1
|
Joyce M, Yang S, Morin K, Duque A, Arellano J, Datta D, Wang M, Arnsten A. β1-adrenoceptor expression on GABAergic interneurons in primate dorsolateral prefrontal cortex: potential role in stress-induced cognitive dysfunction. Neurobiol Stress 2024; 30:100628. [PMID: 38550854 PMCID: PMC10973161 DOI: 10.1016/j.ynstr.2024.100628] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 11/12/2024] Open
Abstract
Uncontrollable stress exposure impairs working memory and reduces the firing of dorsolateral prefrontal cortex (dlPFC) "Delay cells", involving high levels of norepinephrine and dopamine release. Previous work has focused on catecholamine actions on dlPFC pyramidal cells, but inhibitory interneurons may contribute as well. The current study combined immunohistochemistry and multi-scale microscopy with iontophoretic physiology and behavioral analyses to examine the effects of beta1-noradrenergic receptors (β1-ARs) on inhibitory neurons in layer III dlPFC. We found β1-AR robustly expressed on different classes of inhibitory neurons labeled by the calcium-binding proteins calbindin (CB), calretinin (CR), and parvalbumin (PV). Immunoelectron microscopy confirmed β1-AR expression on the plasma membrane of PV-expressing dendrites. PV interneurons can be identified as fast-spiking (FS) in physiological recordings, and thus were studied in macaques performing a working memory task. Iontophoresis of a β1-AR agonist had a mixed effect, increasing the firing of a subset and decreasing the firing of others, likely reflecting loss of firing of the entire microcircuit. This loss of overall firing likely contributes to impaired working memory during stress, as pretreatment with the selective β1-AR antagonist, nebivolol, prevented stress-induced working memory deficits. Thus, selective β1-AR antagonists may be helpful in treating stress-related disorders.
Collapse
Affiliation(s)
- M.K.P. Joyce
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - S. Yang
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - K. Morin
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - A. Duque
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - J. Arellano
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - D. Datta
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - M. Wang
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| | - A.F.T. Arnsten
- Dept. Neuroscience, Yale Medical School, New Haven, CT, 06510, USA
| |
Collapse
|
2
|
Ali Vafaei A, Nazari M, Omoumi S, Rashidy-Pour A, Raise-Abdullahi P. Corticosterone injection into the basolateral amygdala before and after memory reactivation impairs the subsequent expression of fear memory in rats: An interaction of glucocorticoids and β-adrenoceptors. Neurobiol Learn Mem 2023; 205:107829. [PMID: 37734437 DOI: 10.1016/j.nlm.2023.107829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 08/15/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
Glucocorticoid administration, before or after fear memory reactivation, impairs subsequent fear memory expression, but the underlying mechanisms are not well understood. The present study examined the role of basolateral amygdala (BLA) β-adrenoceptors in the effects of intra-BLA corticosterone injection on fear memory in rats. Bilateral cannulae were implanted in the BLA of Wistar male rats. The rats were trained and tested using an inhibitory avoidance task (1 mA footshock for 3 s). Forty-eight hours after training, corticosterone (CORT, 5, 10, or 20 ng/0.5 µl/side) and the β2-adrenoceptor agonist clenbuterol (CLEN, 10 or 20 ng/0.5 µl/side) or the β-adrenoceptor antagonist propranolol (PROP, 250 or 500 ng/0.5 µl/side) were injected into the BLA before or right after memory reactivation (retrieval, Test 1). We performed subsequent tests 2 (Test 2), 5 (Test 3), 7 (Test 4), and 9 (Test 5) days after Test 1. The results demonstrated that CORT injection before Test 1 disrupted memory retrieval and reduced fear expression in Tests 2-5, possibly due to enhanced extinction or impaired reconsolidation. CORT injection after Test 1 also impaired reconsolidation and reduced fear expression in Tests 2-5. CLEN prevented, but PROP exacerbated, the effects of CORT on fear expression. The reminder shock did not recover fear memory in CORT-treated animals, suggesting that reconsolidation, not extinction, was affected. These results indicate that glucocorticoids and β-adrenoceptors in the BLA jointly modulate fear memory reconsolidation and expression. Comprehending the neurobiology of stress and the impact of glucocorticoids on fear memory may lead to new treatments for stress and trauma-induced disorders such as PTSD.
Collapse
Affiliation(s)
- Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Nazari
- Department of Physiology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Samira Omoumi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | | |
Collapse
|
3
|
Ko B, Yoo JY, Yoo T, Choi W, Dogan R, Sung K, Um D, Lee SB, Kim HJ, Lee S, Beak ST, Park SK, Paik SB, Kim TK, Kim JH. Npas4-mediated dopaminergic regulation of safety memory consolidation. Cell Rep 2023; 42:112678. [PMID: 37379214 DOI: 10.1016/j.celrep.2023.112678] [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: 10/05/2022] [Revised: 04/18/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
Amygdala circuitry encodes associations between conditioned stimuli and aversive unconditioned stimuli and also controls fear expression. However, whether and how non-threatening information for unpaired conditioned stimuli (CS-) is discretely processed remains unknown. The fear expression toward CS- is robust immediately after fear conditioning but then becomes negligible after memory consolidation. The synaptic plasticity of the neural pathway from the lateral to the anterior basal amygdala gates the fear expression of CS-, depending upon neuronal PAS domain protein 4 (Npas4)-mediated dopamine receptor D4 (Drd4) synthesis, which is precluded by stress exposure or corticosterone injection. Herein, we show cellular and molecular mechanisms that regulate the non-threatening (safety) memory consolidation, supporting the fear discrimination.
Collapse
Affiliation(s)
- BumJin Ko
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jong-Yeon Yoo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Taesik Yoo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Woochul Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Rumeysa Dogan
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Kibong Sung
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Dahun Um
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hyun Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Sangjun Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seung Tae Beak
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea; Institute of Convergence Science, Yonsei University, Seoul 03722, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea; Institute of Convergence Science, Yonsei University, Seoul 03722, Republic of Korea
| | - Se-Bum Paik
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea; Institute of Convergence Science, Yonsei University, Seoul 03722, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea; Institute of Convergence Science, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
4
|
Nazari M, Rashidy-Pour A, Ali Vafaei A, Raise-Abdullahi P. Systemic corticosterone administration impairs the late fear memory reconsolidation via basolateral amygdala glucocorticoid receptors: dependence on the time window and memory age. Neurobiol Learn Mem 2023:107797. [PMID: 37385522 DOI: 10.1016/j.nlm.2023.107797] [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: 12/07/2022] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Glucocorticoid receptors (GRs) of the basolateral amygdala (BLA) play an important role in memory reconsolidation. The present study investigated the role of the BLA GRs in the late reconsolidation of fear memory using an inhibitory avoidance (IA) task in male Wistar rats. Stainless steel cannulae were implanted bilaterally into the BLA of the rats. After 7 days of recovery, the animals were trained in a one-trial IA task (1mA, 3s). In Experiment one, 48h after the training session, the animals received 3 systemic doses of corticosterone (CORT; 1, 3, or 10 mg/kg, i.p.) followed by an intra-BLA microinjection of the vehicle (0.3µl/side) at different time points (immediately, 12, or 24h) after memory reactivation. Memory reactivation was performed by returning the animals to the light compartment while the sliding door was open. No shock was delivered during memory reactivation. CORT (10 mg/kg) injection 12h after memory reactivation most effectively impaired the late memory reconsolidation (LMR). In the second part of Experiment one, immediately, 12, or 24h after memory reactivation, GR antagonist RU38486 (RU; 1ng/0.3µl/side) was injected into BLA following a systemic injection of CORT (10 mg/kg) to examine whether it would block the CORT effect. RU inhibited the impairing effects of CORT on LMR. In Experiment two, the animals received CORT (10 mg/kg) with time windows immediately, 3, 6, 12, and 24h after memory reactivation. Again, CORT (10 mg/kg) injection 12h after MR impaired LMR. Memory reactivation was performed in the third Experiment, 7, 14, 28, or 56 days after the training session. Injection of CORT (10 mg/kg) 12h later had no significant effect on the LMR. The impairing effect of CORT was seen only in 2-day-old but not 7, 14, 28, and 56-day-old memories. GRs located in BLA seem to play an important role in the LMR of young memory, as with increasing the age of memories, they become less sensitive to manipulation.
Collapse
Affiliation(s)
- Maryam Nazari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | |
Collapse
|
5
|
Yang SS, Mack NR, Shu Y, Gao WJ. Prefrontal GABAergic Interneurons Gate Long-Range Afferents to Regulate Prefrontal Cortex-Associated Complex Behaviors. Front Neural Circuits 2021; 15:716408. [PMID: 34322002 PMCID: PMC8313241 DOI: 10.3389/fncir.2021.716408] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/14/2021] [Indexed: 01/11/2023] Open
Abstract
Prefrontal cortical GABAergic interneurons (INs) and their innervations are essential for the execution of complex behaviors such as working memory, social behavior, and fear expression. These behavior regulations are highly dependent on primary long-range afferents originating from the subcortical structures such as mediodorsal thalamus (MD), ventral hippocampus (vHPC), and basolateral amygdala (BLA). In turn, the regulatory effects of these inputs are mediated by activation of parvalbumin-expressing (PV) and/or somatostatin expressing (SST) INs within the prefrontal cortex (PFC). Here we review how each of these long-range afferents from the MD, vHPC, or BLA recruits a subset of the prefrontal interneuron population to exert precise control of specific PFC-dependent behaviors. Specifically, we first summarize the anatomical connections of different long-range inputs formed on prefrontal GABAergic INs, focusing on PV versus SST cells. Next, we elaborate on the role of prefrontal PV- and SST- INs in regulating MD afferents-mediated cognitive behaviors. We also examine how prefrontal PV- and SST- INs gate vHPC afferents in spatial working memory and fear expression. Finally, we discuss the possibility that prefrontal PV-INs mediate fear conditioning, predominantly driven by the BLA-mPFC pathway. This review will provide a broad view of how multiple long-range inputs converge on prefrontal interneurons to regulate complex behaviors and novel future directions to understand how PFC controls different behaviors.
Collapse
Affiliation(s)
- Sha-Sha Yang
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States,Institute for Translational Brain Research, Fudan University, Shanghai, China
| | - Nancy R. Mack
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Yousheng Shu
- Institute for Translational Brain Research, Fudan University, Shanghai, China
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States,*Correspondence: Wen-Jun Gao,
| |
Collapse
|
6
|
Abstract
Treatment for critical illness typically focuses on a patient's short-term physical recovery; however, recent work has broadened our understanding of the long-term implications of illness and treatment strategies. In particular, survivors of critical illness have significantly elevated risk of developing lasting cognitive impairment and psychiatric disorders. In this review, we examine the role of endogenous and exogenous glucocorticoids in neuropsychiatric outcomes following critical illness. Illness is marked by acute elevation of free cortisol and adrenocorticotropic hormone suppression, which typically normalize after recovery; however, prolonged dysregulation can sometimes occur. High glucocorticoid levels can cause lasting alterations to the plasticity and structural integrity of the hippocampus and prefrontal cortex, and this mechanism may plausibly contribute to impaired memory and cognition in critical illness survivors, though specific evidence is lacking. Glucocorticoids may also exacerbate inflammation-associated neural damage. Conversely, current evidence indicates that glucocorticoids during illness may protect against the development of post-traumatic stress disorder. We propose future directions for research in this field, including determining the role of persistent glucocorticoid elevations after illness in neuropsychiatric outcomes, the role of systemic vs neuroinflammation, and probing unexplored lines of investigation on the role of mineralocorticoid receptors and the gut-brain axis. Progress toward personalized medicine in this area has the potential to produce tangible improvements to the lives patients after a critical illness, including Coronavirus Disease 2019.
Collapse
Affiliation(s)
- Alice R Hill
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Joanna L Spencer-Segal
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Deparment of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
7
|
García‐Pérez D, Milanés MV. Role of glucocorticoids on noradrenergic and dopaminergic neurotransmission within the basolateral amygdala and dentate gyrus during morphine withdrawal place aversion. Addict Biol 2020; 25:e12728. [PMID: 30784175 DOI: 10.1111/adb.12728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/28/2022]
Abstract
Aversive memories related to drug withdrawal can generate a motivational state leading to compulsive drug taking. However, the mechanisms underlying the generation of these withdrawal memories remain unclear. Limbic structures, such as the basolateral amygdala (BLA) and the dentate gyrus (DG) of the hippocampus, play a crucial role in the negative affective component of morphine withdrawal. Given the prominent role of glucocorticoids (GCs), noradrenaline (NA), and dopamine (DA) in memory-related processes, in the present study, we employed the conditioned place aversion (CPA) paradigm to uncover the role of GCs on NA and DA neurotransmission within the BLA and NA neurotransmission within the DG during opiate-withdrawal conditioning (memory formation consolidation), and after reexposure to the conditioned environment (memory retrieval). We observed that adrenalectomy impaired naloxone-induced CPA. Memory retrieval was associated with an increase in dihydroxyphenylacetic acid (DOPAC) levels in the BLA in morphine-addicted animals in a GC-independent manner. Importantly, NA turnover was related with the expression of withdrawal physical signs during the conditioning phase and with locomotor activity during the test phase. On the other hand, reduced DA concentration in the BLA was correlated with the CPA score. Our results indicate that while noradrenergic system is more associated with the somatic consequences of withdrawal, dopaminergic neurotransmission modulates the affective state. Nevertheless, it seems necessary that both systems work together with GCs to enable aversive-memory formation and recall.
Collapse
Affiliation(s)
| | - Maria Victoria Milanés
- Department of PharmacologyUniversity of Murcia Murcia Spain
- Farmacología Celular y MolecularMurcia Institute of Biomedical Research (IMIB) Murcia Spain
| |
Collapse
|
8
|
Colucci P, Mancini GF, Santori A, Zwergel C, Mai A, Trezza V, Roozendaal B, Campolongo P. Amphetamine and the Smart Drug 3,4-Methylenedioxypyrovalerone (MDPV) Induce Generalization of Fear Memory in Rats. Front Mol Neurosci 2019; 12:292. [PMID: 31849606 PMCID: PMC6895769 DOI: 10.3389/fnmol.2019.00292] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/14/2019] [Indexed: 11/13/2022] Open
Abstract
Human studies have consistently shown that drugs of abuse affect memory function. The psychostimulants amphetamine and the "bath salt" 3,4-methylenedioxypyrovalerone (MDPV) increase brain monoamine levels through a similar, yet not identical, mechanism of action. Findings indicate that amphetamine enhances the consolidation of memory for emotional experiences, but still MDPV effects on memory function are underinvestigated. Here, we tested the effects induced by these two drugs on generalization of fear memory and their relative neurobiological underpinnings. To this aim, we used a modified version of the classical inhibitory avoidance task, termed inhibitory avoidance discrimination task. According to such procedure, adult male Sprague-Dawley rats were first exposed to one inhibitory avoidance apparatus and, with a 1-min delay, to a second apparatus where they received an inescapable footshock. Forty-eight hours later, retention latencies were tested, in a randomized order, in the two training apparatuses as well as in a novel contextually modified apparatus to assess both strength and generalization of memory. Our results indicated that both amphetamine and MDPV induced generalization of fear memory, whereas only amphetamine enhanced memory strength. Co-administration of the β-adrenoceptor antagonist propranolol prevented the effects of both amphetamine and MDPV on the strength and generalization of memory. The dopaminergic receptor blocker cis-flupenthixol selectively reversed the amphetamine effect on memory generalization. These findings indicate that amphetamine and MDPV induce generalization of fear memory through different modulations of noradrenergic and dopaminergic neurotransmission.
Collapse
Affiliation(s)
- Paola Colucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Giulia Federica Mancini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Alessia Santori
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry & Technologies, Sapienza University of Rome, Rome, Italy.,Department of Medicine of Precision, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Antonello Mai
- Department of Drug Chemistry & Technologies, Sapienza University of Rome, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University Roma Tre, Rome, Italy
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands.,Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
9
|
Nitschke JP, Chu S, Pruessner JC, Bartz JA, Sheldon S. Post-learning stress reduces the misinformation effect: effects of psychosocial stress on memory updating. Psychoneuroendocrinology 2019; 102:164-171. [PMID: 30562688 DOI: 10.1016/j.psyneuen.2018.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022]
Abstract
Episodic memories can be modified when exposed to new and related information. This phenomenon, known as memory updating, is generally thought to be adaptive but can also lead to incorporating false information into a memory trace. Given the well-known effects of stress on episodic memory, we used a false information paradigm to investigate if acute stress during memory updating (i.e., post-learning stage) affected false memory formation. In a between-subject design, young healthy participants completed the initial phases of the misinformation experiment - they studied an event via a slideshow and then were exposed a related narrative that contained misleading information about that event. After, half of the participants were exposed to acute psychosocial stress and the other half completed a control task. Once stress levels returned to baseline, all of the participants completed the final phase of the experiment, which was a memory test for slideshow that included items containing true facts and misinformation. Participants in the stress condition showed a reduced misinformation effect and were better able to discriminate true from false information compared to control participants. This pattern of results held even when participants were tested on the same memory test after a multiple day delay, illustrating the long-lasting effects of stress on false memory formation specifically, and memory updating generally. We discuss how our results add to the understanding of the time-dependent factors that moderate stress effects on memory, and speculate how stress effects on memory updating can be positive, by limiting intrusions into encoded events, but also negative, by limiting the ability to integrate information with other concepts, harming memory generalization.
Collapse
Affiliation(s)
- Jonas P Nitschke
- Department of Psychology, McGill University, Avenue McGill College 2001, H3A 1G1 Montreal, Canada.
| | - Sonja Chu
- Department of Psychology, McGill University, Avenue McGill College 2001, H3A 1G1 Montreal, Canada
| | - Jens C Pruessner
- Department of Psychology, University of Konstanz, 78457 Konstanz, Germany
| | - Jennifer A Bartz
- Department of Psychology, McGill University, Avenue McGill College 2001, H3A 1G1 Montreal, Canada
| | - Signy Sheldon
- Department of Psychology, McGill University, Avenue McGill College 2001, H3A 1G1 Montreal, Canada
| |
Collapse
|
10
|
Peters JR, Eisenlohr-Moul TA, Walsh EC, Derefinko KJ. Exploring the pathophysiology of emotion-based impulsivity: The roles of the sympathetic nervous system and hostile reactivity. Psychiatry Res 2018; 267:368-375. [PMID: 29957555 PMCID: PMC6309543 DOI: 10.1016/j.psychres.2018.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 11/22/2022]
Abstract
The tendency to engage in impulsive behavior in the context of negative affect, known as negative urgency, has emerged as a powerful transdiagnostic predictor of behavioral dysregulation. Although general vulnerability to negative affect (neuroticism) correlates with negative urgency, not all neurotic individuals engage in urgent behavior. Given prior experimental evidence that sympathetic nervous system (SNS) activation may promote emotion-related impulsivity, the present study examines tonic SNS activity as a moderator of the link between neuroticism and negative urgency. Participants (N = 194) completed measures of neuroticism and negative urgency, as well as a stress task. They also underwent assessment of tonic SNS activity (cardiac pre-ejection period). The link between neuroticism and negative urgency was strengthened for individuals with higher tonic SNS activity; however, this was not the case for behavioral performance on the task. A similar pattern was demonstrated for hostile reactivity to the stress task; increased hostile response partially explained the interaction between SNS activation and neuroticism on negative urgency. These findings suggest a potential facilitative role of the SNS in hostile reactivity and emotion-driven impulsivity among more neurotic individuals.
Collapse
Affiliation(s)
- Jessica R Peters
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA.
| | | | - Erin C Walsh
- Department of Physical Medicine and Rehabilitation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Karen J Derefinko
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
11
|
Zhu R, Liu X, Shi Y, Wang X, Xue L, Zhao H. Propranolol can induce PTSD-like memory impairments in rats. Brain Behav 2018; 8:e00905. [PMID: 29484264 PMCID: PMC5822589 DOI: 10.1002/brb3.905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction One hallmark symptom of post-traumatic stress disorder (PTSD) is an inability to restrict fear responses to the appropriate predictor. An infusion of glucocorticoids (GCs) after a high-intensity shock has been shown to induce PTSD-like memory impairments. In addition to GCs, noradrenergic signalling is also recognized as a key biomarker underlying PTSD symptomatology. Methods To explore the role of the noradrenergic system in PTSD-like memory impairments, in this study, various doses of the β-adrenoceptor antagonist propranolol were systemically or bilaterally injected into the dorsal hippocampus immediately after unpaired cue-shock contextual fear conditioning, and then the rats were tested 24 h later. Results Interestingly, we found that only low-dose propranolol could induce PTSD-like memory impairments, as rats showed reduced freezing to the correct predictor and generalized fear responses to the safe cues, accompanied by increased NE levels in the hippocampus and altered neural activity within the frontal-subcortical circuit. Conclusion These findings demonstrate that the noradrenergic system is involved in regulating the consolidation of contextual fear memory and that propranolol can dose-dependently induce PTSD-like memory impairments.
Collapse
Affiliation(s)
- Rong‐Ting Zhu
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Xiang‐Hui Liu
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Yan‐Wei Shi
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Xiao‐Guang Wang
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Li Xue
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Hu Zhao
- Faculty of Forensic MedicineZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
12
|
Villain H, Benkahoul A, Birmes P, Ferry B, Roullet P. Influence of early stress on memory reconsolidation: Implications for post-traumatic stress disorder treatment. PLoS One 2018; 13:e0191563. [PMID: 29352277 PMCID: PMC5774817 DOI: 10.1371/journal.pone.0191563] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 01/08/2018] [Indexed: 11/19/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a common consequence of exposure to a life-threatening event. Currently, pharmacological treatments are limited by high rates of relapse, and novel treatment approaches are needed. We have recently demonstrated that propranolol, a β-adrenergic antagonist, inhibited aversive memory reconsolidation in animals. Following this, in an open-label study 70% of patients with PTSD treated with propranolol during reactivation of traumatic memory exhibited full remission. However, the reason why 30% of these patients did not respond positively to propranolol treatment is still unclear. One of the major candidates as factor of treatment resistance is the patient's early-life traumatic history. To test the role of this factor, mice with pre- or postnatal stress are being tested in fear conditioning and in a new behavioral task, the "city-like", specifically designed as a mouse model of PTSD. After reactivation of the traumatic event, mice received propranolol injection to block the noradrenergic system during memory reconsolidation. Results show that, in the “city-like” test, control mice strongly avoided the shock compartment but also the compartments containing cues associated with the electric shocks. Injection of propranolol after reactivation greatly reduced the memory of the traumatic event, but this effect was not present when mice had received pre- or postnatal stress. Moreover, propranolol produced only a very weak effect in the fear conditioning test, and never changed the corticosterone level whatever the behavioral experiment. Taken together our results suggest that our new behavioural paradigm is well adapted to PTSD study in mice, and that early stress exposure may have an impact on propranolol PTSD treatment outcome. These data are critical to understanding the effect of propranolol treatment, in order to improve the therapeutic protocol currently used in humans.
Collapse
Affiliation(s)
- Hélène Villain
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Aïcha Benkahoul
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Philippe Birmes
- Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS,Toulouse, France
| | - Barbara Ferry
- Centre of Research in Neuroscience Lyon—UMR CNRS 5292—INSERM U 1028—Université Claude Bernard Lyon 1,Lyon, France
| | - Pascal Roullet
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
- * E-mail:
| |
Collapse
|
13
|
Urueña-Méndez GP, Lamprea M. Efectos de la inactivación sistémica de los receptores GR y MR sobre el daño rápido en la recuperación de la memoria espacial inducido por corticosterona. UNIVERSITAS PSYCHOLOGICA 2017. [DOI: 10.11144/javeriana.upsy15-5.eisr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Estudios previos de nuestro laboratorio han mostrado que la inyección de corticosterona aplicada diez minutos antes de la prueba de recobre afecta la recuperación de la memoria espacial en el laberinto de Barnes. Se ha propuesto que los efectos de esta hormona están mediados por la activación de los receptores clásicos de glucocorticoides; sin embargo la rápida aparición de los efectos permite suponer un mecanismo alternativo de tipo no genómico. El objetivo del presente estudio fue determinar la participación de los receptores de glucocorticoides GR y MR sobre el efecto rápido de la corticosterona en la recuperación de la memoria espacial en el laberinto de Barnes. Treinta y siete ratas Wistar macho fueron entrenadas en la tarea y 24h después recibieron una inyección subcutánea de antagonista GR, antagonista MR o vehículo. Cincuenta minutos después de la inyección, los animales fueron inyectados con corticosterona o vehículo y diez minutos después se evaluó la recuperación de la memoria espacial. Los resultados mostraron que la corticosterona perjudica rápidamente la recuperación de la memoria espacial a largo plazo, pues los animales inyectados presentaron mayores latencias de escape, mayor número de errores, mayor número de exploraciones y mayor distancia recorrida hasta alcanzar la meta. Este efecto sólo fue revertido con la administración del antagonista MR. Este hallazgo concuerda con estudios in vitro donde se muestra que los efectos rápidos de la corticosterona sobre la trasmisión glutamatérgica en el hipocampo están mediados por los receptores MR, posiblemente localizados en la membrana celular.
Collapse
|
14
|
Atsak P, Guenzel FM, Kantar-Gok D, Zalachoras I, Yargicoglu P, Meijer OC, Quirarte GL, Wolf OT, Schwabe L, Roozendaal B. Glucocorticoids mediate stress-induced impairment of retrieval of stimulus-response memory. Psychoneuroendocrinology 2016; 67:207-15. [PMID: 26923851 DOI: 10.1016/j.psyneuen.2016.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/09/2016] [Accepted: 02/09/2016] [Indexed: 11/26/2022]
Abstract
Acute stress and elevated glucocorticoid hormone levels are well known to impair the retrieval of hippocampus-dependent 'declarative' memory. Recent findings suggest that stress might also impair the retrieval of non-hippocampal memories. In particular, stress shortly before retention testing was shown to impair the retrieval of striatal stimulus-response associations in humans. However, the mechanism underlying this stress-induced retrieval impairment of non-hippocampal stimulus-response memory remains elusive. In the present study, we investigated whether an acute elevation in glucocorticoid levels mediates the impairing effects of stress on retrieval of stimulus-response memory. Male Sprague-Dawley rats were trained on a stimulus-response task in an eight-arm radial maze until they learned to associate a stimulus, i.e., cue, with a food reward in one of the arms. Twenty-four hours after successful acquisition, they received a systemic injection of vehicle, corticosterone (1mg/kg), the corticosterone-synthesis inhibitor metyrapone (35mg/kg) or were left untreated 1h before retention testing. We found that the corticosterone injection impaired the retrieval of stimulus-response memory. We further found that the systemic injection procedure per se was stressful as the vehicle administration also increased plasma corticosterone levels and impaired the retrieval of stimulus-response memory. However, memory retrieval was not impaired when rats were tested 2min after the systemic vehicle injection, before any stress-induced elevation in corticosterone levels had occurred. Moreover, metyrapone treatment blocked the effect of injection stress on both plasma corticosterone levels and memory retrieval impairment, indicating that the endogenous corticosterone response mediates the stress-induced memory retrieval impairment. None of the treatments affected rats' locomotor activity or motivation to search for the food reward within the maze. These findings show that stress may affect memory processes beyond the hippocampus and that these stress effects are due to the action of glucocorticoids.
Collapse
Affiliation(s)
- Piray Atsak
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, 6525 EZ Nijmegen, The Netherlands.
| | - Friederike M Guenzel
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Institute of Cognitive Neuroscience, Department of Cognitive Psychology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Deniz Kantar-Gok
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, 07070 Antalya, Turkey
| | - Ioannis Zalachoras
- Department of Endocrinology and Metabolism, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RA Leiden, The Netherlands
| | - Piraye Yargicoglu
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, 07070 Antalya, Turkey
| | - Onno C Meijer
- Department of Endocrinology and Metabolism, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RA Leiden, The Netherlands
| | - Gina L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro 76230, Mexico
| | - Oliver T Wolf
- Institute of Cognitive Neuroscience, Department of Cognitive Psychology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Lars Schwabe
- Department of Cognitive Psychology, University of Hamburg, 20146 Hamburg, Germany
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, 6525 EZ Nijmegen, The Netherlands
| |
Collapse
|
15
|
Abstract
Fear memory is the best-studied form of memory. It was thoroughly investigated in the past 60 years mostly using two classical conditioning procedures (contextual fear conditioning and fear conditioning to a tone) and one instrumental procedure (one-trial inhibitory avoidance). Fear memory is formed in the hippocampus (contextual conditioning and inhibitory avoidance), in the basolateral amygdala (inhibitory avoidance), and in the lateral amygdala (conditioning to a tone). The circuitry involves, in addition, the pre- and infralimbic ventromedial prefrontal cortex, the central amygdala subnuclei, and the dentate gyrus. Fear learning models, notably inhibitory avoidance, have also been very useful for the analysis of the biochemical mechanisms of memory consolidation as a whole. These studies have capitalized on in vitro observations on long-term potentiation and other kinds of plasticity. The effect of a very large number of drugs on fear learning has been intensively studied, often as a prelude to the investigation of effects on anxiety. The extinction of fear learning involves to an extent a reversal of the flow of information in the mentioned structures and is used in the therapy of posttraumatic stress disorder and fear memories in general.
Collapse
Affiliation(s)
- Ivan Izquierdo
- National Institute of Translational Neuroscience, National Research Council of Brazil, and Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristiane R. G. Furini
- National Institute of Translational Neuroscience, National Research Council of Brazil, and Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jociane C. Myskiw
- National Institute of Translational Neuroscience, National Research Council of Brazil, and Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
16
|
Thomas SA. Neuromodulatory signaling in hippocampus-dependent memory retrieval. Hippocampus 2015; 25:415-31. [PMID: 25475876 DOI: 10.1002/hipo.22394] [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] [Accepted: 11/24/2014] [Indexed: 12/15/2022]
Abstract
Considerable advances have been made toward understanding the molecular signaling events that underlie memory acquisition and consolidation. In contrast, less is known about memory retrieval, despite its necessity for utilizing learned information. This review focuses on neuromodulatory and intracellular signaling events that underlie memory retrieval mediated by the hippocampus, for which the most information is currently available. Among neuromodulators, adrenergic signaling is required for the retrieval of various types of hippocampus-dependent memory. Although they contribute to acquisition and/or consolidation, cholinergic and dopaminergic signaling are generally not required for retrieval. Interestingly, while not required for retrieval, serotonergic and opioid signaling may actually constrain memory retrieval. Roles for histamine and non-opioid neuropeptides are currently unclear but possible. A critical effector of adrenergic signaling in retrieval is reduction of the slow afterhyperpolarization mediated by β1 receptors, cyclic AMP, protein kinase A, Epac, and possibly ERK. In contrast, stress and glucocorticoids impair retrieval by decreasing cyclic AMP, mediated in part by the activation of β2 -adrenergic receptors. Clinically, alterations in neuromodulatory signaling and in memory retrieval occur in Alzheimer's disease, Down syndrome, depression, and post-traumatic stress disorder, and recent evidence has begun to link changes in neuromodulatory signaling with effects on memory retrieval.
Collapse
Affiliation(s)
- Steven A Thomas
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Glucocorticoid enhancement of dorsolateral striatum-dependent habit memory requires concurrent noradrenergic activity. Neuroscience 2015; 311:1-8. [PMID: 26470808 DOI: 10.1016/j.neuroscience.2015.10.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/02/2015] [Accepted: 10/08/2015] [Indexed: 11/20/2022]
Abstract
Previous findings indicate that post-training administration of glucocorticoid stress hormones can interact with the noradrenergic system to enhance consolidation of hippocampus- or amygdala-dependent cognitive/emotional memory. The present experiments were designed to extend these findings by examining the potential interaction of glucocorticoid and noradrenergic mechanisms in enhancement of dorsolateral striatum (DLS)-dependent habit memory. In experiment 1, different groups of adult male Long-Evans rats received training in two DLS-dependent memory tasks. In a cued water maze task, rats were released from various start points and were reinforced to approach a visibly cued escape platform. In a response-learning version of the water plus-maze task, animals were released from opposite starting positions and were reinforced to make a consistent egocentric body-turn to reach a hidden escape platform. Immediately post-training, rats received peripheral injections of the glucocorticoid corticosterone (1 or 3 mg/kg) or vehicle solution. In both tasks, corticosterone (3 mg/kg) enhanced DLS-dependent habit memory. In experiment 2, a separate group of animals received training in the response learning version of the water plus-maze task and were given peripheral post-training injections of corticosterone (3 mg/kg), the β-adrenoreceptor antagonist propranolol (3 mg/kg), corticosterone and propranolol concurrently, or control vehicle solution. Corticosterone injections again enhanced DLS-dependent memory, and this effect was blocked by concurrent administration of propranolol. Propranolol administration by itself (3 mg/kg) did not influence DLS-dependent memory. Taken together, the findings indicate an interaction between glucocorticoid and noradrenergic mechanisms in DLS-dependent habit memory. Propranolol administration may be useful in treating stress-related human psychopathologies associated with a dysfunctional DLS-dependent habit memory system.
Collapse
|
18
|
Morena M, De Castro V, Gray JM, Palmery M, Trezza V, Roozendaal B, Hill MN, Campolongo P. Training-Associated Emotional Arousal Shapes Endocannabinoid Modulation of Spatial Memory Retrieval in Rats. J Neurosci 2015; 35:13962-74. [PMID: 26468197 PMCID: PMC6608184 DOI: 10.1523/jneurosci.1983-15.2015] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/05/2015] [Accepted: 08/28/2015] [Indexed: 12/31/2022] Open
Abstract
Variations in environmental aversiveness influence emotional memory processes in rats. We have previously shown that cannabinoid effects on memory are dependent on the stress level at the time of training as well as on the aversiveness of the environmental context. Here, we investigated whether the hippocampal endocannabinoid system modulates memory retrieval depending on the training-associated arousal level. Male adult Sprague Dawley rats were trained on a water maze spatial task at two different water temperatures (19°C and 25°C) to elicit either higher or lower stress levels, respectively. Rats trained under the higher stress condition had better memory and higher corticosterone concentrations than rats trained at the lower stress condition. The cannabinoid receptor agonist WIN55212-2 (10-30 ng/side), the 2-arachidonoyl glycerol (2-AG) hydrolysis inhibitor JZL184 (0.1-1 μg/side), and the anandamide (AEA) hydrolysis inhibitor URB597 (10-30 ng/side) were administered bilaterally into the hippocampus 60 min before probe-trial retention testing. WIN55212-2 or JZL184, but not URB597, impaired probe-trial performances only of rats trained at the higher stressful condition. Furthermore, rats trained under higher stress levels displayed an increase in hippocampal 2-AG, but not AEA, levels at the time of retention testing and a decreased affinity of the main 2-AG-degrading enzyme for its substrate. The present findings indicate that the endocannabinoid 2-AG in the hippocampus plays a key role in the selective regulation of spatial memory retrieval of stressful experience, shedding light on the neurobiological mechanisms involved in the impact of stress effects on memory processing. SIGNIFICANCE STATEMENT Endogenous cannabinoids play a central role in the modulation of memory for emotional events. Here we demonstrate that the endocannabinoid 2-arachidonoylglycerol in the hippocampus, a brain region crucially involved in the regulation of memory processes, selectively modulates spatial memory recall of stressful experiences. Thus, our findings provide evidence that the endocannabinoid 2-arachidonoylglycerol is a key player in mediating the impact of stress on memory retrieval. These findings can pave the way to new potential therapeutic intervention for the treatment of neuropsychiatric disorders, such as post-traumatic stress disorder, where a previous exposure to traumatic events could alter the response to traumatic memory recall leading to mental illness.
Collapse
Affiliation(s)
- Maria Morena
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy, Hotchkiss Brain Institute and Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, Alberta T2N 4N1, Canada,
| | - Valentina De Castro
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - J Megan Gray
- Hotchkiss Brain Institute and Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Maura Palmery
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University Roma Tre, 00146 Rome, Italy, and
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center and Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6525 EZ Nijmegen, The Netherlands
| | - Matthew N Hill
- Hotchkiss Brain Institute and Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy,
| |
Collapse
|
19
|
Pedraza LK, Sierra RO, Boos FZ, Haubrich J, Quillfeldt JA, de Oliveira Alvares L. The dynamic nature of systems consolidation: Stress during learning as a switch guiding the rate of the hippocampal dependency and memory quality. Hippocampus 2015; 26:362-71. [DOI: 10.1002/hipo.22527] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Lizeth K. Pedraza
- Laboratório De Neurobiologia Da Memória; Porto Alegre 91.501-970 Brazil
- Graduate Program in Neuroscience, Institute of Health Sciences; Federal University of Rio Grande Do Sul; Porto Alegre 90.046-900 Brazil
| | - Rodrigo O. Sierra
- Graduate Program in Neuroscience, Institute of Health Sciences; Federal University of Rio Grande Do Sul; Porto Alegre 90.046-900 Brazil
- Laboratório De Psicobiologia E Neurocomputação, Biophysics Department; Biosciences Institute; Porto Alegre 91.501-970 Brazil
| | - Flávia Z. Boos
- Graduate Program in Neuroscience, Institute of Health Sciences; Federal University of Rio Grande Do Sul; Porto Alegre 90.046-900 Brazil
- Laboratório De Psicobiologia E Neurocomputação, Biophysics Department; Biosciences Institute; Porto Alegre 91.501-970 Brazil
| | - Josué Haubrich
- Graduate Program in Neuroscience, Institute of Health Sciences; Federal University of Rio Grande Do Sul; Porto Alegre 90.046-900 Brazil
- Laboratório De Psicobiologia E Neurocomputação, Biophysics Department; Biosciences Institute; Porto Alegre 91.501-970 Brazil
| | - Jorge A. Quillfeldt
- Graduate Program in Neuroscience, Institute of Health Sciences; Federal University of Rio Grande Do Sul; Porto Alegre 90.046-900 Brazil
- Laboratório De Psicobiologia E Neurocomputação, Biophysics Department; Biosciences Institute; Porto Alegre 91.501-970 Brazil
| | - Lucas de Oliveira Alvares
- Laboratório De Neurobiologia Da Memória; Porto Alegre 91.501-970 Brazil
- Graduate Program in Neuroscience, Institute of Health Sciences; Federal University of Rio Grande Do Sul; Porto Alegre 90.046-900 Brazil
| |
Collapse
|
20
|
Maki PM, Mordecai KL, Rubin LH, Sundermann E, Savarese A, Eatough E, Drogos L. Menstrual cycle effects on cortisol responsivity and emotional retrieval following a psychosocial stressor. Horm Behav 2015; 74:201-8. [PMID: 26187711 PMCID: PMC4876953 DOI: 10.1016/j.yhbeh.2015.06.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/27/2015] [Accepted: 06/23/2015] [Indexed: 01/31/2023]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Laboratory-induced stress produces elevations in cortisol and deficits in memory, especially when stress is induced immediately before retrieval of emotionally valent stimuli. Sex and sex steroids appear to influence these stress-induced outcomes, though no study has directly compared the effects of laboratory-induced stress on cortisol and emotional retrieval across the menstrual cycle. We examined the effect of psychosocial stress on cortisol responsivity and emotional retrieval in women tested during either the follicular phase (low estradiol and progesterone) or the luteal phase (higher estradiol and progesterone). Forty women (50% White; age 18-40 years) participated in the study; 20 completed the task during the luteal phase and 20 during the follicular phase. Psychosocial stress was induced with the Trier Social Stress Test (TSST). On the day before the TSST, participants learned two lists of word pairs to 100% criterion. The next day, participants recalled one list after the control condition and the other after the TSST. Women in the follicular phase, but not the luteal phase, demonstrated a significant cortisol response to the TSST. There was a stress-induced decrease in emotional retrieval following the TSST, but this effect was not modified by menstrual phase. However, regression and correlational analyses showed that individual differences in stress-induced cortisol levels were associated with impaired emotional retrieval in the follicular phase only. The present findings indicate that cortisol responsivity and the impairing effects of cortisol on emotional memory are lower when levels of estradiol and progesterone are high compared to when levels are low.
Collapse
Affiliation(s)
- Pauline M Maki
- University of Illinois at Chicago, College of Medicine, Department of Psychiatry, 912 South Wood Street, Chicago, IL 60612, USA; University of Illinois at Chicago, College of Liberal Arts and Sciences, Department of Psychology, 1007 West Harrison Street (M/C 285), Chicago, IL 60607-7137, USA; Rosalind Franklin University of Medicine and Science, College of Health Professions, Clinical Psychology, 3333 Green Bay Road, North Chicago, IL 60064-3095, USA.
| | - Kristen L Mordecai
- University of Illinois at Chicago, College of Medicine, Department of Psychiatry, 912 South Wood Street, Chicago, IL 60612, USA.
| | - Leah H Rubin
- University of Illinois at Chicago, College of Medicine, Department of Psychiatry, 912 South Wood Street, Chicago, IL 60612, USA; University of Illinois at Chicago, College of Liberal Arts and Sciences, Department of Psychology, 1007 West Harrison Street (M/C 285), Chicago, IL 60607-7137, USA.
| | - Erin Sundermann
- University of Illinois at Chicago, College of Liberal Arts and Sciences, Department of Psychology, 1007 West Harrison Street (M/C 285), Chicago, IL 60607-7137, USA.
| | - Antonia Savarese
- University of Illinois at Chicago, College of Medicine, Department of Psychiatry, 912 South Wood Street, Chicago, IL 60612, USA; University of Illinois at Chicago, Graduate Program in Neuroscience, 828S. Wolcott Ave., Chicago, IL 60612, USA.
| | - Erin Eatough
- University of Illinois at Chicago, College of Medicine, Department of Psychiatry, 912 South Wood Street, Chicago, IL 60612, USA.
| | - Lauren Drogos
- University of Illinois at Chicago, College of Liberal Arts and Sciences, Department of Psychology, 1007 West Harrison Street (M/C 285), Chicago, IL 60607-7137, USA.
| |
Collapse
|
21
|
Careaga MBL, Tiba PA, Ota SM, Suchecki D. Pre-test metyrapone impairs memory recall in fear conditioning tasks: lack of interaction with β-adrenergic activity. Front Behav Neurosci 2015; 9:51. [PMID: 25784866 PMCID: PMC4347504 DOI: 10.3389/fnbeh.2015.00051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 02/11/2015] [Indexed: 12/21/2022] Open
Abstract
Cognitive processes, such as learning and memory, are essential for our adaptation to environmental changes and consequently for survival. Numerous studies indicate that hormones secreted during stressful situations, such as glucocorticoids (GCs), adrenaline and noradrenaline, regulate memory functions, modulating aversive memory consolidation and retrieval, in an interactive and complementary way. Thus, the facilitatory effects of GCs on memory consolidation as well as their suppressive effects on retrieval are substantially explained by this interaction. On the other hand, low levels of GCs are also associated with negative effects on memory consolidation and retrieval and the mechanisms involved are not well understood. The present study sought to investigate the consequences of blocking the rise of GCs on fear memory retrieval in multiple tests, assessing the participation of β-adrenergic signaling on this effect. Metyrapone (GCs synthesis inhibitor; 75 mg/kg), administered 90 min before the first test of contextual or tone fear conditioning (TFC), negatively affected animals’ performances, but this effect did not persist on a subsequent test, when the conditioned response was again expressed. This result suggested that the treatment impaired fear memory retrieval during the first evaluation. The administration immediately after the first test did not affect the animals’ performances in contextual fear conditioning (CFC), suggesting that the drug did not interfere with processes triggered by memory reactivation. Moreover, metyrapone effects were independent of β-adrenergic signaling, since concurrent administration with propranolol (2 mg/kg), a β-adrenergic antagonist, did not modify the effects induced by metyrapone alone. These results demonstrate that pre-test metyrapone administration led to negative effects on fear memory retrieval and this action was independent of a β-adrenergic signaling.
Collapse
Affiliation(s)
- Mariella B L Careaga
- Departamento de Psicobiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Paula A Tiba
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC São Paulo, Brazil
| | - Simone M Ota
- Departamento de Psicobiologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia, Universidade Federal de São Paulo São Paulo, Brazil
| |
Collapse
|
22
|
Mohammadkhani R, Darbandi N, Vafaei AA, Ahmadalipour A, Rashidy-Pour A. Glucocorticoid-induced impairment of long-term memory retrieval in female rats: influences of estrous cycle and estrogen. Neurobiol Learn Mem 2015; 118:209-15. [PMID: 25576134 DOI: 10.1016/j.nlm.2014.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/19/2014] [Accepted: 12/27/2014] [Indexed: 10/24/2022]
Abstract
Using an inhibitory avoidance (IA) task, the effects of glucocorticoids on memory retrieval in intact and ovariectomized (OVX) female rats were investigated. Young adult female rats were trained in a one trial IA task (1-mA, 3-s footshock). The latency to reenter the dark compartment of the apparatus was recorded in the retention test performed 48h after training. Pre-retrieval injection of corticosterone (CORT, 1, 3, and 10mg/kg) to OVX rats impaired memory retrieval at all doses tested. Similar administration of CORT (3mg/kg) in intact female rats impaired memory retrieval in the estrus phase (when endogenous plasma levels of estrogen are low) but not in the proestrus phase (when endogenous levels of estrogen are high). Concurrent administration of CORT (3mg/kg) and 17-β-estradiol (15μg/kg) in both proestrus and estrous phases impaired memory retrieval. Our findings indicate that the effects of corticosterone on memory retrieval are modulated by the estrous cycle and 17-β-estradiol.
Collapse
Affiliation(s)
- Raziyeh Mohammadkhani
- Department of Biology, Faculty of Science, Arak University, Arak 38156-8-8349, Iran; Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Niloufar Darbandi
- Department of Biology, Faculty of Science, Arak University, Arak 38156-8-8349, Iran
| | - Abbas Ali Vafaei
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Ahmadalipour
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Student's Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
23
|
Affiliation(s)
- James L. McGaugh
- Center for the Neurobiology of Learning and Memory and Department of Neurobiology and Behavior, University of California, Irvine, California 92697-3800;
| |
Collapse
|
24
|
Jia M, Smerin SE, Zhang L, Xing G, Li X, Benedek D, Ursano R, Li H. Corticosterone mitigates the stress response in an animal model of PTSD. J Psychiatr Res 2015; 60:29-39. [PMID: 25307716 DOI: 10.1016/j.jpsychires.2014.09.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/16/2014] [Accepted: 09/18/2014] [Indexed: 12/18/2022]
Abstract
Activation of glucocorticoid receptor signaling in the stress response to traumatic events has been implicated in the pathogenesis of stress-associated psychiatric disorders such as post-traumatic stress disorder (PTSD). Elevated startle response and hyperarousal are hallmarks of PTSD, and are generally considered to evince fear (DSM V). To further examine the efficacy of corticosterone in treating hyperarousal and elevated fear, the present study utilized a learned helplessness stress model in which rats are restrained and subjected to tail shock for three days. These stressed rats develop a delayed long-lasting exaggeration of the acoustic startle response (ASR) and retarded body weight growth, similar to symptoms of PTSD patients (Myers et al., 2005; Speed et al., 1989). We demonstrate that both pre-stress and post-stress administration of corticosterone (3 mg/kg/day) mitigates a subsequent exaggeration of the ASR measured 14 days after cessation of the stress protocol. Furthermore, the mitigating efficacy of pre-stress administration of corticosterone (3 mg/kg/day for three days) appeared to last significantly longer, up to 21 days after the cessation of the stress protocol, in comparison to that of post-stress administration of corticosterone. However, pre-stress administration of corticosterone at 0.3 mg/kg/day for three days did not mitigate stress-induced exaggeration of the ASR measured at both 14 and 21 days after the cessation of the stress protocol. In addition, pre-stress administration of corticosterone (3 mg/kg/day for three days) mitigates the retardation of body weight growth otherwise resulting from the stress protocol. Congruently, co-administration of the corticosterone antagonist RU486 (40 mg/kg/day for three days) with corticosterone (3 mg/kg/day) prior to stress diminished the mitigating efficacy of the exogenous corticosterone on exaggerated ASR and stress-retarded body weight. The relative efficacy of pre versus post administration of corticosterone and high versus low dose of corticosterone on stress-induced exaggeration of innate fear response and stress-retarded body weight growth indicate that exogenous corticosterone administration within an appropriate time window and dosage are efficacious in diminishing traumatic stress induced pathophysiological processes. Clinical implications associated with the efficacy of prophylactic and therapeutic corticosterone therapy for mitigating symptoms of PTSD are discussed, particularly in relation to diminishing hyperarousal and exaggerated innate fear response.
Collapse
Affiliation(s)
- Min Jia
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Stanley E Smerin
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Lei Zhang
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Guoqiang Xing
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Xiaoxia Li
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - David Benedek
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Robert Ursano
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - He Li
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Service University of Health Sciences (USUHS), 4301 Jones Bridge Rd., Bethesda, MD 20814, USA.
| |
Collapse
|
25
|
Yehuda R, Bierer LM, Pratchett LC, Lehrner A, Koch EC, Van Manen JA, Flory JD, Makotkine I, Hildebrandt T. Cortisol augmentation of a psychological treatment for warfighters with posttraumatic stress disorder: Randomized trial showing improved treatment retention and outcome. Psychoneuroendocrinology 2015; 51:589-97. [PMID: 25212409 DOI: 10.1016/j.psyneuen.2014.08.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 08/03/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Prolonged exposure (PE) therapy for post-traumatic stress disorder (PTSD) in military veterans has established efficacy, but is ineffective for a substantial number of patients. PE is also associated with high dropout rates. We hypothesized that hydrocortisone augmentation would enhance symptom improvement and reduce drop-out rates by diminishing the distressing effects of traumatic memories retrieved during imaginal exposure. We also hypothesized that in responders, hydrocortisone augmentation would be more effective in reversing glucocorticoid indices associated with PTSD than placebo augmentation. METHOD Twenty-four veterans were randomized to receive either 30 mg oral hydrocortisone or placebo prior to PE sessions 3-10 in a double-blind protocol. Glucocorticoid receptor sensitivity was assessed in cultured peripheral blood mononuclear cells (PBMC) using the in vitro lysozyme inhibition test and was determined before and after treatment. Intent-to-treat analysis was performed using latent growth curve modeling of treatment effects on change in PTSD severity over time. Veterans who no longer met diagnostic criteria for PTSD at post-treatment were designated as responders. RESULTS Veterans randomized to hydrocortisone or placebo augmentation did not differ significantly in clinical severity or glucocorticoid sensitivity at pre-treatment. Hydrocortisone augmentation was associated with greater reduction in total PTSD symptoms compared to placebo, a finding that was explained by significantly greater patient retention in the hydrocortisone augmentation condition. A significant treatment condition by responder status interaction for glucocorticoid sensitivity indicated that responders to hydrocortisone augmentation had the highest pre-treatment glucocorticoid sensitivity (lowest lysozyme IC50-DEX) that diminished over the course of treatment. There was a significant association between decline in glucocorticoid responsiveness and improvement in PTSD symptoms among hydrocortisone recipients. CONCLUSIONS The results of this pilot study suggest that hydrocortisone augmentation of PE may result in greater retention in treatment and thereby promote PTSD symptom improvement. Further, the results suggest that particularly elevated glucocorticoid responsiveness at pre-treatment may identify veterans likely to respond to PE combined with an intervention that targets glucocorticoid sensitivity. Confirmation of these findings will suggest that pharmacologic interventions that target PTSD-associated glucocorticoid dysregulation may be particularly helpful in promoting a positive clinical response to PTSD psychotherapy.
Collapse
Affiliation(s)
- Rachel Yehuda
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States; Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Linda M Bierer
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States; Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Laura C Pratchett
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States; Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Amy Lehrner
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States; Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Erin C Koch
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States
| | - Jaklyn A Van Manen
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States
| | - Janine D Flory
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States; Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Iouri Makotkine
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States; Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tom Hildebrandt
- Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
26
|
Sase S, Meyer K, Lubec G, Korz V. Different expression of membrane-bound and cytosolic hippocampal steroid receptor complexes during spatial training in young male rats. Eur Neuropsychopharmacol 2014; 24:1819-27. [PMID: 25258178 DOI: 10.1016/j.euroneuro.2014.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/31/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
Brain steroid receptors are involved in mediating stress responses and cognitive processes throughfast non-genomic signaling of membrane-bound receptors or through the slower genomic actions of cytosolic receptors. Although the contribution of these different pathways in the formation and maintenance of memories has been widely discussed, little is known about the regulation of membrane versus cytosolic receptors during a learning task. Besides the relatively well studied corticosterone-binding glucocorticoid (GR) and mineralocorticoid (MR) receptors, sex steroid hormone receptors, such as the androgen and estrogen (ERα and ERβ) receptors, have also been shown to be involved in the regulation of stress and cognition. Moreover, the latter receptors are known to be functional in both sexes. Therefore, we studied the expression of hippocampal receptors in both cellular fractions during spatial learning in male rats. Membrane and cytosolic GR were shown to be downregulated after memory acquisition and unaffected after consolidation, whereas membrane MR was upregulated after both learning phases and unaffected in the cytosol. Cytosolic ERα was downregulated after both phases and unaffected in the membrane. The remaining receptors were not regulated. The data suggest a specific role of MR and ERα during training as fast and slow mediators, respectively.
Collapse
Affiliation(s)
- Sunetra Sase
- University of Vienna, Department of Pediatrics, Vienna, Austria
| | - Katrin Meyer
- Leibniz Institute for Neurobiology, Magdeburg, Germany; University of Magdeburg, Institute of Biology, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Gert Lubec
- University of Vienna, Department of Pediatrics, Vienna, Austria
| | - Volker Korz
- University of Magdeburg, Institute of Biology, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
27
|
High-dose corticosterone after fear conditioning selectively suppresses fear renewal by reducing anxiety-like response. Pharmacol Biochem Behav 2014; 124:188-95. [PMID: 24933336 DOI: 10.1016/j.pbb.2014.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/22/2014] [Accepted: 06/07/2014] [Indexed: 11/21/2022]
Abstract
Exposure therapy is widely used to treat anxiety disorders, including posttraumatic stress disorder (PTSD). However, preventing the return of fear is still a major challenge after this behavioral treatment. An increasing number of studies suggest that high-dose glucocorticoid treatment immediately after trauma can alleviate the symptoms of PTSD in humans. Unknown is whether high-dose glucocorticoid treatment following fear conditioning suppresses the return of fear. In the present study, a typical fear renewal paradigm (AAB) was used, in which the fear response to an auditory cue can be restored in a novel context (context B) when both training and extinction occur in the same context (context A). We trained rats for auditory fear conditioning and administered corticosterone (CORT; 5 and 25mg/kg, i.p.) or vehicle with different delays (1 and 24h). Forty-eight hours after drug injection, extinction was conducted with no drug in the training context, followed by a test of tone-induced freezing behavior in the same (AAA) or a shifted (AAB) context. Both immediate and delayed administration of high-dose CORT after fear conditioning reduced fear renewal. To examine the anxiolytic effect of CORT, independent rats were trained for cued or contextual fear conditioning, followed by an injection of CORT (5 and 25mg/kg, i.p.) or vehicle at a 1 or 24h delay. One week later, anxiety-like behavior was assessed in the elevated plus maze (EPM) before and after fear expression. We found that high-dose CORT decreased anxiety-like behavior without changing tone- or context-induced freezing. These findings indicate that a single high-dose CORT administration given after fear conditioning may selectively suppress fear renewal by reducing anxiety-like behavior and not by altering the consolidation, retrieval, or extinction of fear memory.
Collapse
|
28
|
Conversi D, Cruciani F, Accoto A, Cabib S. Positive emotional arousal increases duration of memory traces: different role of dopamine D1 receptor and β-adrenoceptor activation. Pharmacol Biochem Behav 2014; 122:158-63. [PMID: 24727402 DOI: 10.1016/j.pbb.2014.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/19/2014] [Accepted: 04/02/2014] [Indexed: 11/16/2022]
Abstract
We investigated the effects of post-training administration of dopamine D1 receptor antagonist SCH 23390 and β-adrenergic receptor antagonist Propranolol on memory retention of an object sampled in a state of positive emotional arousal. Saline-treated mice trained and tested under high emotional/motivational arousal (High) showed discrimination of a novel object both 24 and 96 h post-training. Instead, mice trained and tested under low motivational arousal (Low) were unable to discriminate the novel object 96 h post-training. Both a high (2 mg/kg) and a low (1 mg/kg) dose of Propranolol reduced object discrimination in High mice tested 24 h post-training, whereas neither dose was effective in Low mice. A high dose of SCH 23390 (0.025 mg/kg) reduced discrimination of the novel object in High mice tested both 24 and 96 h post-training, whereas a low dose of the D1 antagonist (0.01 mg/kg) reduced discrimination in High mice tested 96 h post-training and abolished discrimination in Low mice tested 24h after training.
Collapse
Affiliation(s)
- D Conversi
- Department of Psychology, Center D. Bovet, University "Sapienza", Rome, Italy; Fondazione Santa Lucia IRCCS, European Centre for Brain Research, Rome, Italy.
| | - F Cruciani
- Department of Psychology, Center D. Bovet, University "Sapienza", Rome, Italy
| | - A Accoto
- Department of Psychology, Center D. Bovet, University "Sapienza", Rome, Italy
| | - S Cabib
- Department of Psychology, Center D. Bovet, University "Sapienza", Rome, Italy; Fondazione Santa Lucia IRCCS, European Centre for Brain Research, Rome, Italy
| |
Collapse
|
29
|
The endocannabinoid system: an emotional buffer in the modulation of memory function. Neurobiol Learn Mem 2013; 112:30-43. [PMID: 24382324 DOI: 10.1016/j.nlm.2013.12.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 12/16/2013] [Accepted: 12/20/2013] [Indexed: 01/12/2023]
Abstract
Extensive evidence indicates that endocannabinoids modulate cognitive processes in animal models and human subjects. However, the results of endocannabinoid system manipulations on cognition have been contradictory. As for anxiety behavior, a duality has indeed emerged with regard to cannabinoid effects on memory for emotional experiences. Here we summarize findings describing cannabinoid effects on memory acquisition, consolidation, retrieval and extinction. Additionally, we review findings showing how the endocannabinoid system modulates memory function differentially, depending on the level of stress and arousal associated with the experimental context. Based on the evidence reviewed here, we propose that the endocannabinoid system is an emotional buffer that moderates the effects of environmental context and stress on cognitive processes.
Collapse
|
30
|
Hauer D, Kaufmann I, Strewe C, Briegel I, Campolongo P, Schelling G. The role of glucocorticoids, catecholamines and endocannabinoids in the development of traumatic memories and posttraumatic stress symptoms in survivors of critical illness. Neurobiol Learn Mem 2013; 112:68-74. [PMID: 24125890 DOI: 10.1016/j.nlm.2013.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 10/01/2013] [Accepted: 10/03/2013] [Indexed: 12/23/2022]
Abstract
Critically ill patients are at an increased risk for traumatic memories and post-traumatic stress disorder (PTSD). Memories of one or more traumatic events play an important part in the symptom pattern of PTSD. Studies in long-term survivors of intensive care unit (ICU) treatment demonstrated a clear and vivid recall of traumatic experiences and the incidence and intensity of PTSD symptoms increased with the number of traumatic memories present. Preclinical evidence has clearly shown that the consolidation and retrieval of traumatic memories is regulated by an interaction between the noradrenergic, the glucocorticoid and the endocannabinoid system. Critically ill patients in the ICU frequently require treatment with adrenenergic or glucocorticoid drugs and often receive sedative medications; among them propofol is known to influence endocannabinoid signaling. Critical illness could therefore represent a useful model for investigating adrenergic, glucocorticoid as well as endocannabinoid effects on traumatic memory and PTSD development in stressed humans. The endocannabinoid system is an important regulator of HPA-axis activity during stress, an effect which has also been demonstrated in humans. Likewise, a single nucleotide polymorphism (SNP) of the glucocorticoid receptor (GR) gene (the BclI-SNP), which enhances the sensitivity of the glucocorticoid receptors to cortisol and possibly HPA-axis feedback function, was associated with enhanced emotional memory performance in healthy volunteers. The presence of the BclI-SNP increased the risk for traumatic memories and PTSD symptoms in patients after ICU therapy and was linked to lower basal cortisol levels. A number of small studies have demonstrated that the administration of cortisol to critically ill or injured patients results in a significant reduction of PTSD symptoms after recovery without influencing the number of traumatic memories. These glucocorticoid effects can possibly be explained by a cortisol-induced temporary impairment in traumatic memory retrieval which has previously been demonstrated in both rats and humans. The hypothesis that stress doses of glucocorticoids or the pharmacologic manipulation of glucocorticoid-endocannabinoid interaction during traumatic memory consolidation and retrieval could be useful for prophylaxis and treatment of PTSD after critical illness should be tested in larger controlled studies.
Collapse
Affiliation(s)
- Daniela Hauer
- Department of Anaesthesiology, University of Munich, Campus Grosshadern, 81377 Munich, Germany
| | - Ines Kaufmann
- Department of Anaesthesiology, University of Munich, Campus Grosshadern, 81377 Munich, Germany
| | - Claudia Strewe
- Department of Anaesthesiology, University of Munich, Campus Grosshadern, 81377 Munich, Germany
| | - Isabel Briegel
- Department of Anaesthesiology, University of Munich, Campus Grosshadern, 81377 Munich, Germany
| | - Patrizia Campolongo
- Department of Anaesthesiology, University of Munich, Campus Grosshadern, 81377 Munich, Germany; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Gustav Schelling
- Department of Anaesthesiology, University of Munich, Campus Grosshadern, 81377 Munich, Germany.
| |
Collapse
|
31
|
Takatsu-Coleman AL, Zanin KA, Patti CL, Zager A, Lopes-Silva LB, Longo BM, Tufik S, Andersen ML, Frussa-Filho R. Short-term sleep deprivation reinstates memory retrieval in mice: the role of corticosterone secretion. Psychoneuroendocrinology 2013; 38:1967-78. [PMID: 23545263 DOI: 10.1016/j.psyneuen.2013.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 01/23/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
While the effects of sleep deprivation (SD) on the acquisition and consolidation phases of memory have been extensively characterized, its effects on memory retrieval remain overlooked. SD alone is a stressor, and stress-activated glucocorticoids promote bimodal effects on memory. Because we have recently demonstrated that 72h SD impairs memory retrieval in the plus-maze discriminative avoidance task (PM-DAT) in mice, this study investigated whether shorter SD periods would facilitate retrieval. In Experiment I, the temporal forgetting curve of the PM-DAT was determined and an interval between training/testing in which retrieval was no longer present was used in all subsequent experiments. In Experiments II and III, retrieval performance and corticosterone concentration, respectively, were quantified in mice that were sleep deprived for 12 or 24h before testing. In Experiments IV and V, the effects of the corticosterone synthesis inhibitor metyrapone were evaluated on 12h SD-induced retrieval reinstatement and corticosterone concentration enhancement, respectively. Experiment VI determined whether pre-test acute administration of exogenous corticosterone would mimic the facilitatory effects of 12h SD on retrieval. Thirty days after training, mice presented poor performance of the task; however, SD for 12h (but not for 24) before testing reinstated memory retrieval. This facilitatory effect was accompanied by increased corticosterone concentration, abolished by metyrapone, and mimicked by pre-test acute corticosterone administration. Collectively, short-term SD can facilitate memory retrieval by enhancing corticosterone secretion. This facilitatory effect is abolished by longer periods of SD.
Collapse
Affiliation(s)
- André L Takatsu-Coleman
- Departamento de Farmacologia, Universidade Federal de São Paulo, Rua Botucatu 862, Ed. Leal Prado, 1(o) andar, 04023062 São Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Glucocorticoid-induced enhancement of contextual fear memory consolidation in rats: Involvement of D1 receptor activity of hippocampal area CA1. Brain Res 2013; 1524:26-33. [DOI: 10.1016/j.brainres.2013.05.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 05/01/2013] [Accepted: 05/19/2013] [Indexed: 01/01/2023]
|
33
|
Propranolol reduces cognitive deficits, amyloid β levels, tau phosphorylation and insulin resistance in response to chronic corticosterone administration. Int J Neuropsychopharmacol 2013. [PMID: 23194475 DOI: 10.1017/s1461145712001393] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic exposure to glucocorticoids might result not only in insulin resistance or cognitive deficits, but it is also considered as a risk factor for pathologies such as Alzheimer's disease. Propranolol is a β-adrenergic antagonist commonly used in the treatment of hypertension or acute anxiety. The effects of propranolol (5 mg/kg) have been tested in a model of chronic corticosterone administration (100 μg/ml, 4 wk) in drinking water. Corticosterone administration led to cognitive impairment in the novel object recognition test that was reversed by propranolol. Increased levels of Aβ in the hippocampus of corticosterone-treated mice were counteracted by propranolol treatment, purportedly through an increased IDE expression. Chronic corticosterone treatment induced responses characteristic of insulin resistance, as increased peripheral insulin levels, decreased activation of the insulin receptor (pIR) and decreased associated intracellular pathways (pAkt). These effects might be related to a decreased c-Jun N terminal kinase 1 expression. Again, propranolol was able to counteract all corticosterone-induced effects. One of the main kinases involved in tau phosphorylation, glycogen synthase kinase 3β (GSK3β), which is inactivated by phosphorylation by pAkt, was found to be decreased after corticosterone and increased after propranolol treatment. Concomitant changes in pTau expression were found. Overall, these data further strengthen the potential of propranolol as a therapeutic agent for pathologies associated with the interaction glucocorticoids-insulin resistance and the development of relevant cellular processes for Alzheimer's disease.
Collapse
|
34
|
McReynolds JR, McIntyre CK. Emotional modulation of the synapse. Rev Neurosci 2013; 23:449-61. [PMID: 23096101 DOI: 10.1515/revneuro-2012-0047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/05/2012] [Indexed: 11/15/2022]
Abstract
Acute stress and emotional arousal can enhance the consolidation of long-term memories in a manner that is dependent on β -adrenoceptor activation in the basolateral complex of the amygdala (BLA). The BLA interacts with multiple memory systems in the brain to modulate a variety of classes of memory. However, the synaptic mechanisms of this interaction remain unresolved. This review describes the evidence of modulation of memory and synaptic plasticity produced by emotional arousal,stress hormones, and pharmacological or electrophysiological stimulation of the amygdala. The amygdala modulation of local translation and/or degradation of the synaptic plasticity-related proteins, activity-regulated cytoskeletal-associated protein and calcium/calmodulin dependent protein kinase II α , is offered as a potential mechanism for the rapid memory consolidation that is associated with emotionally arousing events. This model shares features with synaptic tagging and the emotional tagging hypotheses.
Collapse
Affiliation(s)
- Jayme R McReynolds
- Department of Behavioral and BrainSciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| | | |
Collapse
|
35
|
Campolongo P, Morena M, Scaccianoce S, Trezza V, Chiarotti F, Schelling G, Cuomo V, Roozendaal B. Novelty-induced emotional arousal modulates cannabinoid effects on recognition memory and adrenocortical activity. Neuropsychopharmacology 2013; 38:1276-86. [PMID: 23340520 PMCID: PMC3656371 DOI: 10.1038/npp.2013.26] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Although it is well established that cannabinoid drugs can influence cognitive performance, the findings-describing both enhancing and impairing effects-have been ambiguous. Here, we investigated the effects of posttraining systemic administration of the synthetic cannabinoid agonist WIN55,212-2 (0.1, 0.3, or 1.0 mg/kg) on short- and long-term retention of object recognition memory under two conditions that differed in their training-associated arousal level. In male Sprague-Dawley rats that were not previously habituated to the experimental context, WIN55,212-2 administered immediately after a 3-min training trial, biphasically impaired retention performance at a 1-h interval. In contrast, WIN55,212-2 enhanced 1-h retention of rats that had received extensive prior habituation to the experimental context. Interestingly, immediate posttraining administration of WIN55,212-2 to non-habituated rats, in doses that impaired 1-h retention, enhanced object recognition performance at a 24-h interval. Posttraining WIN55,212-2 administration to habituated rats did not significantly affect 24-h retention. In light of intimate interactions between cannabinoids and the hypothalamic-pituitary-adrenal axis, we further investigated whether cannabinoid administration might differently influence training-induced glucocorticoid activity in rats in these two habituation conditions. WIN55,212-2 administered after object recognition training elevated plasma corticosterone levels in non-habituated rats whereas it decreased corticosterone levels in habituated rats. Most importantly, following pretreatment with the corticosterone-synthesis inhibitor metyrapone, WIN55,212-2 effects on 1- and 24-h retention of non-habituated rats became similar to those seen in the low-aroused habituated animals, indicating that cannabinoid-induced regulation of adrenocortical activity contributes to the environmentally sensitive effects of systemically administered cannabinoids on short- and long-term retention of object recognition memory.
Collapse
Affiliation(s)
- Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le A. Moro 5, Rome, Italy.
| | - Maria Morena
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le A. Moro 5, Rome, Italy
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le A. Moro 5, Rome, Italy
| | - Viviana Trezza
- Department of Biology, University of Roma Tre, Viale Marconi 446, Rome, Italy
| | - Flavia Chiarotti
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neuroscience, Instituto Superiore di Sanità, Rome, Italy
| | - Gustav Schelling
- Department of Anaesthesiology, Ludwig-Maximilians University, Munich, Germany
| | - Vincenzo Cuomo
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le A. Moro 5, Rome, Italy
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands,Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| |
Collapse
|
36
|
An XL, Zheng XG, Liang J, Bai YJ. Corticosterone combined with intramedial prefrontal cortex infusion of SCH 23390 impairs the strong fear response in high-fear-reactivity rats. Psych J 2013; 2:1-10. [PMID: 26272859 DOI: 10.1002/pchj.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 03/15/2012] [Indexed: 11/07/2022]
Abstract
Accumulating evidence suggests that stress-dose corticosteroids impair fear memory in animals and humans. Corticosteroid treatment after critical illness is seen as a potential psychotropic medication by which to prevent posttraumatic stress disorder. However, individual difference in the responsiveness to stress (i.e., stress reactivity) is a factor that modulates the efficacy of corticosteroids. To understand the contribution of fear reactivity to the effect of post-stress corticosterone, male Sprague-Dawley rats were subjected to classical tone-cued fear conditioning and separated into high and low reactivity (HR and LR, respectively) responder groups based on their levels of freezing during conditioning. The HR rats showed significantly higher fear responses than the LR rats during conditioning as assessed by freezing behavior. At two intervals, 30 min and 48 hr later, the HR rats still displayed more pronounced conditioned responses to cued stimuli compared with the LR rats. Moreover, in contrast to the LR rats, the enhanced fear response in the HR rats was difficult to attenuate by post-training high-dose corticosterone. These results suggest that fear reactivity results in stronger fear memory, and that it is difficult to disrupt this strong fear memory in the HR phenotype using monotherapy. However, the strong fear memory in the HR rats was impaired by concurrent intramedial prefrontal cortex infusion of a high dose of the dopamine D1 receptor antagonist SCH 23390 and systemic administration of corticosterone. SCH 23390 and corticosterone alone did not decrease freezing levels in the HR rats. The fear impairment induced by SCH 23390 combined with corticosterone was not attributable to the effect of these drugs on locomotor activity. This effect was not found with administration of the D2 antagonist eticlopride combined with corticosterone. Our findings demonstrate that the conditioned fear memory in individuals with high stress reactivity is difficult to disrupt using monotherapy, but that combined pharmacotherapy may be useful for treating intervention-resistant fear.
Collapse
Affiliation(s)
- Xian-Li An
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.,Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Xi-Geng Zheng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Jing Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Yun-Jing Bai
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
37
|
Zarrindast MR, Mashayekhi M, Rezayof A, Ahmadi S. β-Adrenoceptors in the dorsal hippocampus are involved in ethanol-induced state-dependent retrieval in mice. Neurobiol Learn Mem 2013; 100:12-7. [DOI: 10.1016/j.nlm.2012.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 11/28/2012] [Accepted: 11/28/2012] [Indexed: 11/27/2022]
|
38
|
Hawley WR, Grissom EM, Patel JM, Hodges KS, Dohanich GP. Reactivation of an aversive memory modulates learning strategy preference in male rats. Stress 2013; 16:73-86. [PMID: 22533611 DOI: 10.3109/10253890.2012.683466] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Reminders of an aversive event adversely impact retrieval of hippocampus-dependent memories and exacerbate stress-induced levels of anxiety. Interestingly, stress and anxiety shift control over learning away from the hippocampus and toward the striatum. The aims of the current study were to determine whether spatial memory and learning strategy are impacted by reminders of a stressor. Adult male Long-Evans rats (N = 47) were subjected to an inhibitory avoidance (IA) training trial in which 32 rats were exposed (3 s) to a single inescapable electrical footshock (0.6 mA). Prior to the retention trial of a Y-maze task and the probe trials of two different learning strategy tasks, some of the rats that were exposed to the footshock (n = 17) were reminded of the stressor on an IA retrieval trial. Both groups of rats exposed to the initial stressor exhibited hypoactivity, but no impairment in spatial memory, on the Y-maze task conducted 1 week after exposure to the footshock. One month after exposure to footshock, both groups of rats exposed to the initial stressor tended to prefer a striatum-dependent learning strategy on a water T-maze task. However, 2 months after exposure to footshock, only shocked rats that were reminded of the stressor exhibited a preference for a striatum-dependent learning strategy on a visible-platform water maze task, which corresponded with lower levels of activity in an open field. The results indicate that reminders of a stressor perpetuate the deleterious effects of stress on affective and cognitive processes.
Collapse
Affiliation(s)
- Wayne R Hawley
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA
| | | | | | | | | |
Collapse
|
39
|
Mochny CR, Kincheski GC, Molina VA, Carobrez AP. Dorsolateral periaqueductal gray stimulation prior to retrieval potentiates a contextual fear memory in rats. Behav Brain Res 2013; 237:76-81. [DOI: 10.1016/j.bbr.2012.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 09/06/2012] [Accepted: 09/10/2012] [Indexed: 11/27/2022]
|
40
|
Joëls M, Sarabdjitsingh RA, Karst H. Unraveling the Time Domains of Corticosteroid Hormone Influences on Brain Activity: Rapid, Slow, and Chronic Modes. Pharmacol Rev 2012; 64:901-38. [DOI: 10.1124/pr.112.005892] [Citation(s) in RCA: 305] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
41
|
Zhou M, Hoogenraad CC, Joëls M, Krugers HJ. Combined β-adrenergic and corticosteroid receptor activation regulates AMPA receptor function in hippocampal neurons. J Psychopharmacol 2012; 26:516-24. [PMID: 21965192 DOI: 10.1177/0269881111424930] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Shortly after stress, limbic neurons are exposed to high levels of noradrenaline and corticosterone. These hormones are necessary for optimal behavioural adaptation. Behavioural effects critically depend on noradrenaline acting via β-adrenergic receptors, but these effects are strongly modulated by corticosterone, indicating putative interactions between the two hormones. Since both noradrenaline and corticosterone are known to quickly affect properties of AMPA-type glutamate receptors (AMPAR), we here examined - in hippocampal neurons - three parameters which give insight in the functionality of AMPARs: phosphorylation, surface expression and spontaneous synaptic transmission. In homogenates of adult hippocampal slices, application of corticosterone (30 nM for 15 min) by itself did not affect phosphorylation of the AMPAR GluA1 subunit at S845 or S831. Co-application of the β-adrenergic receptor agonist isoproterenol (10 µM) largely increased S845 (but not S831) phosphorylation. Corticosterone also did not change GluA1 and GluA2 surface expression in hippocampal primary cultures. However, combined administration of corticosterone and 1 µM isoproterenol - which by itself was ineffective - enhanced surface expression. Interestingly, 10 µM isoproterenol alone enhanced GluA1 surface expression, but this was decreased by corticosterone. Finally, in hippocampal primary cultures, the inter-event interval of miniature excitatory postsynaptic currents (mEPSCs) was decreased by the combination of 1 µM isoproterenol and corticosterone (which were ineffective by themselves) while the same combination did not affect the amplitude. We conclude that AMPAR phosphorylation, surface expression and mEPSC inter-event interval respond most strongly to a combination of corticosterone and β-adrenergic receptors. These combined hormonal effects on glutamate transmission might contribute to their memory-enhancing effects.
Collapse
Affiliation(s)
- Ming Zhou
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | |
Collapse
|
42
|
Atsak P, Roozendaal B, Campolongo P. Role of the endocannabinoid system in regulating glucocorticoid effects on memory for emotional experiences. Neuroscience 2012; 204:104-16. [DOI: 10.1016/j.neuroscience.2011.08.047] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 08/16/2011] [Accepted: 08/22/2011] [Indexed: 11/25/2022]
|
43
|
Glucocorticoids interact with the hippocampal endocannabinoid system in impairing retrieval of contextual fear memory. Proc Natl Acad Sci U S A 2012; 109:3504-9. [PMID: 22331883 DOI: 10.1073/pnas.1200742109] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
There is extensive evidence that glucocorticoid hormones impair the retrieval of memory of emotionally arousing experiences. Although it is known that glucocorticoid effects on memory retrieval impairment depend on rapid interactions with arousal-induced noradrenergic activity, the exact mechanism underlying this presumably nongenomically mediated glucocorticoid action remains to be elucidated. Here, we show that the hippocampal endocannabinoid system, a rapidly activated retrograde messenger system, is involved in mediating glucocorticoid effects on retrieval of contextual fear memory. Systemic administration of corticosterone (0.3-3 mg/kg) to male Sprague-Dawley rats 1 h before retention testing impaired the retrieval of contextual fear memory without impairing the retrieval of auditory fear memory or directly affecting the expression of freezing behavior. Importantly, a blockade of hippocampal CB1 receptors with AM251 prevented the impairing effect of corticosterone on retrieval of contextual fear memory, whereas the same impairing dose of corticosterone increased hippocampal levels of the endocannabinoid 2-arachidonoylglycerol. We also found that antagonism of hippocampal β-adrenoceptor activity with local infusions of propranolol blocked the memory retrieval impairment induced by the CB receptor agonist WIN55,212-2. Thus, these findings strongly suggest that the endocannabinoid system plays an intermediary role in regulating rapid glucocorticoid effects on noradrenergic activity in impairing memory retrieval of emotionally arousing experiences.
Collapse
|
44
|
Li S, Fan YX, Wang W, Tang YY. Effects of acute restraint stress on different components of memory as assessed by object-recognition and object-location tasks in mice. Behav Brain Res 2012; 227:199-207. [DOI: 10.1016/j.bbr.2011.10.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 09/30/2011] [Accepted: 10/04/2011] [Indexed: 12/11/2022]
|
45
|
Segev A, Ramot A, Akirav I. Stress hormones receptors in the amygdala mediate the effects of stress on the consolidation, but not the retrieval, of a non aversive spatial task. PLoS One 2012; 7:e29988. [PMID: 22253850 PMCID: PMC3256198 DOI: 10.1371/journal.pone.0029988] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 12/11/2011] [Indexed: 11/25/2022] Open
Abstract
This study examined the effects of the arousal level of the rat and exposure to a behavioral stressor on acquisition, consolidation and retrieval of a non-aversive hippocampal-dependent learning paradigm, the object location task. Learning was tested under two arousal conditions: no previous habituation to the experimental context (high novelty stress/arousal level) or extensive prior habituation (reduced novelty stress/arousal level). Results indicated that in the habituated rats, exposure to an out-of-context stressor (i.e, elevated platform stress) impaired consolidation and retrieval, but not acquisition, of the task. Non-habituated animals under both stressed and control conditions did not show retention of the task. In habituated rats, RU-486 (10 ng/side), a glucocorticoid receptor (GR) antagonist, or propranolol (0.75 µg/side), a beta-adrenergic antagonist, injected into the basolateral amygdala (BLA), prevented the impairing effects of the stressor on consolidation, but not on retrieval. The CB1/CB2 receptor agonist WIN55,212-2 (WIN, 5 µg/side) microinjected into the BLA did not prevent the effects of stress on either consolidation or retrieval. Taken together the results suggest that: (i) GR and β-adrenergic receptors in the BLA mediate the impairing effects of stress on the consolidation, but not the retrieval, of a neutral, non-aversive hippocampal-dependent task, (ii) the impairing effects of stress on hippocampal consolidation and retrieval are mediated by different neural mechanisms (i.e., different neurotransmitters or different brain areas), and (iii) the effects of stress on memory depend on the interaction between several main factors such as the stage of memory processing under investigation, the animal's level of arousal and the nature of the task (neutral or aversive).
Collapse
Affiliation(s)
- Amir Segev
- Department of Psychology, University of Haifa, Haifa, Israel
| | - Assaf Ramot
- Department of Psychology, University of Haifa, Haifa, Israel
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa, Israel
- * E-mail:
| |
Collapse
|
46
|
Abstract
Our memories are not all created equally strong: Some experiences are well remembered while others are remembered poorly, if at all. Research on memory modulation investigates the neurobiological processes and systems that contribute to such differences in the strength of our memories. Extensive evidence from both animal and human research indicates that emotionally significant experiences activate hormonal and brain systems that regulate the consolidation of newly acquired memories. These effects are integrated through noradrenergic activation of the basolateral amygdala that regulates memory consolidation via interactions with many other brain regions involved in consolidating memories of recent experiences. Modulatory systems not only influence neurobiological processes underlying the consolidation of new information, but also affect other mnemonic processes, including memory extinction, memory recall, and working memory. In contrast to their enhancing effects on consolidation, adrenal stress hormones impair memory retrieval and working memory. Such effects, as with memory consolidation, require noradrenergic activation of the basolateral amygdala and interactions with other brain regions.
Collapse
Affiliation(s)
- Benno Roozendaal
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, The Netherlands.
| | | |
Collapse
|
47
|
Stress and glucocorticoids impair memory retrieval via β2-adrenergic, Gi/o-coupled suppression of cAMP signaling. J Neurosci 2011; 31:14172-81. [PMID: 21976502 DOI: 10.1523/jneurosci.2122-11.2011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acute stress impairs the retrieval of hippocampus-dependent memory, and this effect is mimicked by exogenous administration of stress-responsive glucocorticoid hormones. It has been proposed that glucocorticoids affect memory by promoting the release and/or blocking the reuptake of norepinephrine (NE), a stress-responsive neurotransmitter. It has also been proposed that this enhanced NE signaling impairs memory retrieval by stimulating β(1)-adrenergic receptors and elevating levels of cAMP. In contrast, other evidence indicates that NE, β(1), and cAMP signaling is transiently required for the retrieval of hippocampus-dependent memory. To resolve this discrepancy, wild-type rats and mice with and without gene-targeted mutations were stressed or treated with glucocorticoids and/or adrenergic receptor drugs before testing memory for inhibitory avoidance or fear conditioning. Here we report that glucocorticoids do not require NE to impair retrieval. However, stress- and glucocorticoid-induced impairments of retrieval depend on the activation of β(2) (but not β(1))-adrenergic receptors. Offering an explanation for the opposing functions of these two receptors, the impairing effects of stress, glucocorticoids and β(2) agonists on retrieval are blocked by pertussis toxin, which inactivates signaling by G(i/o)-coupled receptors. In hippocampal slices, β(2) signaling decreases cAMP levels and greatly reduces the increase in cAMP mediated by β(1) signaling. Finally, augmenting cAMP signaling in the hippocampus prevents the impairment of retrieval by systemic β(2) agonists or glucocorticoids. These results demonstrate that the β(2) receptor can be a critical effector of acute stress, and that β(1) and β(2) receptors can have quite distinct roles in CNS signaling and cognition.
Collapse
|
48
|
Fan SJ, Jiang H, Yang LJ, Liu X, Song J, Pan F. Effects of adrenergic agents on stress-induced brain microstructural and immunochemical changes in adult male Wistar rats. Ann Anat 2011; 193:418-24. [DOI: 10.1016/j.aanat.2011.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 06/04/2011] [Accepted: 06/06/2011] [Indexed: 12/31/2022]
|
49
|
Cannabinoid receptor involvement in stress-induced cocaine reinstatement: potential interaction with noradrenergic pathways. Neuroscience 2011; 204:117-24. [PMID: 21871539 DOI: 10.1016/j.neuroscience.2011.08.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Revised: 08/04/2011] [Accepted: 08/10/2011] [Indexed: 11/24/2022]
Abstract
This study examined the role of endocannabinoid signaling in stress-induced reinstatement of cocaine seeking and explored the interaction between noradrenergic and endocannabinergic systems in the process. A well-validated preclinical model for human relapse, the rodent conditioned place preference assay, was used. Cocaine-induced place preference was established in C57BL/6 mice using injections of 15 mg/kg cocaine. Following extinction of preference for the cocaine-paired environment, reinstatement of place preference was determined following 6 min of swim stress or cocaine injection (15 mg/kg, i.p.). The role of endocannabinoid signaling was studied using the cannabinoid antagonist AM-251 (3 mg/kg, i.p.). Another cohort of mice was tested for reinstatement following administration of the cannabinoid agonist CP 55,940 (10, 20, or 40 μg/kg, i.p.). The alpha-2 adrenergic antagonist BRL-44408 (5 mg/kg, i.p.) with or without CP 55,940 (20 μg/kg) was administered to a third group of mice. We found that: (1) AM-251 blocked forced swim-induced, but not cocaine-induced, reinstatement of cocaine-seeking behavior; (2) the cannabinoid agonist CP 55,940 did not reinstate cocaine-seeking behavior when administered alone but did synergize with a non-reinstating dose of the alpha-2 adrenergic antagonist BRL-44408 to cause reinstatement. These results are consistent with the hypothesis that stress exposure triggers the endogenous activation of CB1 receptors and that activation of the endocannabinoid system is required for the stress-induced relapse of the mice to cocaine seeking. Further, the data suggest that the endocannabinoid system interacts with noradrenergic mechanisms to influence stress-induced reinstatement of cocaine-seeking behavior.
Collapse
|
50
|
Cohen H, Kaplan Z, Koresh O, Matar MA, Geva AB, Zohar J. Early post-stressor intervention with propranolol is ineffective in preventing posttraumatic stress responses in an animal model for PTSD. Eur Neuropsychopharmacol 2011; 21:230-40. [PMID: 21194896 DOI: 10.1016/j.euroneuro.2010.11.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 11/22/2010] [Accepted: 11/30/2010] [Indexed: 10/18/2022]
Abstract
The therapeutic value of β-adrenoceptor blockage, using propranolol, in the aftermath of traumatic experience is uncertain. A prospective, controlled animal model of posttraumatic stress disorder (PTSD) was employed to assess the effects of propranolol on long-term behavioral responses to stress. Animals exposed to predator scent stress received a single bolus of propranolol (10 or 15mg/kg) or vehicle 1h post-exposure. Outcomes were assessed using the elevated plus-maze (EPM) and acoustic startle response (ASR) at 30days and freezing response to a trauma reminder (unsoiled litter) on Day 31. Individual animals were classified as having "extreme", "partial" and "minimal" behavioral responses, according to pre-set cut-off criteria for EPM and ASR response patterns. The physiological efficacy of the doses of propranolol was verified by collecting cardiovascular data telemetrically (from exposed or unexposed individuals given propranolol or vehicle). The effect of propranolol on long-term memory was verified using a non-spatial memory task. Both doses of propranolol effectively reduced mean heart rate and impaired the object-recognition task, as expected. No significant effect on prevalence rates of PTSD-like behavioral responses or on trauma reminder response was observed for either dose of propranolol as compared to vehicle. Despite adequate efficacy in terms of heart rate and disruption of memory, single-dose, post-stress β-blockage with propranolol was ineffective in reducing onset of PTSD-like behavioral disruption and trauma cue responses in the long term. Traumatic stress-related processes appear to be affected differently than the others.
Collapse
Affiliation(s)
- Hagit Cohen
- Ministry of Health Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | | | |
Collapse
|