1
|
Karst H, Riera Llobet A, Joëls M, van der Veen R. Complex housing in adulthood state-dependently affects the excitation-inhibition balance in the infralimbic prefrontal cortex of male C57Bl/6 mice. Behav Brain Res 2025; 476:115233. [PMID: 39233145 DOI: 10.1016/j.bbr.2024.115233] [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: 06/05/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024]
Abstract
The prefrontal cortex (PFC) plays an important role in social behavior and is sensitive to stressful circumstances. Challenging life conditions might change PFC function and put individuals at risk for maladaptive social behavior. The excitation-inhibition (EI) balance of prefrontal neurons appears to play a crucial role in this process. Here, we examined how a challenging life condition in C57BL/6JolaHsd mice, i.e. group-housing 6 mice in a complex environment for 10 days in adulthood, changes the EI-balance of infralimbic prefrontal neurons in layer 2/3, compared to standard pair-housing. Slices were prepared from "undisturbed" mice, i.e. the first mouse taken from the cage, or mice taken ∼15 min later, who were mildly aroused after removal of the first mouse. We observed a housing-condition by arousal-state interaction, with in the complex housing group an elevated EI-balance in undisturbed and reduced EI-balance in mildly aroused animals, while no differences were observed in standard housed animals. The change was explained by a shift in mIPSC and mEPSC frequency, while amplitudes remained unaffected. Female mice showed no housing-by-state interaction, but a main effect of housing was found for mIPSCs, with a higher frequency in complex- versus standard-housed females. No effects were observed in males who were complex-housed from a young age onwards. Explorative investigations support a potential mediating role of corticosterone in housing effects on the EI-balance of males. We argue that taking the arousal state of individuals into account is necessary to better understand the consequences of exposure to challenging life conditions for prefrontal function.
Collapse
Affiliation(s)
- Henk Karst
- University of Amsterdam, Swammerdam Institute of Life Science, Amsterdam, the Netherlands; Utrecht University, University Medical Center Utrecht, Translational Neuroscience, Utrecht, the Netherlands
| | - Arianna Riera Llobet
- University of Amsterdam, Swammerdam Institute of Life Science, Amsterdam, the Netherlands
| | - Marian Joëls
- Utrecht University, University Medical Center Utrecht, Translational Neuroscience, Utrecht, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rixt van der Veen
- University of Amsterdam, Swammerdam Institute of Life Science, Amsterdam, the Netherlands; Centre for Urban Mental Health (UMH), University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Lingg RT, Johnson SB, Hinz DC, Skog TD, Lizarazu M, Romig-Martin SA, LaLumiere RT, Narayanan NS, Radley JJ. Prefrontal projections to the bed nuclei of the stria terminalis modulate the specificity of aversive memories. RESEARCH SQUARE 2024:rs.3.rs-4241372. [PMID: 39569181 PMCID: PMC11577250 DOI: 10.21203/rs.3.rs-4241372/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Generalizing aversive memories helps organisms avoid danger, whereas discriminating between dissimilar situations promotes opportunistic behaviors. We identified a novel pathway that controls the contextual specificity of memory consolidation of inhibitory avoidance learning. Optogenetic inhibition of the rostral medial prefrontal cortex (mPFC)-to-anteroventral bed nuclei of the stria terminalis (avBST) pathway after a single footshock exacerbated stress hormonal output, and 2 d later promoted generalization to a novel context. Rostral mPFC-avBST influences were directly mnemonic rather than associated with stress hormone increases, as adrenalectomy did not prevent such influences on generalization. We next observed that fear discrimination between novel and aversive contexts engaged activity along the rostral mPFC and avBST pathway. Finally, post-footshock optogenetic pathway excitation enhanced 2-d discrimination. These findings highlight a prefrontal pathway in which activity immediately after aversive experiences promotes mnemonic discrimination between threatening and non-threatening contexts and may be importance for understanding trauma generalization in psychiatric illnesses.
Collapse
Affiliation(s)
- Ryan T. Lingg
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Shane B. Johnson
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
| | - Dalton C. Hinz
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Timothy D. Skog
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
| | - Manuela Lizarazu
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Sara A. Romig-Martin
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Ryan T. LaLumiere
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Nandakumar S. Narayanan
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Jason J. Radley
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
3
|
Schwabe L. Memory Under Stress: From Adaptation to Disorder. Biol Psychiatry 2024:S0006-3223(24)01385-4. [PMID: 38880463 DOI: 10.1016/j.biopsych.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/02/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Stressful events are ubiquitous in everyday life. Exposure to these stressors initiates the temporally orchestrated release of a multitude of hormones, peptides, and neurotransmitters that target brain areas that have been critically implicated in learning and memory. This review summarizes recent insights on the profound impact of stress on 4 fundamental processes of memory: memory formation, memory contextualization, memory retrieval, and memory flexibility. Stress mediators instigate dynamic alterations in these processes, thereby facilitating efficient responding under stress and the creation of a decontextualized memory representation that can effectively aid coping with novel future threats. While they are generally adaptive, the same stress-related changes may contribute to the rigid behaviors, uncontrollable intrusions, and generalized fear responding seen in anxiety disorders and posttraumatic stress disorder. Drawing on recent discoveries in cognitive neuroscience and psychiatry, this review discusses how stress-induced alterations in memory processes can simultaneously foster adaptation to stressors and fuel psychopathology. The transition from adaptive to maladaptive changes in the impact of stress on memory hinges on the nuanced interplay of stressor characteristics and individual predispositions. Thus, taking individual differences in the cognitive response to stressors into account is essential for any successful treatment of stress-related mental disorders.
Collapse
Affiliation(s)
- Lars Schwabe
- Department of Cognitive Psychology, Institute of Psychology, Universität Hamburg, Hamburg, Germany.
| |
Collapse
|
4
|
Mbiydzenyuy NE, Joanna Hemmings SM, Shabangu TW, Qulu-Appiah L. Exploring the influence of stress on aggressive behavior and sexual function: Role of neuromodulator pathways and epigenetics. Heliyon 2024; 10:e27501. [PMID: 38486749 PMCID: PMC10937706 DOI: 10.1016/j.heliyon.2024.e27501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
Stress is a complex and multifaceted phenomenon that can significantly influence both aggressive behavior and sexual function. This review explores the intricate relationship between stress, neuromodulator pathways, and epigenetics, shedding light on the various mechanisms that underlie these connections. While the role of stress in both aggression and sexual behavior is well-documented, the mechanisms through which it exerts its effects are multifarious and not yet fully understood. The review begins by delving into the potential influence of stress on the Hypothalamic-Pituitary-Adrenal (HPA) axis, glucocorticoids, and the neuromodulators involved in the stress response. The intricate interplay between these systems, which encompasses the regulation of stress hormones, is central to understanding how stress may contribute to aggressive behavior and sexual function. Several neuromodulator pathways are implicated in both stress and behavior regulation. We explore the roles of norepinephrine, serotonin, oxytocin, and androgens in mediating the effects of stress on aggression and sexual function. It is important to distinguish between general sexual behavior, sexual motivation, and the distinct category of "sexual aggression" as separate constructs, each necessitating specific examination. Additionally, epigenetic mechanisms emerge as crucial factors that link stress to changes in gene expression patterns and, subsequently, to behavior. We then discuss how epigenetic modifications can occur in response to stress exposure, altering the regulation of genes associated with stress, aggression, and sexual function. While numerous studies support the association between epigenetic changes and stress-induced behavior, more research is necessary to establish definitive links. Throughout this exploration, it becomes increasingly clear that the relationship between stress, neuromodulator pathways, and epigenetics is intricate and multifaceted. The review emphasizes the need for further research, particularly in the context of human studies, to provide clinical significance and to validate the existing findings from animal models. By better understanding how stress influences aggressive behavior and sexual function through neuromodulator pathways and epigenetic modifications, this research aims to contribute to the development of innovative protocols of precision medicine and more effective strategies for managing the consequences of stress on human behavior. This may also pave way for further research into risk factors and underlying mechanisms that may associate stress with sexual aggression which finds application not only in neuroscience, but also law, ethics, and the humanities in general.
Collapse
Affiliation(s)
- Ngala Elvis Mbiydzenyuy
- Basic Science Department, School of Medicine, Copperbelt University, P.O Box 71191, Ndola, Zambia
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Private Bag X1, Matieland, 7602, Cape Town South Africa
| | - Sian Megan Joanna Hemmings
- Division of Molecular Biology & Human Genetics, Biomedical Science Research Institute, Stellenbosch University, Private Bag X1, Matieland, 7602, Cape Town South Africa
| | - Thando W. Shabangu
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Private Bag X1, Matieland, 7602, Cape Town South Africa
| | - Lihle Qulu-Appiah
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Private Bag X1, Matieland, 7602, Cape Town South Africa
| |
Collapse
|
5
|
de Kloet ER, Joëls M. The cortisol switch between vulnerability and resilience. Mol Psychiatry 2024; 29:20-34. [PMID: 36599967 DOI: 10.1038/s41380-022-01934-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023]
Abstract
In concert with neuropeptides and transmitters, the end products of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone (CORT), promote resilience: i.e., the ability to cope with threats, adversity, and trauma. To exert this protective action, CORT activates mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) that operate in a complementary manner -as an on/off switch- to coordinate circadian events, stress-coping, and adaptation. The evolutionary older limbic MR facilitates contextual memory retrieval and supports an on-switch in the selection of stress-coping styles at a low cost. The rise in circulating CORT concentration after stress subsequently activates a GR-mediated off-switch underlying recovery of homeostasis by providing the energy for restraining the primary stress reactions and promoting cognitive control over emotional reactivity. GR activation facilitates contextual memory storage of the experience to enable future stress-coping. Such complementary MR-GR-mediated actions involve rapid non-genomic and slower gene-mediated mechanisms; they are time-dependent, conditional, and sexually dimorphic, and depend on genetic background and prior experience. If coping fails, GR activation impairs cognitive control and promotes emotional arousal which eventually may compromise resilience. Such breakdown of resilience involves a transition to a chronic stress construct, where information processing is crashed; it leads to an imbalanced MR-GR switch and hence increased vulnerability. Novel MR-GR modulators are becoming available that may reset a dysregulated stress response system to reinstate the cognitive flexibility required for resilience.
Collapse
Affiliation(s)
- E Ronald de Kloet
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, The Netherlands.
- Leiden/Amsterdam Center of Drug Research, Leiden University, Leiden, The Netherlands.
| | - Marian Joëls
- Dept. Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Jeanneteau F. Fast signaling by glucocorticoids shapes neural representations of behaviors. Steroids 2023; 199:109294. [PMID: 37549777 DOI: 10.1016/j.steroids.2023.109294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Glucocorticoids are stress hormones that play central roles in the immediate and slower adaptive responses of the brain and body to new behavioral experience. The exact mechanisms by which the rapid and slow processes underlying glucocorticoid mnemonic effects unfold are under intensive scrutiny. It is possible that glucocorticoids rapidly modify memory representations in the brain by interfering with synaptic functions between inhibitory and excitatory neurons in a timing and context dependent manner. In particular, activity-dependent trans-synaptic messengers appear to have all the necessary attributes to engage in the rapid signaling by glucocorticoids and regulate the brain and behaviors. Novel frameworks for the treatment of stress-related disorders could emerge from a better characterization of the dynamic interplay between the rapid and slow signaling components by glucocorticoids on large-scale brain networks. Here I present some of the exact factors that could help reach this objective.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle , Université de Montpellier, INSERM, CNRS, 141 rue de la Cardonille, 34090, Montpellier, France.
| |
Collapse
|
7
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
8
|
Fricke K, Alexander N, Jacobsen T, Krug H, Wehkamp K, Vogel S. The effects of hydrocortisone and yohimbine on human behavior in approach-avoidance conflicts. Psychopharmacology (Berl) 2023; 240:1705-1717. [PMID: 37314480 PMCID: PMC10265560 DOI: 10.1007/s00213-023-06396-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 05/31/2023] [Indexed: 06/15/2023]
Abstract
RATIONALE Balancing approach of positive and avoidance of negative stimuli is essential when faced with approach-avoidance conflicts, e.g., situations with both positive and negative outcomes. This balance is disturbed in several mental disorders, e.g., excessive avoidance in anxiety disorders, and heightened approach in substance use disorders. Since stress is assumed to impact these disorders' etiology and maintenance, it seems crucial to understand how stress influences behavior in approach-avoidance conflicts. Indeed, some studies suggested altered approach-avoidance behavior under acute stress, but the mechanism underlying these effects is unknown. OBJECTIVES Investigate how the pharmacological manipulation of major stress mediators (cortisol and noradrenaline) influences task-based approach-avoidance conflict behavior in healthy individuals. METHODS Ninety-six participants (48 women, 48 men) received either 20mg hydrocortisone, 20mg yohimbine, both, or placebo before performing a task targeting foraging under predation in a fully crossed double-blind between-subject design. Moreover, we investigated effects of gender and endogenous testosterone and estradiol levels on approach-avoidance behavior. RESULTS While biological stress markers (cortisol concentration, alpha amylase activity) indicated successful pharmacological manipulation, behavior in approach-avoidance conflicts was not affected as expected. Although yohimbine administration affected risky foraging latency under predation, we found no main effect of hydrocortisone or their interaction on behavior. In contrast, we found gender differences for almost all behavioral outcome measures, which might be explained by differences in endogenous testosterone levels. CONCLUSIONS The investigated major stress mediators were not sufficient to imitate previously shown stress effects on approach-avoidance conflict behavior. We discuss potential reasons for our findings and implications for future research.
Collapse
Affiliation(s)
- Kim Fricke
- Department of Psychology, Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany.
- ICAN Institute for Cognitive and Affective Neuroscience, Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany.
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, Rudolf-Bultmann-Str. 8, 35039, Marburg, Germany
| | - Thomas Jacobsen
- Experimental Psychology Unit, Helmut-Schmidt-University/University of the Federal Armed Forces Hamburg, Holstenhofweg 85, 22043, Hamburg, Germany
| | - Henriette Krug
- Faculty of Health Sciences, Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany
| | - Kai Wehkamp
- Faculty of Health Sciences, Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany
| | - Susanne Vogel
- Department of Psychology, Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany
- ICAN Institute for Cognitive and Affective Neuroscience, Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany
| |
Collapse
|
9
|
Skupio U, Welte J, Serrat R, Eraso-Pichot A, Julio-Kalajzić F, Gisquet D, Cannich A, Delcasso S, Matias I, Fundazuri UB, Pouvreau S, Pagano Zottola AC, Lavanco G, Drago F, Ruiz de Azua I, Lutz B, Bellocchio L, Busquets-Garcia A, Chaouloff F, Marsicano G. Mitochondrial cannabinoid receptors gate corticosterone impact on novel object recognition. Neuron 2023; 111:1887-1897.e6. [PMID: 37098353 DOI: 10.1016/j.neuron.2023.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/21/2023] [Accepted: 03/30/2023] [Indexed: 04/27/2023]
Abstract
Corticosteroid-mediated stress responses require the activation of complex brain circuits involving mitochondrial activity, but the underlying cellular and molecular mechanisms are scantly known. The endocannabinoid system is implicated in stress coping, and it can directly regulate brain mitochondrial functions via type 1 cannabinoid (CB1) receptors associated with mitochondrial membranes (mtCB1). In this study, we show that the impairing effect of corticosterone in the novel object recognition (NOR) task in mice requires mtCB1 receptors and the regulation of mitochondrial calcium levels in neurons. Different brain circuits are modulated by this mechanism to mediate the impact of corticosterone during specific phases of the task. Thus, whereas corticosterone recruits mtCB1 receptors in noradrenergic neurons to impair NOR consolidation, mtCB1 receptors in local hippocampal GABAergic interneurons are required to inhibit NOR retrieval. These data reveal unforeseen mechanisms mediating the effects of corticosteroids during different phases of NOR, involving mitochondrial calcium alterations in different brain circuits.
Collapse
Affiliation(s)
- Urszula Skupio
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Julia Welte
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Roman Serrat
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Abel Eraso-Pichot
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Francisca Julio-Kalajzić
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Doriane Gisquet
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Astrid Cannich
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | | | - Isabelle Matias
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Unai B Fundazuri
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Sandrine Pouvreau
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Antonio C Pagano Zottola
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France; Institute for Cellular Biochemistry and Genetics, UMR 5095, Bordeaux 33077, France
| | - Gianluca Lavanco
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Filippo Drago
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
| | - Inigo Ruiz de Azua
- Institute of Physiological Chemistry, University Medical Center, Mainz 55131 Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center, Mainz 55131 Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | - Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Arnau Busquets-Garcia
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Program, IMIM Hospital del Mar Medical Research Institute, Barcelona 08003, Spain
| | - Francis Chaouloff
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
10
|
Lin X, Huang L, Huang H, Ke Z, Chen Y. Disturbed relationship between glucocorticoid receptor and 5-HT1AR/5-HT2AR in ADHD rats: A correlation study. Front Neurosci 2023; 16:1064369. [PMID: 36699537 PMCID: PMC9869156 DOI: 10.3389/fnins.2022.1064369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Objective This work is to investigate the alterations of the central 5-hydroxytryptamine (5-HT) system in spontaneously hypertensive rats (SHR) and the correlation with the behaviors of SHR, and to explore the effects of glucocorticoid intervention on the central 5-HT system and SHR behaviors. Materials and methods Three weeks old SHR were chosen as the attention-deficit hyperactivity disorder (ADHD) model and treated with glucocorticoid receptor (GR) agonist or inhibitor, whereas Wista Kyoto rats (WKY) were chosen as the normal control group. Open-field test and Làt maze test were used to evaluate the spontaneous activities and non-selective attention. The levels of 5-HT in the extracellular fluid specimens of the prefrontal cortex of rats were analyzed by high-performance liquid chromatography. The expressions of GR, 5-HT1A receptor (5-HT1AR), and 5-HT2A receptor (5-HT2AR) in the prefrontal cortex were analyzed through immunohistochemistry. Results Our study demonstrated that the 5-HT level was lower in the prefrontal cortex of SHR compared to that of WKY. The Open-field test and Làt maze test showed that GR agonist (dexamethasone, DEX) intervention ameliorated attention deficit and hyperactive behavior, whereas GR inhibitor (RU486) aggravated the disorders. With DEX, the expression levels of 5-HT and 5-HT2AR in the prefrontal cortex of SHR were significantly higher than those in the control group, whereas the expression level of 5-HT1AR was lower. However, the expression levels of 5-HT and 5-HT2AR were significantly decreased after the intervention with RU486, while the expression level of 5-HT1AR increased. Results showed that glucocorticoid was negatively correlated with 5-HT1AR and positively correlated with 5-HT2AR. Conclusion In the prefrontal cortex of ADHD rats, the down-regulation of 5-HT and 5-HT2AR expressions and the up-regulation of 5-HT1AR, compared with WYK rats, suggested a dysfunctional central 5-HT system in ADHD rats. The GR agonist can upregulate the expression of 5-HT and 5-HT2AR and downregulate the expression of 5-HT1AR in the prefrontal cortex of SHR as well as reduce the hyperactivity and attention deficit behavior in SHR, while the opposite was true for the GR inhibitor. It is suggested that the dysfunction of the 5-HT system in ADHD rats is closely related to glucocorticoid receptor activity.
Collapse
|
11
|
Hansen N, Neyazi A, Lüdecke D, Hasan A, Wiltfang J, Malchow B. Repositioning synthetic glucocorticoids in psychiatric disease associated with neural autoantibodies: a narrative review. J Neural Transm (Vienna) 2022:10.1007/s00702-022-02578-2. [PMID: 36576564 PMCID: PMC10374711 DOI: 10.1007/s00702-022-02578-2] [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: 09/23/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
Synthetic glucocorticoids (sGCs) are a well-investigated and standard drug therapy for disorders associated with CNS inflammation. Less is known about treating psychiatric disorders associated with neural autoantibodies. Our aim is to elucidate the repositioning of sGCs in psychiatric diseases that co-exist with neural autoantibodies. We used PubMed to identify articles for this narrative review. To our knowledge, no randomized, placebo-controlled trials have yet been conducted on applying sGC to treat neural autoantibody-associated psychiatric disorders. We describe initial results of cohort studies and single cases or case series often associated with autoantibodies against membrane-surface antigens demonstrating a largely beneficial response to sGCs either as monotherapy or polytherapy together with other immunosuppressive agents. However, sGCs may be less efficient in patients with psychiatric diseases associated with autoantibodies directed against intracellular antigens. These results reveal potential benefits of the novel usage of sGCs for the indication of neural autoantibody-associated psychiatric disease. Further large-scale randomized, placebo-controlled trials are needed to discover whether sGCs are safe, well tolerated, and beneficial in subgroups of neural autoantibody-associated psychiatric diseases.
Collapse
Affiliation(s)
- Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany.
| | - Alexandra Neyazi
- Department of Psychiatry and Psychotherapy, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Daniel Lüdecke
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, 86156, Augsburg, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Goettingen, Germany.,Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | | |
Collapse
|
12
|
Chao OY, Nikolaus S, Yang YM, Huston JP. Neuronal circuitry for recognition memory of object and place in rodent models. Neurosci Biobehav Rev 2022; 141:104855. [PMID: 36089106 PMCID: PMC10542956 DOI: 10.1016/j.neubiorev.2022.104855] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022]
Abstract
Rats and mice are used for studying neuronal circuits underlying recognition memory due to their ability to spontaneously remember the occurrence of an object, its place and an association of the object and place in a particular environment. A joint employment of lesions, pharmacological interventions, optogenetics and chemogenetics is constantly expanding our knowledge of the neural basis for recognition memory of object, place, and their association. In this review, we summarize current studies on recognition memory in rodents with a focus on the novel object preference, novel location preference and object-in-place paradigms. The evidence suggests that the medial prefrontal cortex- and hippocampus-connected circuits contribute to recognition memory for object and place. Under certain conditions, the striatum, medial septum, amygdala, locus coeruleus and cerebellum are also involved. We propose that the neuronal circuitry for recognition memory of object and place is hierarchically connected and constructed by different cortical (perirhinal, entorhinal and retrosplenial cortices), thalamic (nucleus reuniens, mediodorsal and anterior thalamic nuclei) and primeval (hypothalamus and interpeduncular nucleus) modules interacting with the medial prefrontal cortex and hippocampus.
Collapse
Affiliation(s)
- Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Susanne Nikolaus
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany.
| |
Collapse
|
13
|
Jeanneteau F, Coutellier L. The glucocorticoid footprint on the memory engram. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25:100378. [PMID: 38486965 PMCID: PMC10938917 DOI: 10.1016/j.coemr.2022.100378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
The complexity of the classical inverted U-shaped relationship between cortisol levels and responses transposable to stress reactivity has led to an incomplete understanding of the mechanisms enabling healthy and toxic effects of stress on brain and behavior. A clearer, more detailed, picture of those relationships can be obtained by integrating cortisol effects on large-scale brain networks, in particular, by focusing on neural network configurations from the perspective of inhibition and excitation. A unifying view of Semon and Hebb's theories of cellular memory links the biophysical and metabolic changes in neuronal ensembles to the strengthening of collective synapses. In that sense, the neuronal capacity to record, store, and retrieve information directly relates to the adaptive capacity of its connectivity and metabolic reserves. Here, we use task-activated cell ensembles or simply engram cells as an example to demonstrate that the adaptive behavioral responses to stress result from collective synapse strength within and across networks of interneurons and excitatory ones.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Laurence Coutellier
- Departments of Psychology and Neuroscience, Ohio State University, Columbus, USA
| |
Collapse
|
14
|
Osorio-Gómez D, Guzmán-Ramos K, Bermúdez-Rattoni F. Dopamine activity on the perceptual salience for recognition memory. Front Behav Neurosci 2022; 16:963739. [PMID: 36275849 PMCID: PMC9583835 DOI: 10.3389/fnbeh.2022.963739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
To survive, animals must recognize relevant stimuli and distinguish them from inconspicuous information. Usually, the properties of the stimuli, such as intensity, duration, frequency, and novelty, among others, determine the salience of the stimulus. However, previously learned experiences also facilitate the perception and processing of information to establish their salience. Here, we propose “perceptual salience” to define how memory mediates the integration of inconspicuous stimuli into a relevant memory trace without apparently altering the recognition of the physical attributes or valence, enabling the detection of stimuli changes in future encounters. The sense of familiarity is essential for successful recognition memory; in general, familiarization allows the transition of labeling a stimulus from the novel (salient) to the familiar (non-salient). The novel object recognition (NOR) and object location recognition (OLRM) memory paradigms represent experimental models of recognition memory that allow us to study the neurobiological mechanisms involved in episodic memory. The catecholaminergic system has been of vital interest due to its role in several aspects of recognition memory. This review will discuss the evidence that indicates changes in dopaminergic activity during exposure to novel objects or places, promoting the consolidation and persistence of memory. We will discuss the relationship between dopaminergic activity and perceptual salience of stimuli enabling learning and consolidation processes necessary for the novel-familiar transition. Finally, we will describe the effect of dopaminergic deregulation observed in some pathologies and its impact on recognition memory.
Collapse
Affiliation(s)
- Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Mexico, Mexico
| | - Kioko Guzmán-Ramos
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Lerma, Estado de México, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Mexico, Mexico
- *Correspondence: Federico Bermúdez-Rattoni
| |
Collapse
|
15
|
Chen YF, Song Q, Colucci P, Maltese F, Siller-Pérez C, Prins K, McGaugh JL, Hermans EJ, Campolongo P, Kasri NN, Roozendaal B. Basolateral amygdala activation enhances object recognition memory by inhibiting anterior insular cortex activity. Proc Natl Acad Sci U S A 2022; 119:e2203680119. [PMID: 35622887 PMCID: PMC9295787 DOI: 10.1073/pnas.2203680119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Noradrenergic activation of the basolateral amygdala (BLA) by emotional arousal enhances different forms of recognition memory via functional interactions with the insular cortex (IC). Human neuroimaging studies have revealed that the anterior IC (aIC), as part of the salience network, is dynamically regulated during arousing situations. Emotional stimulation first rapidly increases aIC activity but suppresses it in a delayed fashion. Here, we investigated in male Sprague-Dawley rats whether the BLA influence on recognition memory is associated with an increase or suppression of aIC activity during the postlearning consolidation period. We first employed anterograde and retrograde viral tracing and found that the BLA sends dense monosynaptic projections to the aIC. Memory-enhancing norepinephrine administration into the BLA following an object training experience suppressed aIC activity 1 h later, as determined by a reduced expression of the phosphorylated form of the transcription factor cAMP response element-binding (pCREB) protein and neuronal activity marker c-Fos. In contrast, the number of perisomatic γ-aminobutyric acid (GABA)ergic inhibitory synapses per pCREB-positive neuron was significantly increased, suggesting a dynamic up-regulation of GABAergic tone. In support of this possibility, pharmacological inhibition of aIC activity with a GABAergic agonist during consolidation enhanced object recognition memory. Norepinephrine administration into the BLA did not affect neuronal activity within the posterior IC, which receives sparse innervation from the BLA. The evidence that noradrenergic activation of the BLA enhances the consolidation of object recognition memory via a mechanism involving a suppression of aIC activity provides insight into the broader brain network dynamics underlying emotional regulation of memory.
Collapse
Affiliation(s)
- Yan-Fen Chen
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| | - Qi Song
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| | - Paola Colucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico, Santa Lucia Foundation, 00179 Rome, Italy
| | - Federica Maltese
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| | | | - Karina Prins
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| | - James L. McGaugh
- Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-3800
| | - Erno J. Hermans
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico, Santa Lucia Foundation, 00179 Rome, Italy
| | - Nael Nadif Kasri
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| |
Collapse
|
16
|
dos Santos Corrêa M, Grisanti GDV, Franciscatto IAF, Tarumoto TSA, Tiba PA, Ferreira TL, Fornari RV. Remote contextual fear retrieval engages activity from salience network regions in rats. Neurobiol Stress 2022; 18:100459. [PMID: 35601686 PMCID: PMC9118522 DOI: 10.1016/j.ynstr.2022.100459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/22/2022] [Accepted: 05/04/2022] [Indexed: 11/30/2022] Open
Abstract
The ability to retrieve contextual fear memories depends on the coordinated activation of a brain-wide circuitry. Transition from recent to remote memories seems to involve the reorganization of this circuitry, a process called systems consolidation that has been associated with time-dependent fear generalization. However, it is unknown whether emotional memories acquired under different stress levels can undergo different systems consolidation processes. Here, we explored the activation pattern and functional connectivity of key brain regions associated with contextual fear conditioning (CFC) retrieval after recent (2 days) or remote (28 days) memory tests performed in rats submitted to strong (1.0 mA footshock) or mild (0.3 mA footshock) training. We used brain tissue from Wistar rats from a previous study, where we observed that increasing training intensity promotes fear memory generalization over time, possibly due to an increase in corticosterone (CORT) levels during memory consolidation. Analysis of Fos expression across 8 regions of interest (ROIs) allowed us to identify coactivation between them at both timepoints following memory recall. Our results showed that strong CFC elicits higher Fos activation in the anterior insular and prelimbic cortices during remote retrieval, which was positively correlated with freezing along with the basolateral amygdala. Rats trained either with mild or strong CFC showed broad functional connectivity at the recent timepoint whereas only animals submitted to the strong CFC showed a widespread loss of coactivation during remote retrieval. Post-training plasma CORT levels are positively correlated with FOS expression during recent retrieval in strong CFC, but negatively correlated with FOS expression during remote retrieval in mild CFC. Our findings suggest that increasing training intensity results in differential processes of systems consolidation, possibly associated with increased post-training CORT release, and that strong CFC engages activity from the aIC, BLA and PrL - areas associated with the Salience Network in rats - during remote retrieval.
Collapse
Affiliation(s)
- Moisés dos Santos Corrêa
- Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | - Gabriel David Vieira Grisanti
- Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | | | - Tatiana Suemi Anglas Tarumoto
- Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | - Paula Ayako Tiba
- Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | - Tatiana Lima Ferreira
- Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | - Raquel Vecchio Fornari
- Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC (UFABC), São Bernardo do Campo, SP, Brazil
| |
Collapse
|
17
|
Schwabe L, Hermans EJ, Joëls M, Roozendaal B. Mechanisms of memory under stress. Neuron 2022; 110:1450-1467. [PMID: 35316661 DOI: 10.1016/j.neuron.2022.02.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/11/2022]
Abstract
It is well established that stress has a major impact on memory, driven by the concerted action of various stress mediators on the brain. Recent years, however, have seen considerable advances in our understanding of the cellular, neural network, and cognitive mechanisms through which stress alters memory. These novel insights highlight the intricate interplay of multiple stress mediators, including-beyond corticosteroids, catecholamines, and peptides-for instance, endocannabinoids, which results in time-dependent shifts in large-scale neural networks. Such stress-induced network shifts enable highly specific memories of the stressful experience in the long run at the cost of transient impairments in mnemonic flexibility during and shortly after a stressful event. Based on these recent discoveries, we provide a new integrative framework that links the cellular, systems, and cognitive mechanisms underlying acute stress effects on memory processes and points to potential targets for treating aberrant memory in stress-related mental disorders.
Collapse
Affiliation(s)
- Lars Schwabe
- Department of Cognitive Psychology, Universität Hamburg, Hamburg, Germany.
| | - Erno J Hermans
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marian Joëls
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Benno Roozendaal
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Role of Stress-Related Dopamine Transmission in Building and Maintaining a Protective Cognitive Reserve. Brain Sci 2022; 12:brainsci12020246. [PMID: 35204009 PMCID: PMC8869980 DOI: 10.3390/brainsci12020246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 01/27/2023] Open
Abstract
This short review presents the hypothesis that stress-dependent dopamine (DA) transmission contributes to developing and maintaining the brain network supporting a cognitive reserve. Research has shown that people with a greater cognitive reserve are better able to avoid symptoms of degenerative brain changes. The paper will review evidence that: (1) successful adaptation to stressors involves development and stabilization of effective but flexible coping strategies; (2) this process requires dynamic reorganization of functional networks in the adult brain; (3) DA transmission is amongst the principal mediators of this process; (4) age- and disease-dependent cognitive impairment is associated with dysfunctional connectivity both between and within these same networks as well as with reduced DA transmission.
Collapse
|
19
|
The Entorhinal Cortex as a Gateway for Amygdala Influences on Memory Consolidation. Neuroscience 2022; 497:86-96. [PMID: 35122874 DOI: 10.1016/j.neuroscience.2022.01.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/16/2022]
Abstract
The amygdala, specifically its basolateral nucleus (BLA), is a critical site integrating neuromodulatory influences on memory consolidation in other brain areas. Almost 20 years ago, we reported the first direct evidence that BLA activity is required for modulatory interventions in the entorhinal cortex (EC) to affect memory consolidation (Roesler, Roozendaal, and McGaugh, 2002). Since then, significant advances have been made in our understanding of how the EC participates in memory. For example, the characterization of grid cells specialized in processing spatial information in the medial EC (mEC) that act as major relayers of information to the hippocampus (HIP) has changed our view of memory processing by the EC; and the development of optogenetic technologies for manipulation of neuronal activity has recently enabled important new discoveries on the role of the BLA projections to the EC in memory. Here, we review the current evidence on interactions between the BLA and EC in synaptic plasticity and memory formation. The findings suggest that the EC may function as a gateway and mediator of modulatory influences from the BLA, which are then processed and relayed to the HIP. Through extensive reciprocal connections among the EC, HIP, and several cortical areas, information related to new memories is then consolidated by these multiple brain systems, through various molecular and cellular mechanisms acting in a distributed and highly concerted manner, during several hours after learning. A special note is made on the contribution by Ivan Izquierdo to our understanding of memory consolidation at the brain system level.
Collapse
|
20
|
Implicit and explicit emotional memory recall in anxiety and depression: Role of basolateral amygdala and cortisol-norepinephrine interaction. Psychoneuroendocrinology 2022; 136:105598. [PMID: 34894424 DOI: 10.1016/j.psyneuen.2021.105598] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/14/2021] [Accepted: 11/17/2021] [Indexed: 01/05/2023]
Abstract
Anxiety and depression are linked to both explicit and implicit memory biases, which are defined as the tendency to preferentially recall emotionally negative information at conscious and subconscious levels, respectively. Functional connectivity (FC) of the basolateral amygdala (BLA) and related stress hormones (i.e., cortisol and norepinephrine) are purportedly implicated in these biases. However, previous findings on memory biases in anxiety and depression have been inconsistent, likely due to their symptomatic complications. Therefore, the underlying neurobiological mechanism remains unclear. We thus investigated whether anxiety and depression as premorbid predispositions are related to the memory biases, and whether FC of BLA, cortisol, and 3-methoxy-4-hydroxyphenylglycol (MHPG: a major metabolite of norepinephrine) would affect the anxiety/depression-related biased memory recall in 100 participants without psychiatric symptomatology. Psycho-behavioral assessment, resting-state fMRI scans, and saliva collection at 10-points-in-time across two days were conducted. Correlations of memory biases with anxiety/depression and neurobiological markers were explored. As a result, neither anxiety nor depression were correlated with explicit memory bias to negative (vs. positive) information, although depression was associated with better recall of the negative stimuli only when they were perceived as self-relevant. In contrast, both anxiety and depression were correlated with implicit memory bias; however, the effects were solely explained by anxiety. Furthermore, FC of the BLA with subgenual anterior cingulate cortex (sgACC) and the synergetic effect of cortisol and MHPG uniquely affected the implicit memory bias. These findings suggest that anxiety facilitates an initial snapshot of negative information and can be accompanied by depression when the information creates negative semantic associations with the self. The BLA-sgACC neural connectivity and cortisol-norepinephrine interaction that are associated with the implicit memory bias might be one of the important neurobiological targets in the prevention and treatment for comorbid anxiety and depressive disorders.
Collapse
|
21
|
Faucher P, Huguet C, Mons N, Micheau J. Acute pre-learning stress selectively impairs hippocampus-dependent fear memory consolidation: Behavioral and molecular evidence. Neurobiol Learn Mem 2022; 188:107585. [PMID: 35021061 DOI: 10.1016/j.nlm.2022.107585] [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: 07/27/2021] [Revised: 12/22/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
Despite compelling evidence that stress or stress-related hormones influence fear memory consolidation processes, the understanding of molecular mechanisms underlying the effects of stress is still fragmentary. The release of corticosterone in response to pre-learning stress exposure has been demonstrated to modulate positively or negatively memory encoding and/or consolidation according to many variables such as stress intensity, the emotional valence of the learned material or the interval between stressful episode and learning experience. Here, we report that contextual but not cued fear memory consolidation was selectively impaired in male mice exposed to a 50 min-period of restraint stress just before the unpaired fear conditioning session. In addition to behavioral impairment, acute stress down-regulated activated/phosphorylated ERK1/2 (pERK1/2) in dorsal hippocampal area CA1 in mice sacrificed 60 min and 9 h after unpaired conditioning. In lateral amygdala, although acute stress by itself increased the level of pERK1/2 it nevertheless blocked the peak of pERK1/2 that was normally observed 15 min after unpaired conditioning. To examine whether stress-induced corticosterone overflow was responsible of these detrimental effects, the corticosterone synthesis inhibitor, metyrapone, was administered 30 min before stress exposure. Metyrapone abrogated the stress-induced contextual fear memory deficits but did not alleviate the effects of stress on pERK1/2 and its downstream target phosphorylated CREB (pCREB) in hippocampus CA1 and lateral amygdala. Collectively, our observations suggest that consolidation of hippocampus-dependent memory and the associated signaling pathway are particularly sensitive to stress. However, behavioral normalization by preventive metyrapone treatment was not accompanied by renormalization of the canonical signaling pathway. A new avenue would be to consider surrogate mechanisms involving proper metyrapone influence on both nongenomic and genomic actions of glucocorticoid receptors.
Collapse
Affiliation(s)
- Pierre Faucher
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Célia Huguet
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Nicole Mons
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Jacques Micheau
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France.
| |
Collapse
|
22
|
Ventura-Bort C, Wirkner J, Wendt J, Hamm AO, Weymar M. Establishment of Emotional Memories Is Mediated by Vagal Nerve Activation: Evidence from Noninvasive taVNS. J Neurosci 2021; 41:7636-7648. [PMID: 34281991 PMCID: PMC8425981 DOI: 10.1523/jneurosci.2329-20.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 11/21/2022] Open
Abstract
Emotional memories are better remembered than neutral ones, but the mechanisms leading to this memory bias are not well understood in humans yet. Based on animal research, it is suggested that the memory-enhancing effect of emotion is based on central noradrenergic release, which is triggered by afferent vagal nerve activation. To test the causal link between vagus nerve activation and emotional memory in humans, we applied continuous noninvasive transcutaneous auricular vagus nerve stimulation (taVNS) during exposure to emotional arousing and neutral scenes and tested subsequent, long-term recognition memory after 1 week. We found that taVNS, compared with sham, increased recollection-based memory performance for emotional, but not neutral, material. These findings were complemented by larger recollection-related brain potentials (parietal ERP Old/New effect) during retrieval of emotional scenes encoded under taVNS, compared with sham. Furthermore, brain potentials recorded during encoding also revealed that taVNS facilitated early attentional discrimination between emotional and neutral scenes. Extending animal research, our behavioral and neural findings confirm a modulatory influence of the vagus nerve in emotional memory formation in humans.SIGNIFICANCE STATEMENT Emotionally relevant information elicits stronger and more enduring memories than nonrelevant information. Animal research has shown that this memory-enhancing effect of emotion is related to the noradrenergic activation in the brain, which is triggered by afferent fibers of the vagus nerve (VN). In the current study, we show that noninvasive transcutaneous auricular VN stimulation enhances recollection-based memory formation specifically for emotionally relevant information as indicated by behavioral and electrophysiological indices. These human findings give novel insights into the mechanisms underlying the establishment of emotional episodic memories by confirming the causal link between the VN and memory formation which may help understand the neural mechanisms underlying disorders associated with altered memory functions and develop treatment options.
Collapse
Affiliation(s)
- Carlos Ventura-Bort
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, 14476, Potsdam, Germany
| | - Janine Wirkner
- Department of Clinical Psychology and Psychotherapy, University of Greifswald, Greifswald, 17489, Germany
| | - Julia Wendt
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, 14476, Potsdam, Germany
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, 17487, Germany
| | - Alfons O Hamm
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, 17487, Germany
| | - Mathias Weymar
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, 14476, Potsdam, Germany
- Faculty of Health Sciences Brandenburg, University of Potsdam, Potsdam, 14476, Germany
| |
Collapse
|
23
|
Song Q, Bolsius YG, Ronzoni G, Henckens MJAG, Roozendaal B. Noradrenergic enhancement of object recognition and object location memory in mice. Stress 2021; 24:181-188. [PMID: 32233890 DOI: 10.1080/10253890.2020.1747427] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Extensive evidence indicates that noradrenergic activation is essentially involved in mediating the enhancing effects of emotional arousal on memory consolidation. Our current understanding of the neurobiological mechanisms underlying the memory-modulatory effects of the noradrenergic system is primarily based on pharmacological studies in rats, employing targeted administration of noradrenergic drugs into specific brain regions. However, the further delineation of the specific neural circuitry involved would benefit from experimental tools that are currently more readily available in mice. Previous studies have not, as yet, investigated the effect of noradrenergic enhancement of memory in mice, which show different cognitive abilities and higher endogenous arousal levels induced by a training experience compared to rats. In the present study, we investigated the effect of posttraining noradrenergic activation in male C57BL/6J mice on the consolidation of object recognition and object location memory. We found that the noradrenergic stimulant yohimbine (0.3 or 1.0 mg/kg) administered systemically immediately after an object training experience dose-dependently enhanced 24-h memory of both the identity and location of the object. Thus, these findings indicate that noradrenergic activation also enhances memory consolidation processes in mice, paving the way for a systematic investigation of the neural circuitry underlying these emotional arousal effects on memory.LAY SUMMARY: The current study successfully validated the effect of noradrenergic activation on both object recognition and object location memory in mice. This study thereby provides a fundamental proof-of-principle for the investigation of the neural circuitry underlying noradrenergic and arousal effects on long-term memory in mice.
Collapse
Affiliation(s)
- Qi Song
- Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| | - Youri G Bolsius
- Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Giacomo Ronzoni
- Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| | - Marloes J A G Henckens
- Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Espinoza S, Arredondo SB, Barake F, Carvajal F, Guerrero FG, Segovia-Miranda F, Valenzuela DM, Wyneken U, Rojas-Fernández A, Cerpa W, Massardo L, Varela-Nallar L, González A. Neuronal surface P antigen (NSPA) modulates postsynaptic NMDAR stability through ubiquitination of tyrosine phosphatase PTPMEG. BMC Biol 2020; 18:164. [PMID: 33158444 PMCID: PMC7648380 DOI: 10.1186/s12915-020-00877-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cognitive dysfunction (CD) is common among patients with the autoimmune disease systemic lupus erythematosus (SLE). Anti-ribosomal P autoantibodies associate with this dysfunction and have neuropathogenic effects that are mediated by cross-reacting with neuronal surface P antigen (NSPA) protein. Elucidating the function of NSPA can then reveal CD pathogenic mechanisms and treatment opportunities. In the brain, NSPA somehow contributes to glutamatergic NMDA receptor (NMDAR) activity in synaptic plasticity and memory. Here we analyze the consequences of NSPA absence in KO mice considering its structural features shared with E3 ubiquitin ligases and the crucial role of ubiquitination in synaptic plasticity. Results Electrophysiological studies revealed a decreased long-term potentiation in CA3-CA1 and medial perforant pathway-dentate gyrus (MPP-DG) hippocampal circuits, reflecting glutamatergic synaptic plasticity impairment in NSPA-KO mice. The hippocampal dentate gyrus of these mice showed a lower number of Arc-positive cells indicative of decreased synaptic activity and also showed proliferation defects of neural progenitors underlying less adult neurogenesis. All this translates into poor spatial and recognition memory when NSPA is absent. A cell-based assay demonstrated ubiquitination of NSPA as a property of RBR-type E3 ligases, while biochemical analysis of synaptic regions disclosed the tyrosine phosphatase PTPMEG as a potential substrate. Mice lacking NSPA have increased levels of PTPMEG due to its reduced ubiquitination and proteasomal degradation, which correlated with lower levels of GluN2A and GluN2B NMDAR subunits only at postsynaptic densities (PSDs), indicating selective trafficking of these proteins out of PSDs. As both GluN2A and GluN2B interact with PTPMEG, tyrosine (Tyr) dephosphorylation likely drives their endocytic removal from the PSD. Actually, immunoblot analysis showed reduced phosphorylation of the GluN2B endocytic signal Tyr1472 in NSPA-KO mice. Conclusions NSPA contributes to hippocampal plasticity and memory processes ensuring appropriate levels of adult neurogenesis and PSD-located NMDAR. PTPMEG qualifies as NSPA ubiquitination substrate that regulates Tyr phosphorylation-dependent NMDAR stability at PSDs. The NSPA/PTPMEG pathway emerges as a new regulator of glutamatergic transmission and plasticity and may provide mechanistic clues and therapeutic opportunities for anti-P-mediated pathogenicity in SLE, a still unmet need.
Collapse
Affiliation(s)
- Sofía Espinoza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, 7510157, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile
| | - Sebastián B Arredondo
- Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Francisca Barake
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, 7510157, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile.,Fundación Ciencia y Vida, 7780272, Santiago, Chile
| | - Francisco Carvajal
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330028, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), 6213029, Punta Arenas, Chile
| | - Fernanda G Guerrero
- Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Fabian Segovia-Miranda
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile
| | | | - Ursula Wyneken
- Laboratorio de Neurociencias, Facultad de Medicina, Universidad de los Andes, 7620001, Santiago, Chile
| | - Alejandro Rojas-Fernández
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - Waldo Cerpa
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile.,Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330028, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), 6213029, Punta Arenas, Chile
| | - Loreto Massardo
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, 7510157, Santiago, Chile
| | - Lorena Varela-Nallar
- Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, 7510157, Santiago, Chile. .,Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile. .,Fundación Ciencia y Vida, 7780272, Santiago, Chile.
| |
Collapse
|
25
|
Norepinephrine and glucocorticoid effects on the brain mechanisms underlying memory accuracy and generalization. Mol Cell Neurosci 2020; 108:103537. [DOI: 10.1016/j.mcn.2020.103537] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/31/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
|
26
|
Li Z, Gao C, Peng J, Liu M, Cong B. Multi-omics analysis of pathological changes in the amygdala of rats subjected to chronic restraint stress. Behav Brain Res 2020; 392:112735. [PMID: 32502515 DOI: 10.1016/j.bbr.2020.112735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Overwhelming stress potentially results in the occurrence of many mental diseases. The amygdala is one region in the brain targeted by stress. Recent studies have shown that changes in the amygdala of subjects under stress are related to depression, anxiety and post-traumatic stress disorder (PTSD). However, researchers have not clearly elucidated the changes in the amygdala in response to stress and the underlying mechanism. We conducted several experiments to understand this mechanism. METHODS In this study, we first established a rat model of chronic restraint stress (CRS) and observed the changes in behavior and neurons in the amygdala. Second, an integrated metabolomics and proteomics experiment was conducted to identify potential stress-related biomarkers. Finally, we validated two molecules of interest and detected four apoptosis-related proteins using Western blotting to further determine the related mechanisms. RESULTS Our study revealed the presence of anxiety-like behaviors and pathological changes in amygdalar neurons in the rat model. In the multi-omics analysis, 19 potential molecules were identified. Western blotting confirmed consistent changes in the levels of Cry1 and Brcc36 obtained in previous results. The levels of proteins in the ataxia telangiectasia mutated (ATM) pathway were increased in the CRS group. CONCLUSIONS CRS causes anxiety-like behaviors that are potentially related to decreased levels of GABA in the amygdala. Moreover, CRS potentially alters the levels of Cry1 and Brcc36 and results in circadian rhythm disorder and impairments in DNA repair and apoptosis in the amygdala through a mechanism mediated by the ATM pathway.
Collapse
Affiliation(s)
- Zhonghua Li
- West China School of Basic Medical Sciences & Forensic Science, Sichuan University, Chengdu, 610041 China; Procuratorial Technology Department of the People's Procuratorate of Sichuan Province, Chengdu, 610041 China
| | - Chong Gao
- Procuratorial Technology Information Center of the Supreme People's Procuratorate, Beijing, 100726 China
| | - Jin Peng
- West China School of Basic Medical Sciences & Forensic Science, Sichuan University, Chengdu, 610041 China
| | - Min Liu
- West China School of Basic Medical Sciences & Forensic Science, Sichuan University, Chengdu, 610041 China
| | - Bin Cong
- West China School of Basic Medical Sciences & Forensic Science, Sichuan University, Chengdu, 610041 China; Department of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017 China.
| |
Collapse
|
27
|
Roozendaal B, Mirone G. Opposite effects of noradrenergic and glucocorticoid activation on accuracy of an episodic-like memory. Psychoneuroendocrinology 2020; 114:104588. [PMID: 32085987 DOI: 10.1016/j.psyneuen.2020.104588] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/05/2019] [Accepted: 01/20/2020] [Indexed: 10/25/2022]
Abstract
Stressful and emotionally arousing experiences activate hormonal systems that create strong memories. It remains unclear, however, how this strengthening affects the quality of such memories. In the present study, we examined whether the noradrenergic and glucocorticoid hormonal systems affect accuracy of episodic-like memory. We trained male Sprague-Dawley rats on an episodic-like association task, termed inhibitory avoidance discrimination task, in which they explored two different contexts, but shock was given only in the latter context. Forty-eight hours later, retention latencies were tested in the two training contexts as well as in a novel context. The noradrenergic stimulant yohimbine, administered systemically immediately after the training session, enhanced both accuracy and strength of the memory, as shown by long latencies specific to the shock context. By contrast, the glucocorticoid corticosterone induced a generalized strengthening of memory and enhanced latencies in both the shock and non-shock training contexts. Retention latencies in the novel context were not significantly affected. These findings indicate that the noradrenergic and glucocorticoid systems, while both strengthening memory of the shock experience per se, produce opposite effects on accuracy of the shock-context association.
Collapse
Affiliation(s)
- Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands.
| | - Gabriele Mirone
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| |
Collapse
|
28
|
Hakamata Y, Mizukami S, Izawa S, Moriguchi Y, Hori H, Kim Y, Hanakawa T, Inoue Y, Tagaya H. Basolateral Amygdala Connectivity With Subgenual Anterior Cingulate Cortex Represents Enhanced Fear-Related Memory Encoding in Anxious Humans. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 5:301-310. [PMID: 32001192 DOI: 10.1016/j.bpsc.2019.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/31/2019] [Accepted: 11/15/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND The amygdala can enhance emotional memory encoding as well as anxiogenesis via corticotropin-releasing factor neurons. However, the amygdala's explicit role in emotional encoding remains unclarified in humans. We examined how functional connectivity (FC) of amygdala subnuclei affects emotional encoding, considering its mechanism in which anxiety, attention, and cortisol conceivably participate. METHODS A total of 65 healthy humans underwent resting-state functional magnetic resonance imaging scans and saliva collection at 10 points in time over 2 days. FC analysis was performed for basolateral amygdala subnucleus (BLA) and centromedial amygdala subnucleus. We assessed attentional control via an emotional Stroop task and assessed emotional encoding via a facial identification task that examines how strongly a neutral face is memorized when accompanied by an emotional face (fearful, sad, or happy). FC and task performance were compared between high-anxious and non-high-anxious groups classified by anxious personality scores. RESULTS BLA connected with subgenual anterior cingulate cortex (sgACC) in proportion to the strength of fear-related encoding, whereas centromedial subnucleus connected with caudate nucleus for happy-related encoding. The high-anxious group showed more enhanced fear-related encoding but impaired happy-related encoding compared with the non-high-anxious group. BLA-sgACC FC was more intensified in the high-anxious group than in the non-high-anxious group; however, centromedial-caudate FC did not differ between them. Although emotional encoding was uncorrelated with either attentional control or cortisol, BLA-sgACC was positively correlated with cortisol increase after awakening. CONCLUSIONS The study revealed that neural interactions of BLA, specifically with sgACC, might play a critical role in fear-related memory encoding, depending on the individual's level of anxiety. These findings aid in understanding the complicated mechanisms of emotional memory in anxiety disorders.
Collapse
Affiliation(s)
- Yuko Hakamata
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Health Science, Kitasato University School of Allied Health Sciences, Kanagawa, Japan.
| | - Shinya Mizukami
- Department of Radiological Technology, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Shuhei Izawa
- Occupational Stress Research Group, National Institute of Occupational Safety and Health, Kanagawa, Japan
| | - Yoshiya Moriguchi
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroaki Hori
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshiharu Kim
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Hanakawa
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yusuke Inoue
- Department of Diagnostic Radiology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hirokuni Tagaya
- Department of Health Science, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| |
Collapse
|
29
|
Santori A, Colucci P, Mancini GF, Morena M, Palmery M, Trezza V, Puglisi-Allegra S, Hill MN, Campolongo P. Anandamide modulation of circadian- and stress-dependent effects on rat short-term memory. Psychoneuroendocrinology 2019; 108:155-162. [PMID: 31302498 DOI: 10.1016/j.psyneuen.2019.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 05/22/2019] [Accepted: 06/28/2019] [Indexed: 10/26/2022]
Abstract
The endocannabinoid system plays a key role in the control of emotional responses to environmental challenges. CB1 receptors are highly expressed within cortico-limbic brain areas, where they modulate stress effects on memory processes. Glucocorticoid and endocannabinoid release is influenced by circadian rhythm. Here, we investigated how different stress intensities immediately after encoding influence rat short-term memory in an object recognition task, whether the effects depend on circadian rhythm and if exogenous augmentation of anandamide levels could restore any observed impairment. Two separate cohorts of male adult Sprague-Dawley rats were tested at two different times of the day, morning (inactivity phase) or afternoon (before the onset of the activity phase) in an object recognition task. The anandamide hydrolysis inhibitor URB597 was intraperitoneally administered immediately after the training trial. Rats were thereafter subjected to a forced swim stress under low or high stress conditions and tested 1 h after training. Control rats underwent the same experimental procedure except for the forced swim stress (no stress). We further investigated whether URB597 administration might modulate corticosterone release in rats subjected to the different stress conditions, both in the morning or afternoon. The low stressor elevated plasma corticosterone levels and impaired 1 h recognition memory performance when animals were tested in the morning. Exposure to the higher stress condition elevated plasma corticosterone levels and impaired memory performance, independently of the testing time. These findings show that stress impairing effects on short-term recognition memory are dependent on the intensity of stress and circadian rhythm. URB597 (0.3 mg kg-1) rescued the altered memory performance and decreased corticosterone levels in all the impaired groups yet leaving memory unaltered in the non-impaired groups.
Collapse
Affiliation(s)
- Alessia Santori
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | - Paola Colucci
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | | | - Maria Morena
- Hotchkiss Brain Institute, Depts. of Cell Biology and Anatomy &Psychiatry, University of Calgary, T2N 4N1, Calgary, Canada
| | - Maura Palmery
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | - Viviana Trezza
- Dept. of Science, Section of Biomedical Sciences and Technologies, University Roma Tre, 00146, Rome, Italy
| | | | - Matthew N Hill
- Hotchkiss Brain Institute, Depts. of Cell Biology and Anatomy &Psychiatry, University of Calgary, T2N 4N1, Calgary, Canada
| | - Patrizia Campolongo
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy; Neurobiology of Behavior Laboratory, Santa Lucia Foundation, 00143, Rome, Italy.
| |
Collapse
|
30
|
Rapid Reconfiguration of the Functional Connectome after Chemogenetic Locus Coeruleus Activation. Neuron 2019; 103:702-718.e5. [DOI: 10.1016/j.neuron.2019.05.034] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/15/2019] [Accepted: 05/21/2019] [Indexed: 12/14/2022]
|