1
|
Hartmann J, Klengel C, Dillmann LJ, Hisey EE, Hafner K, Shukla R, Soliva Estruch M, Bajaj T, Ebert T, Mabbott KG, Rostin L, Philipsen A, Carlezon WA, Gisabella B, McCullumsmith RE, Vergis JM, Klengel T, Berretta S, Daskalakis NP, Pantazopoulos H, Gassen NC, Ressler KJ. SKA2 enhances stress-related glucocorticoid receptor signaling through FKBP4-FKBP5 interactions in neurons. Proc Natl Acad Sci U S A 2024; 121:e2417728121. [PMID: 39705315 DOI: 10.1073/pnas.2417728121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/15/2024] [Indexed: 12/22/2024] Open
Abstract
Genes involved in regulating the hypothalamic-pituitary-adrenal (HPA) axis, including the glucocorticoid receptor (GR), are linked to various stress-related psychopathologies including bipolar disorder as well as other mood and trauma-related disorders. The protein product of the cell cycle gene, SKA2, is a GR interaction partner in peripheral cells. However, the precise roles of SKA2 in stress and GR signaling in the brain, specifically in nonreplicating postmitotic neurons, and its involvement in HPA axis regulation remain unclear. Here, we demonstrate, using diverse in vitro cell assays, a mechanism by which SKA2 promotes GR signaling through enhancing GR-FKBP4 interaction leading to dissociation of FK506-bindingprotein 51 (FKBP5) from the complex. FKBP4 and FKBP5 are cochaperones known to regulate GR function in opposite directions. Notably in mice, SKA2 in Crh+ neurons of the paraventricular nucleus of the hypothalamus is crucial for HPA axis responsiveness and for maintaining the negative feedback loop underlying allostasis. Moreover, we show that SKA2 expression is increased in postmortem human hippocampus and amygdala from individuals with BD. Our study highlights a critical role of SKA2 in HPA axis function, adds to the understanding of the molecular basis of stress-related psychiatric disorders, and points to potential targets for intervention.
Collapse
Affiliation(s)
- Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Claudia Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Larissa J Dillmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Erin E Hisey
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Rammohan Shukla
- Department of Neuroscience, University of Wyoming, Laramie, WY 82071
| | - Marina Soliva Estruch
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Thomas Bajaj
- Department of Psychiatry and Psychotherapy, Research Group Neurohomeostasis, University Hospital, Boon 53127, Germany
| | - Tim Ebert
- Department of Psychiatry and Psychotherapy, Research Group Neurohomeostasis, University Hospital, Boon 53127, Germany
| | - Katharine G Mabbott
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Luise Rostin
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital, Bonn 53127, Germany
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | | | - John M Vergis
- Department of Neurosciences, University of Toledo, Toledo, OH 43614
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Nils C Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- Department of Psychiatry and Psychotherapy, Research Group Neurohomeostasis, University Hospital, Boon 53127, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| |
Collapse
|
2
|
Bordes J, Bajaj T, Miranda L, van Doeselaar L, Brix LM, Narayan S, Yang H, Mitra S, Kovarova V, Springer M, Kleigrewe K, Müller-Myhsok B, Gassen NC, Schmidt MV. Sex-specific fear acquisition following early life stress is linked to amygdala and hippocampal purine and glutamate metabolism. Commun Biol 2024; 7:1684. [PMID: 39702524 DOI: 10.1038/s42003-024-07396-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
Early life stress (ELS) can negatively impact health, increasing the risk of stress-related disorders, such as post-traumatic stress disorder (PTSD). Importantly, PTSD disproportionately affects women, emphasizing the critical need to explore how sex differences influence the genetic and metabolic neurobiological pathways underlying trauma-related behaviors. This study uses the limited bedding and nesting (LBN) paradigm to model ELS and investigate its sex-specific effects on fear memory formation. Employing innovative unsupervised behavioral classification, the current study reveals distinct behavioral patterns associated with fear acquisition and retrieval in male and female mice following ELS. Females exposed to LBN display heightened active fear responses, contrasting with males. Furthermore, the study examined the crucial link between behavioral regulation and cellular metabolism in key brain regions involved in fear and stress processing. Sex-specific and stress-dependent alterations were observed in purine, pyrimidine, and glutamate metabolism within the basolateral amygdala, the dorsal hippocampus, and the ventral hippocampus. These findings provide crucial insights into the complex interplay between metabolic pathways, the neurobiological underpinnings of fear memory, and stress responses. Importantly, they emphasize the significance of considering sex-specific metabolic alterations when investigating stress-related disorders, opening potential avenues for the development of targeted interventions.
Collapse
Affiliation(s)
- Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Thomas Bajaj
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127, Bonn, Germany
| | - Lucas Miranda
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Lotte van Doeselaar
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Lea Maria Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Sowmya Narayan
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Huanqing Yang
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Shiladitya Mitra
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Veronika Kovarova
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Margherita Springer
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Bertram Müller-Myhsok
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Nils C Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127, Bonn, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
3
|
Göver T, Slezak M. Targeting glucocorticoid receptor signaling pathway for treatment of stress-related brain disorders. Pharmacol Rep 2024; 76:1333-1345. [PMID: 39361217 PMCID: PMC11582215 DOI: 10.1007/s43440-024-00654-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 11/22/2024]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis plays a central role in governing stress-related disorders such as major depressive disorder (MDD), anxiety, and post-traumatic stress disorder. Chronic stress or early life trauma, known risk factors of disease, alter HPA axis activity and pattern of glucocorticoid (GC) secretion. These changes have consequences for physiological processes controlled by glucocorticoid receptor (GR) signaling, such as immune response and metabolism. In the brain, the aberrant GR signaling translates to altered behavior, making the GR pathway a viable target for therapies of stress-related disorders. One of the crucial elements of the pathway is FKBP5, a regulator of GR sensitivity and feedback control within the HPA axis, in which genetic variants were shown to moderate the risk of developing psychiatric conditions. The difficulty in targeting the GR-FKBP5 pathway stems from tailoring the intervention to specific brain regions and cell types, in the context of personalized genetic variations in GR and GR-associated genes, like FKBP5. The development of selective inhibitors, antagonists, and approaches based on targeted protein degradation offer insights into mechanistic aspects of disease and pave the way for improved therapy. These strategies can be employed either independently or in conjunction with conventional medications. Concomitant advancements in personalized drug screening (e.g. in vitro models exploiting induced pluripotent stem cells, iPSCs) bring the potential for optimization of therapy aiming to rescue central deficits originating from the HPA imbalance. In this mini-review, we discuss potential therapeutic strategies targeting GR signaling in stress-related disorders, with a focus on personalized approaches and advancements in drug development.
Collapse
Affiliation(s)
- Tansu Göver
- Lukasiewicz Research Network - PORT Polish Center for Technology Development, ul. Stabłowicka 147, 54-066, Wroclaw, Poland
- Department of Biophysics and Neuroscience, Wroclaw Medical University, ul. Chałubińskiego 3A, 50-368, Wroclaw, Poland
| | - Michal Slezak
- Lukasiewicz Research Network - PORT Polish Center for Technology Development, ul. Stabłowicka 147, 54-066, Wroclaw, Poland.
| |
Collapse
|
4
|
Hu L, Qiu MJ, Fan WJ, Wang WE, Liu SH, Liu XQ, Liu SW, Shen ZJ, Zheng YF, Liu GC, Jia ZY, Wang XQ, Fang N. Characterization of GABAergic marker expression in prefrontal cortex in dexamethasone induced depression/anxiety model. Front Endocrinol (Lausanne) 2024; 15:1433026. [PMID: 39483976 PMCID: PMC11524930 DOI: 10.3389/fendo.2024.1433026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background The pivotal responsibility of GABAergic interneurons is inhibitory neurotransmission; in this way, their significance lies in regulating the maintenance of excitation/inhibition (E/I) balance in cortical circuits. An abundance of glucocorticoids (GCs) exposure results in a disorder of GABAergic interneurons in the prefrontal cortex (PFC); the relationship between this status and an enhanced vulnerability to neuropsychiatric ailments, like depression and anxiety, has been identified, but this connection is still poorly understood because systematic and comprehensive research is lacking. Here, we aim to investigate the impact of dexamethasone (DEX, a GC receptor agonist) on GABAergic interneurons in the PFC of eight-week-old adult male mice. Methods A double-blind study was conducted where thirty-two mice were treated subcutaneously either saline or DEX (0.2 mg/10 ml per kg of body weight) dissolved in saline daily for 21 days. Weight measurements were taken at five-day intervals to assess the emotional changes in mice as well as the response to DEX treatment. Following the 21-day regimen of DEX injections, mice underwent examinations for depression/anxiety-like behaviours and GABAergic marker expression in PFC. Results In a depression/anxiety model generated by chronic DEX treatment, we found that our DEX procedure did trigger depression/anxiety-like behaviors in mice. Furthermore, DEX treatment reduced the expression levels of a GABA-synthesizing enzyme (GAD67), Reelin, calcium-binding proteins (parvalbumin and calretinin) and neuropeptides co-expressed in GABAergic neurons (somatostatin, neuropeptide Y and vasoactive intestinal peptide) in the PFC were reduced after 21 days of DEX treatment; these reductions were accompanied by decreases in brain size and cerebral cortex thickness. Conclusion Our results indicate that a reduction in the number of GABAergic interneurons may result in deficiencies in cortical inhibitory neurotransmission, potentially causing an E/I imbalance in the PFC; this insight suggests a potential breakthrough strategy for the treatment of depression and anxiety.
Collapse
Affiliation(s)
- Ling Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ming-Jing Qiu
- Neurological Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen-Juan Fan
- Luohe Medical College, Henan Province Engineering Research Center of Nutrition and Health, Luohe, China
| | - Wan-Er Wang
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Shao-Hao Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xiao-Qi Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Shi-Wei Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Ze-Jin Shen
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Ya-Fei Zheng
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Guang-Chao Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Zi-Yi Jia
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xiao-Qing Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Na Fang
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
5
|
Bigot M, De Badts CH, Benchetrit A, Vicq É, Moigneu C, Meyrel M, Wagner S, Hennrich AA, Houenou J, Lledo PM, Henry C, Alonso M. Disrupted basolateral amygdala circuits supports negative valence bias in depressive states. Transl Psychiatry 2024; 14:382. [PMID: 39300117 DOI: 10.1038/s41398-024-03085-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
Negative bias is an essential characteristic of depressive episodes leading patients to attribute more negative valence to environmental cues. This negative bias affects all levels of information processing including emotional response, attention and memory, leading to the development and maintenance of depressive symptoms. In this context, pleasant stimuli become less attractive and unpleasant ones more aversive, yet the related neural circuits underlying this bias remain largely unknown. By studying a mice model for depression chronically receiving corticosterone (CORT), we showed a negative bias in valence attribution to olfactory stimuli that responds to antidepressant drug. This result paralleled the alterations in odor value assignment we observed in bipolar depressed patients. Given the crucial role of amygdala in valence coding and its strong link with depression, we hypothesized that basolateral amygdala (BLA) circuits alterations might support negative shift associated with depressive states. Contrary to humans, where limits in spatial resolution of imaging tools impair easy amygdala segmentation, recently unravelled specific BLA circuits implicated in negative and positive valence attribution could be studied in mice. Combining CTB and rabies-based tracing with ex vivo measurements of neuronal activity, we demonstrated that negative valence bias is supported by disrupted activity of specific BLA circuits during depressive states. Chronic CORT administration induced decreased recruitment of BLA-to-NAc neurons preferentially involved in positive valence encoding, while increasing recruitment of BLA-to-CeA neurons preferentially involved in negative valence encoding. Importantly, this dysfunction was dampened by chemogenetic hyperactivation of BLA-to-NAc neurons. Moreover, altered BLA activity correlated with durable presynaptic connectivity changes coming from the paraventricular nucleus of the thalamus, recently demonstrated as orchestrating valence assignment in the amygdala. Together, our findings suggest that specific BLA circuits alterations might support negative bias in depressive states and provide new avenues for translational research to understand the mechanisms underlying depression and treatment efficacy.
Collapse
Affiliation(s)
- Mathilde Bigot
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Claire-Hélène De Badts
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
| | - Axel Benchetrit
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
| | - Éléonore Vicq
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
| | - Carine Moigneu
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
| | - Manon Meyrel
- Assistance Publique-Hôpitaux de Paris, Department of psychiatry, Mondor University Hospital, Créteil, France
- NeuroSpin, PsyBrain Team, UNIACT Lab, CEA Saclay, Gif-sur-Yvette, France
- Université Paris Est Créteil, Faculté de Santé de Créteil, INSERM U955, IMRB, Translational Neuropsychiatry team, Créteil, France
| | - Sébastien Wagner
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
| | - Alexandru Adrian Hennrich
- Max von Pettenkofer-Institute Virology, Medical Faculty, and Gene Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Josselin Houenou
- Assistance Publique-Hôpitaux de Paris, Department of psychiatry, Mondor University Hospital, Créteil, France
- NeuroSpin, PsyBrain Team, UNIACT Lab, CEA Saclay, Gif-sur-Yvette, France
- Université Paris Est Créteil, Faculté de Santé de Créteil, INSERM U955, IMRB, Translational Neuropsychiatry team, Créteil, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France
| | - Chantal Henry
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France.
- Université de Paris Cité, Paris, France.
- Departement of Psychiatry, Service Hospitalo-Universitaire, GHU Paris Psychiatrie & Neurosciences, Paris, France.
| | - Mariana Alonso
- Institut Pasteur, Université Paris Cité, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Perception and Action Unit, F-75015, Paris, France.
| |
Collapse
|
6
|
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
|
7
|
Wei Q, Kumar V, Moore S, Li F, Murphy GG, Watson SJ, Akil H. High emotional reactivity is associated with activation of a molecularly distinct hippocampal-amygdala circuit modulated by the glucocorticoid receptor. Neurobiol Stress 2023; 27:100581. [PMID: 37928820 PMCID: PMC10623371 DOI: 10.1016/j.ynstr.2023.100581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/25/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023] Open
Abstract
Emotions are characterized not only by their valence but also by whether they are stable or labile. Yet, we do not understand the molecular or circuit mechanisms that control the dynamic nature of emotional responses. We have shown that glucocorticoid receptor overexpression in the forebrain (GRov) leads to a highly reactive mouse with increased anxiety behavior coupled with greater swings in emotional responses. This phenotype is established early in development and persists into adulthood. However, the neural circuitry mediating this lifelong emotional lability remains unknown. In the present study, optogenetic stimulation in ventral dentate gyrus (vDG) of GRov mice led to a greater range and a prolonged duration of anxiety behavior. cFos expression analysis showed that the amplified behavioral response to vDG activation in GRov mice is coupled to increased neuronal activity in specific brain regions. Relative to wild type mice, GRov mice displayed glutamatergic/GABAergic activation imbalance in ventral CA1 (vCA1) and selectively increased glutamatergic activation in the basal posterior amygdaloid complex. Moreover, forebrain GR overexpression led to increased activation of molecularly distinct subpopulations of neurons within the hippocampus and the posterior basolateral amygdala (pBLA) as evident from the increased cFos co-labeling in the calbindin1+ glutamatergic neurons in vCA1 and in the DARPP-32/Ppp1r1b+ glutamatergic neurons in pBLA. We propose that a molecularly distinct hippocampal-amygdala circuit is shaped by stress early in life and tunes the dynamics of emotional responses.
Collapse
Affiliation(s)
- Qiang Wei
- Corresponding author. Michigan Neuroscience Institute University of Michigan 205 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| | - Vivek Kumar
- Corresponding author. Michigan Neuroscience Institute, University of Michigan, 205 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| | - Shannon Moore
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Fei Li
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Geoffrey G. Murphy
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | |
Collapse
|
8
|
Peay DN, Acuna A, Reynolds CM, Willis C, Takalkar R, Bryce Ortiz J, Conrad CD. Chronic stress leads to persistent and contrasting stellate neuron dendritic hypertrophy in the amygdala of male and female rats, an effect not found in the hippocampus. Neurosci Lett 2023; 812:137403. [PMID: 37473795 DOI: 10.1016/j.neulet.2023.137403] [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: 04/07/2023] [Revised: 07/02/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
In males, chronic stress enhances dendritic complexity in the amygdala, a region important in emotion regulation. An amygdalar subregion, the basolateral amygdala (BLA), is influenced by the hippocampus and prefrontal cortex to coordinate emotional learning and memory. This study quantified changes in dendritic complexity of BLA stellate neurons ten days after an unpredictable chronic stressor ended in both male and female rats. In addition, dendritic complexity of hippocampal neurons in male rats was assessed at a similar timepoint. Following Golgi processing, stressed male and female rats showed enhanced BLA dendritic complexity; increased arborization occurred near the soma in males and distally in females. As the brain was sampled ten days after chronic stress ended, BLA dendritic hypertrophy persisted in both sexes after the stressor had ended. For the hippocampus, CA3 dendritic complexity was similar for control and stressed males when assessed eight days after stress ended, suggesting that any stress-induced changes had resolved. These results show persistent enhancement of BLA dendritic arborization in both sexes following chronic stress, reveal sex differences in how BLA hypertrophy manifests, and suggest a putative neurobiological substrate by which chronic stress may create a vulnerable phenotype for emotional dysfunction.
Collapse
Affiliation(s)
- Dylan N Peay
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Amanda Acuna
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Cindy M Reynolds
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Chris Willis
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Rujuta Takalkar
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - J Bryce Ortiz
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States
| | - Cheryl D Conrad
- Department of Psychology, Arizona State University, Tempe, AZ, 85287-1104, United States.
| |
Collapse
|
9
|
Ye B, Yuan Y, Liu R, Zhou H, Li Y, Sheng Z, Li T, Zhang B, Xu Z, Li Y, Liu Z. Restoring Wnt signaling in a hormone-simulated postpartum depression model remediated imbalanced neurotransmission and depressive-like behaviors. Mol Med 2023; 29:101. [PMID: 37491227 PMCID: PMC10369844 DOI: 10.1186/s10020-023-00697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/09/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Postpartum depression (PPD) is a prevalent mental disorder that negatively impacts mothers and infants. The mechanisms of vulnerability to affective illness in the postpartum period remain largely unknown. Drastic fluctuations in reproductive hormones during the perinatal period generally account for triggering PPD. However, the molecular mechanism underlying the PPD-like behaviors induced by the fluctuations in hormones has rarely been reported. METHODS We utilized hormones-simulated pseudopregnancy (HSP) and hormones-simulated postpartum period (HSPP) rat models to determine how drastic fluctuations in hormone levels affect adult neurotransmission and contribute to depressive-like behaviors. The electrophysiological response of CA1 pyramidal neurons was evaluated by whole-cell patch clamping to identify the hormone-induced modulations of neurotransmission. The statistical significance of differences was assessed with One-way ANOVA and t-test (p < 0.05 was considered significant). RESULTS Reproductive hormones withdrawal induced depressive-like behaviors and disturbed the balance of excitatory and inhibitory transmission in the pyramidal neurons in the hippocampus. Molecular analyses revealed that the blunted Wnt signaling might be responsible for the deficits of synaptic transmission and behaviors. Activation of Wnt signaling increased excitatory and inhibitory synaptic transmission in the hippocampus. Reactivation of Wnt signaling alleviated the anhedonic behaviors and abnormal synaptic transmission. CONCLUSIONS Restoring Wnt signaling in the hormones-simulated postpartum period rat models remediated depression-related anhedonia symptoms and rebalanced the excitation/inhibition ratio by collectively enhancing the plasticity of GABAergic and glutamatergic synapses. The investigations carried out in this research might provide an alternative and prospective treatment strategy for PPD.
Collapse
Affiliation(s)
- Binglu Ye
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Yawei Yuan
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Rui Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Haitao Zhou
- State Key Laboratory of Drug Research and Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.9A Yuquan Road, Beijing, 100049, China
| | - Yujie Li
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Zhihao Sheng
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Tianyu Li
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
- State Key Laboratory of Drug Research and Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bing Zhang
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Zhendong Xu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
| | - Yang Li
- State Key Laboratory of Drug Research and Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No.9A Yuquan Road, Beijing, 100049, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Zhiqiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
| |
Collapse
|
10
|
Nguyen TML, Defaix C, Mendez-David I, Tritschler L, Etting I, Alvarez JC, Choucha W, Colle R, Corruble E, David DJ, Gardier AM. Intranasal (R, S)-ketamine delivery induces sustained antidepressant effects associated with changes in cortical balance of excitatory/inhibitory synaptic activity. Neuropharmacology 2023; 225:109357. [PMID: 36462636 DOI: 10.1016/j.neuropharm.2022.109357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
In 2019, an intranasal (IN) spray of esketamine SPRAVATO® was approved as a fast-acting antidepressant by drug Agencies US FDA and European EMA. At sub-anesthetic doses, (±)-ketamine, a non-competitive glutamate N-methyl-d-aspartate (NMDA) receptor antagonist, increases the overall excitability of the medial prefrontal cortex (mPFC), an effect being essential for its rapid antidepressant activity. We wondered if this effect of ketamine could come from changes in the balance between neuronal excitation and inhibition (E/I balance) in the mPFC. Here, we performed a preclinical approach to study neurochemical and behavioral responses to a single IN ketamine dose in BALB/cJ mice, a strain more sensitive to stress. By using in vivo microdialysis, we measured cortical E/I balance as the ratio between glutamate to GABA extracellular levels 24 h post-ketamine. We found, for the first time, that E/I balance was shifted in favor of excitation rather than inhibition in the mPFC but more robustly with IN KET than with a single intraperitoneal (IP) dose. Increases in plasma and brain ketamine, norketamine and HNKs levels suggest different metabolic profiles of IP and IN ketamine 30 min post-dose. A significantly larger proportion of ketamine and HNKs in the brain are derived from the IN route 30 min post-dose. It may be linked to the greater magnitude in E/I ratio following IN delivery relative to IP at t24 h. This study suggests that both IP and IN are effective brain delivery methods inducing similar sustained antidepressant efficacy of KET, but the way they induced neurotransmitter changes is slightly different.
Collapse
Affiliation(s)
- Thi Mai Loan Nguyen
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Céline Defaix
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Indira Mendez-David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Laurent Tritschler
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Isabelle Etting
- Lab. Pharmacologie-Toxicologie, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | - Jean-Claude Alvarez
- Lab. Pharmacologie-Toxicologie, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | - Walid Choucha
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Romain Colle
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Denis J David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Alain M Gardier
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France.
| |
Collapse
|
11
|
Huang J, Xu F, Yang L, Tuolihong L, Wang X, Du Z, Zhang Y, Yin X, Li Y, Lu K, Wang W. Involvement of the GABAergic system in PTSD and its therapeutic significance. Front Mol Neurosci 2023; 16:1052288. [PMID: 36818657 PMCID: PMC9928765 DOI: 10.3389/fnmol.2023.1052288] [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: 09/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The neurobiological mechanism of post-traumatic stress disorder (PTSD) is poorly understood. The inhibition of GABA neurons, especially in the amygdala, is crucial for the precise regulation of the consolidation, expression, and extinction of fear conditioning. The GABAergic system is involved in the pathophysiological process of PTSD, with several studies demonstrating that the function of the GABAergic system decreases in PTSD patients. This paper reviews the preclinical and clinical studies, neuroimaging techniques, and pharmacological studies of the GABAergic system in PTSD and summarizes the role of the GABAergic system in PTSD. Understanding the role of the GABAergic system in PTSD and searching for new drug targets will be helpful in the treatment of PTSD.
Collapse
Affiliation(s)
| | - Fei Xu
- Department of Psychiatry of School of Public Health, Southern Medical University, Guangzhou, China
| | - Liping Yang
- Department of Applied Psychology of School of Public Health, Southern Medical University, Guangzhou, China
| | - Lina Tuolihong
- Department of Basic Medical of Basic Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyu Wang
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zibo Du
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yiqi Zhang
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xuanlin Yin
- Department of Basic Medical of Basic Medical College, Southern Medical University, Guangzhou, China
| | - Yingjun Li
- Department of Medical Laboratory Science, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Kangrong Lu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, China
| | - Wanshan Wang
- Department of Laboratory Animal Center, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Haikonen J, Englund J, Amarilla SP, Kharybina Z, Shintyapina A, Kegler K, Garcia MS, Atanasova T, Taira T, Hartung H, Lauri SE. Aberrant cortical projections to amygdala GABAergic neurons contribute to developmental circuit dysfunction following early life stress. iScience 2022; 26:105724. [PMID: 36582824 PMCID: PMC9792886 DOI: 10.1016/j.isci.2022.105724] [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: 05/10/2022] [Revised: 10/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Early life stress (ELS) results in enduring dysfunction of the corticolimbic circuitry, underlying emotional and social behavior. However, the neurobiological mechanisms involved remain elusive. Here, we have combined viral tracing and electrophysiological techniques to study the effects of maternal separation (MS) on frontolimbic connectivity and function in young (P14-21) rats. We report that aberrant prefrontal inputs to basolateral amygdala (BLA) GABAergic interneurons transiently increase the strength of feed-forward inhibition in the BLA, which raises LTP induction threshold in MS treated male rats. The enhanced GABAergic activity after MS exposure associates with lower functional synchronization within prefrontal-amygdala networks in vivo. Intriguingly, no differences in these parameters were detected in females, which were also resistant to MS dependent changes in anxiety-like behaviors. Impaired plasticity and synchronization during the sensitive period of circuit refinement may contribute to long-lasting functional changes in the prefrontal-amygdaloid circuitry that predispose to neuropsychiatric conditions later on in life.
Collapse
Affiliation(s)
- Joni Haikonen
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Jonas Englund
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Shyrley Paola Amarilla
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Zoia Kharybina
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Alexandra Shintyapina
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Kristel Kegler
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Marta Saez Garcia
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Tsvetomira Atanasova
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Henrike Hartung
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Sari E. Lauri
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland,Corresponding author
| |
Collapse
|
13
|
McCullough KM, Katrinli S, Hartmann J, Lori A, Klengel C, Missig G, Klengel T, Langford NA, Newman EL, Anderson KJ, Smith AK, Carroll FI, Ressler KJ, Carlezon WA. Blood levels of T-Cell Receptor Excision Circles (TRECs) provide an index of exposure to traumatic stress in mice and humans. Transl Psychiatry 2022; 12:423. [PMID: 36192377 PMCID: PMC9530209 DOI: 10.1038/s41398-022-02159-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Exposure to stress triggers biological changes throughout the body. Accumulating evidence indicates that alterations in immune system function are associated with the development of stress-associated illnesses such as major depressive disorder and post-traumatic stress disorder, increasing interest in identifying immune markers that provide insight into mental health. Recombination events during T-cell receptor rearrangement and T-cell maturation in the thymus produce circular DNA fragments called T-cell receptor excision circles (TRECs) that can be utilized as indicators of thymic function and numbers of newly emigrating T-cells. Given data suggesting that stress affects thymus function, we examined whether blood levels of TRECs might serve as a quantitative peripheral index of cumulative stress exposure and its physiological correlates. We hypothesized that chronic stress exposure would compromise thymus function and produce corresponding decreases in levels of TRECs. In male mice, exposure to chronic social defeat stress (CSDS) produced thymic involution, adrenal hypertrophy, and decreased levels of TRECs in blood. Extending these studies to humans revealed robust inverse correlations between levels of circulating TRECs and childhood emotional and physical abuse. Cell-type specific analyses also revealed associations between TREC levels and blood cell composition, as well as cell-type specific methylation changes in CD4T + and CD8T + cells. Additionally, TREC levels correlated with epigenetic age acceleration, a common biomarker of stress exposure. Our findings demonstrate alignment between findings in mice and humans and suggest that blood-borne TRECs are a translationally-relevant biomarker that correlates with, and provides insight into, the cumulative physiological and immune-related impacts of stress exposure in mammals.
Collapse
Affiliation(s)
- Kenneth M McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Jakob Hartmann
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Adriana Lori
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Claudia Klengel
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Galen Missig
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Torsten Klengel
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Nicole A Langford
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Emily L Newman
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kasey J Anderson
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, USA
| | - Kerry J Ressler
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - William A Carlezon
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
14
|
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
|
15
|
Loureirin C and Xanthoceraside Prevent Abnormal Behaviors Associated with Downregulation of Brain Derived Neurotrophic Factor and AKT/mTOR/CREB Signaling in the Prefrontal Cortex Induced by Chronic Corticosterone Exposure in Mice. Neurochem Res 2022; 47:2865-2879. [DOI: 10.1007/s11064-022-03694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/27/2022] [Accepted: 07/14/2022] [Indexed: 11/27/2022]
|
16
|
de Kloet ER. Brain mineralocorticoid and glucocorticoid receptor balance in neuroendocrine regulation and stress-related psychiatric etiopathologies. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 24:100352. [PMID: 38037568 PMCID: PMC10687720 DOI: 10.1016/j.coemr.2022.100352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cortisol and corticosterone (CORT) coordinate circadian events and manage the stress response by differential activation of two complementary brain receptor systems, i.e., the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR), which mediate rapid non-genomic and slow genomic actions. Several recent discoveries are highlighted from molecular fine-tuning of the MR/GR balance by FKBP5 to CORTs role in neural network regulation underlying stress adaptation in emotional, cognitive, and social domains of behavior. The data suggest that MR mediates CORT action on risk assessment, social interaction, and response selection, while GR activation promotes memory consolidation and behavioral adaptation; there are also sex differences in CORT action. New evidence suggests that targeting the MR/GR balance resets a dysregulated stress response system and promotes resilience.
Collapse
Affiliation(s)
- Edo Ronald de Kloet
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, University of Leiden, Leiden, the Netherlands
| |
Collapse
|
17
|
Ketamine exerts its sustained antidepressant effects via cell-type-specific regulation of Kcnq2. Neuron 2022; 110:2283-2298.e9. [PMID: 35649415 DOI: 10.1016/j.neuron.2022.05.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/01/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022]
Abstract
A single sub-anesthetic dose of ketamine produces a rapid and sustained antidepressant response, yet the molecular mechanisms responsible for this remain unclear. Here, we identified cell-type-specific transcriptional signatures associated with a sustained ketamine response in mice. Most interestingly, we identified the Kcnq2 gene as an important downstream regulator of ketamine action in glutamatergic neurons of the ventral hippocampus. We validated these findings through a series of complementary molecular, electrophysiological, cellular, pharmacological, behavioral, and functional experiments. We demonstrated that adjunctive treatment with retigabine, a KCNQ activator, augments ketamine's antidepressant-like effects in mice. Intriguingly, these effects are ketamine specific, as they do not modulate a response to classical antidepressants, such as escitalopram. These findings significantly advance our understanding of the mechanisms underlying the sustained antidepressant effects of ketamine, with important clinical implications.
Collapse
|
18
|
Díaz-Hung ML, Hetz C. Proteostasis and resilience: on the interphase between individual's and intracellular stress. Trends Endocrinol Metab 2022; 33:305-317. [PMID: 35337729 DOI: 10.1016/j.tem.2022.02.003] [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: 11/23/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
Abstract
A long proportion of the population is resilient to the negative consequences of stress. Glucocorticoids resulting from endocrine responses to stress are essential adaptive mediators, but also drive alterations to brain function, negatively impacting neuronal connectivity, synaptic plasticity, and memory-related processes. Recent evidence has indicated that organelle function and cellular stress responses are relevant determinant of vulnerability and resistance to environmental stress. At the molecular level, a fundamental mechanism of cellular stress adaptation is the maintenance of proteostasis, which also have key roles in sustaining basal neuronal function. Here, we discuss recent evidence suggesting that proteostasis unbalance at the level of the endoplasmic reticulum, the main site for protein folding in the cell, represents a possible mechanistic link between individuals and cellular stress.
Collapse
Affiliation(s)
- Mei-Li Díaz-Hung
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
19
|
Glucocorticoid Receptor β Isoform Predominates in the Human Dysplastic Brain Region and Is Modulated by Age, Sex, and Antiseizure Medication. Int J Mol Sci 2022; 23:ijms23094940. [PMID: 35563330 PMCID: PMC9099578 DOI: 10.3390/ijms23094940] [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: 04/07/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
The glucocorticoid receptor (GR) at the blood−brain barrier (BBB) is involved in the pathogenesis of drug-resistant epilepsy with focal cortical dysplasia (FCD); however, the roles of GR isoforms GRα and GRβ in the dysplastic brain have not been revealed. We utilized dysplastic/epileptic and non-dysplastic brain tissue from patients who underwent resective epilepsy surgery to identify the GRα and GRβ levels, subcellular localization, and cellular specificity. BBB endothelial cells isolated from the dysplastic brain tissue (EPI-ECs) were used to decipher the key BBB proteins related to drug regulation and BBB integrity compared to control and transfected GRβ-overexpressed BBB endothelial cells. GRβ was upregulated in dysplastic compared to non-dysplastic tissues, and an imbalance of the GRα/GRβ ratio was significant in females vs. males and in patients > 45 years old. In EPI-ECs, the subcellular localization and expression patterns of GRβ, Hsp90, CYP3A4, and CYP2C9 were consistent with GRβ+ brain endothelial cells. Active matrix metalloproteinase levels and activity increased, whereas claudin-5 levels decreased in both EPI-ECs and GRβ+ endothelial cells. In conclusion, the GRβ has a major effect on dysplastic BBB functional proteins and is age and gender-dependent, suggesting a critical role of brain GRβ in dysplasia as a potential biomarker and therapeutic target in epilepsy.
Collapse
|
20
|
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: 77] [Impact Index Per Article: 25.7] [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
|
21
|
Hwang HM, Hashimoto-Torii K. Activation of the anterior cingulate cortex ameliorates anxiety in a preclinical model of fetal alcohol spectrum disorders. Transl Psychiatry 2022; 12:24. [PMID: 35058425 PMCID: PMC8776849 DOI: 10.1038/s41398-022-01789-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 11/30/2022] Open
Abstract
People with fetal alcohol spectrum disorders (FASD) are suffered from a wide range of interlinked cognitive and psychological problems. However, few therapeutic options are available for those patients due to limited dissection of its underlying etiology. Here we found that prenatal alcohol exposure (PAE) increases anxiety in mice due to a dysregulated functional connectivity between the anterior cingulate cortex (ACC) and basolateral amygdala (BLA). We also show that chemogenetic activation of excitatory neurons in the ACC reduced this anxiety behavior in the PAE mice. Interestingly, although the level of plasma corticosterone correlated with the increase in anxiety in the PAE, this level was not altered by chemogenetic activation of the ACC, suggesting that the functional connectivity between the ACC and the BLA does not alter the activity of the hypothalamic-pituitary-adrenal axis. Altogether, this study demonstrated that reduced excitation in the ACC is a cause of anxiety in the PAE mice, providing critical insights into the ACC-BLA neural circuit as a potential target for treating anxiety in FASD patients.
Collapse
Affiliation(s)
- Hye M. Hwang
- grid.239560.b0000 0004 0482 1586Center for Neuroscience Research, The Children’s Research Institute, Children’s National Hospital, Washington, DC USA ,grid.253615.60000 0004 1936 9510The Institute for Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
22
|
Chakraborty P, Chattarji S, Jeanneteau F. A salience hypothesis of stress in PTSD. Eur J Neurosci 2021; 54:8029-8051. [PMID: 34766390 DOI: 10.1111/ejn.15526] [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: 07/10/2021] [Revised: 09/13/2021] [Accepted: 10/30/2021] [Indexed: 11/30/2022]
Abstract
Attention to key features of contexts and things is a necessary tool for all organisms. Detecting these salient features of cues, or simply, salience, can also be affected by exposure to traumatic stress, as has been widely reported in individuals suffering from post-traumatic stress disorder (PTSD). Interestingly, similar observations have been robustly replicated across many animal models of stress as well. By using evidence from such rodent stress paradigms, in the present review, we explore PTSD through the lens of salience processing. In this context, we propose that interaction between the neurotrophin brain-derived neurotrophic factor (BDNF) and glucocorticoids determines the long lasting cellular and behavioural consequences of stress salience. We also describe the dual effect of glucocorticoid therapy in the amelioration of PTSD symptoms. Finally, by integrating in vivo observations at multiple scales of plasticity, we propose a unifying hypothesis that pivots on a crucial role of glucocorticoid signalling in dynamically orchestrating stress salience.
Collapse
Affiliation(s)
- Prabahan Chakraborty
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France.,Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India
| | - Sumantra Chattarji
- Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Freddy Jeanneteau
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France
| |
Collapse
|
23
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
24
|
Gulyaeva NV. Glucocorticoid Regulation of the Glutamatergic
Synapse: Mechanisms of Stress-Dependent Neuroplasticity. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021030091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
Hartmann J, Bajaj T, Klengel C, Chatzinakos C, Ebert T, Dedic N, McCullough KM, Lardenoije R, Joëls M, Meijer OC, McCann KE, Dudek SM, Sarabdjitsingh RA, Daskalakis NP, Klengel T, Gassen NC, Schmidt MV, Ressler KJ. Mineralocorticoid receptors dampen glucocorticoid receptor sensitivity to stress via regulation of FKBP5. Cell Rep 2021; 35:109185. [PMID: 34077736 PMCID: PMC8244946 DOI: 10.1016/j.celrep.2021.109185] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/04/2021] [Accepted: 05/05/2021] [Indexed: 01/23/2023] Open
Abstract
Responding to different dynamic levels of stress is critical for mammalian survival. Disruption of mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) signaling is proposed to underlie hypothalamic-pituitary-adrenal (HPA) axis dysregulation observed in stress-related psychiatric disorders. In this study, we show that FK506-binding protein 51 (FKBP5) plays a critical role in fine-tuning MR:GR balance in the hippocampus. Biotinylated-oligonucleotide immunoprecipitation in primary hippocampal neurons reveals that MR binding, rather than GR binding, to the Fkbp5 gene regulates FKBP5 expression during baseline activity of glucocorticoids. Notably, FKBP5 and MR exhibit similar hippocampal expression patterns in mice and humans, which are distinct from that of the GR. Pharmacological inhibition and region- and cell type-specific receptor deletion in mice further demonstrate that lack of MR decreases hippocampal Fkbp5 levels and dampens the stress-induced increase in glucocorticoid levels. Overall, our findings demonstrate that MR-dependent changes in baseline Fkbp5 expression modify GR sensitivity to glucocorticoids, providing insight into mechanisms of stress homeostasis.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Gene Deletion
- Gene Expression Regulation
- Hippocampus/metabolism
- Humans
- Male
- Mice, Inbred C57BL
- Models, Biological
- Neurons/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/metabolism
- Stress, Physiological
- Tacrolimus Binding Proteins/genetics
- Tacrolimus Binding Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA.
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127 Bonn, Germany
| | - Claudia Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| | - Chris Chatzinakos
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tim Ebert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127 Bonn, Germany
| | - Nina Dedic
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| | - Kenneth M McCullough
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| | - Roy Lardenoije
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Marian Joëls
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht, 3584 CG Utrecht, the Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Katharine E McCann
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - R Angela Sarabdjitsingh
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht, 3584 CG Utrecht, the Netherlands
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127 Bonn, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA.
| |
Collapse
|
26
|
Yin YY, Wang YH, Liu WG, Yao JQ, Yuan J, Li ZH, Ran YH, Zhang LM, Li YF. The role of the excitation:inhibition functional balance in the mPFC in the onset of antidepressants. Neuropharmacology 2021; 191:108573. [PMID: 33945826 DOI: 10.1016/j.neuropharm.2021.108573] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Currently available antidepressants, such as selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs), generally require weeks to months to produce a therapeutic response, but the mechanism of action underlying the delayed onset of antidepressant-like action remains to be elucidated. The balance between excitatory glutamatergic pyramidal neurons and inhibitory γ-aminobutyric acid (GABA) interneurons, i.e., the excitation:inhibition functional (E:I) balance, in the medial prefrontal cortex (mPFC) is critical in regulating several behaviors and might play an important mediating role in the mechanism of rapid antidepressant-like action reported by several studies. In the present study, the multichannel electrophysiological technique was used to record the firing activities of pyramidal neurons and interneurons and investigate the effects of a single dose of fluoxetine and ketamine (both 10 mg/kg, i.p.) on the E:I functional balance in the rat mPFC after 90 min or 24 h, and the forced swimming test (FST) was used to evaluate the antidepressant-like effects of fluoxetine and ketamine. The present study also explored the effects of chronic treatment with fluoxetine (10 mg/kg, i.g.) for 7 d or 21 d on the E:I functional balance in the mPFC. The present results suggested that a single dose of ketamine could both significantly increase the firing activities of pyramidal neurons and significantly decrease the firing activities of interneurons in the mPFC and exerted significant antidepressant-like action on the FST after 90 min and 24 h, but fluoxetine had no such effects under the same conditions. However, chronic treatment with fluoxetine for 21 d (but not 7 d) could significantly affect the firing activities of pyramidal neurons and interneurons in the mPFC. Taken together, the present results indicated that rapid regulation of the E:I functional balance in the mPFC might be an important common mechanism of rapid-acting antidepressants and the delayed onset of SSRIs might be partly attributed to their inability to rapidly regulate the E:I functional balance in the mPFC. The present study provided a new entry point to the development of rapid-acting antidepressants.
Collapse
Affiliation(s)
- Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yun-Hui Wang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | | | - Jun-Qi Yao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Jin Yuan
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Ze-Han Li
- Capital Normal University High School, Beijing, China
| | - Yu-Hua Ran
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China; Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|
27
|
HUZARD D, RAPPENEAU V, MEIJER OC, TOUMA C, ARANGO-LIEVANO M, GARABEDIAN MJ, JEANNETEAU F. Experience and activity-dependent control of glucocorticoid receptors during the stress response in large-scale brain networks. Stress 2021; 24:130-153. [PMID: 32755268 PMCID: PMC7907260 DOI: 10.1080/10253890.2020.1806226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.
Collapse
Affiliation(s)
- Damien HUZARD
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Virginie RAPPENEAU
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Onno C. MEIJER
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Chadi TOUMA
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Margarita ARANGO-LIEVANO
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Freddy JEANNETEAU
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
- Corresponding author:
| |
Collapse
|
28
|
Molendijk ML, de Kloet ER. Forced swim stressor: Trends in usage and mechanistic consideration. Eur J Neurosci 2021; 55:2813-2831. [PMID: 33548153 PMCID: PMC9291081 DOI: 10.1111/ejn.15139] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022]
Abstract
The acquired immobility response during the “forced swim test (FST)” is not a rodent model of depression, but the test has some validity in predicting a compound's antidepressant potential. Nevertheless, 60% of the about 600 papers that were published annually the past 2 years label the rodent's immobility response as depression‐like behaviour, but the relative contribution per country is changing. When the Editors‐in‐Chief of 5 journals publishing most FST papers were asked for their point of view on labelling immobility as depression‐like behaviour and despair, they responded that they primarily rely on the reviewers regarding scientific merit of the submission. One Editor informs authors of the recent NIMH notice (https://grants.nih.gov/grants/guide/notice‐files/NOT‐MH‐19‐053.html) which encourages investigators to use animal models “for” addressing neurobiological questions rather than as model “of” specific mental disorders. The neurobiological questions raised by use of the FST fall in two categories. First, research on the role of endocrine and metabolic factors, with roots in the 1980s, and with focus on the bottom‐up action of glucocorticoids on circuits processing salient information, executive control and memory consolidation. Second, recent findings using novel technological and computational advances that have allowed great progress in charting top‐down control in the switch from active to passive coping with the inescapable stressor executed by neuronal ensembles of the medial prefrontal cortex via the peri‐aquaductal grey. It is expected that combining neural top‐down and endocrine bottom‐up approaches will provide new insights in the role of stress‐coping and adaptation in pathogenesis of mental disorders.
Collapse
Affiliation(s)
- Marc L Molendijk
- Institute of Psychology, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden University Medical Center, Leiden, The Netherlands
| | - E Ronald de Kloet
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Pervin M, Unno K, Konishi T, Nakamura Y. L-Arginine Exerts Excellent Anti-Stress Effects on Stress-Induced Shortened Lifespan, Cognitive Decline and Depression. Int J Mol Sci 2021; 22:E508. [PMID: 33419170 PMCID: PMC7825557 DOI: 10.3390/ijms22020508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/23/2022] Open
Abstract
The anti-stress potential of dietary L-arginine (Arg) was assessed in psychosocially stress-loaded senescence-accelerated (SAMP10) mice. Although this strain of mouse is sensitive to stress, daily administration of Arg at 3 mg/kg significantly suppressed aging-related cognitive decline and behavioral depression at nine months of age and counteracted stress-induced shortened lifespan. To investigate the mechanism of the anti-stress effect of Arg in the brain, early changes in oxidative damage and gene expression levels were measured using SAMP10 mice that were stress-loaded for three days. Increased lipid peroxidation in the brains of stressed mice was significantly lowered by Arg intake. Several genes associated with oxidative stress response and neuronal excitotoxic cell death, including Nr4a1, Arc, and Cyr61, remarkably increased in response to psychosocial stress; however, their expression was significantly suppressed in mice that ingested Arg even under stress conditions. In contrast, the genes that maintain mitochondrial functions and neuronal survival, including Hba-a2 and Hbb-b2, were significantly increased in mice that ingested Arg. These results indicate that Arg reduces oxidative damage and enhances mitochondrial functions in the brain. We suggest that the daily intake of Arg plays important roles in reducing stress-induced brain damage and slowing aging.
Collapse
Affiliation(s)
- Monira Pervin
- Tea Science Center, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| | - Keiko Unno
- Tea Science Center, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| | - Tomokazu Konishi
- Faculty of Bioresource Sciences, Akita Prefectural University, Shimoshinjo Nakano, Akita 010-0195, Japan;
| | - Yoriyuki Nakamura
- Tea Science Center, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| |
Collapse
|
30
|
Silkis IG. The Possible Mechanism of the Appearance of Nightmares in Post-Traumatic Stress Disorder and Approaches to Their Prevention. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419030127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
31
|
Besnard A, Langberg T, Levinson S, Chu D, Vicidomini C, Scobie KN, Dwork AJ, Arango V, Rosoklija GB, Mann JJ, Hen R, Leonardo ED, Boldrini M, Sahay A. Targeting Kruppel-like Factor 9 in Excitatory Neurons Protects against Chronic Stress-Induced Impairments in Dendritic Spines and Fear Responses. Cell Rep 2019; 23:3183-3196. [PMID: 29898391 PMCID: PMC7453932 DOI: 10.1016/j.celrep.2018.05.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 04/09/2018] [Accepted: 05/14/2018] [Indexed: 11/01/2022] Open
Abstract
Stress exposure is associated with the pathogenesis of psychiatric disorders, including post-traumatic stress disorder (PTSD) and major depressive disorder (MDD). Here, we show in rodents that chronic stress exposure rapidly and transiently elevates hippocampal expression of Kruppel-like factor 9 (Klf9). Inducible genetic silencing of Klf9 expression in excitatory forebrain neurons in adulthood prior to, but not after, onset of stressor prevented chronic restraint stress (CRS)-induced potentiation of contextual fear acquisition in female mice and chronic corticosterone (CORT) exposure-induced fear generalization in male mice. Klf9 silencing prevented chronic CORT and CRS induced enlargement of dendritic spines in the ventral hippocampus of male and female mice, respectively. KLF9 mRNA density was increased in the anterior dentate gyrus of women, but not men, with more severe recent stressful life events and increased mortality. Thus, Klf9 functions as a stress-responsive transcription factor that mediates circuit and behavioral resilience in a sex-specific manner.
Collapse
Affiliation(s)
- Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Tomer Langberg
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Sally Levinson
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Duong Chu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Cinzia Vicidomini
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Kimberly N Scobie
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Divisions of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA; Macedonian Academy of Sciences & Arts, Skopje 1000, Republic of Macedonia
| | - Victoria Arango
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Divisions of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Gorazd B Rosoklija
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Divisions of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA; Macedonian Academy of Sciences & Arts, Skopje 1000, Republic of Macedonia
| | - J John Mann
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Divisions of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA
| | - René Hen
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Department of Neuroscience, Columbia University Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Medical Center, New York, NY 10032, USA; Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - E David Leonardo
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Maura Boldrini
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Divisions of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA; BROAD Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
32
|
Maddox SA, Hartmann J, Ross RA, Ressler KJ. Deconstructing the Gestalt: Mechanisms of Fear, Threat, and Trauma Memory Encoding. Neuron 2019; 102:60-74. [PMID: 30946827 DOI: 10.1016/j.neuron.2019.03.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/03/2019] [Accepted: 03/12/2019] [Indexed: 01/14/2023]
Abstract
Threat processing is central to understanding debilitating fear- and trauma-related disorders such as posttraumatic stress disorder (PTSD). Progress has been made in understanding the neural circuits underlying the "engram" of threat or fear memory formation that complements a decades-old appreciation of the neurobiology of fear and threat involving hub structures such as the amygdala. In this review, we examine key recent findings, as well as integrate the importance of hormonal and physiological approaches, to provide a broader perspective of how bodily systems engaged in threat responses may interact with amygdala-based circuits in the encoding and updating of threat-related memory. Understanding how trauma-related memories are encoded and updated throughout the brain and the body will ultimately lead to novel biologically-driven approaches for treatment and prevention.
Collapse
Affiliation(s)
- Stephanie A Maddox
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Jakob Hartmann
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Rachel A Ross
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Kerry J Ressler
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Dedic N, Kühne C, Gomes KS, Hartmann J, Ressler KJ, Schmidt MV, Deussing JM. Deletion of CRH From GABAergic Forebrain Neurons Promotes Stress Resilience and Dampens Stress-Induced Changes in Neuronal Activity. Front Neurosci 2019; 13:986. [PMID: 31619956 PMCID: PMC6763571 DOI: 10.3389/fnins.2019.00986] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of the corticotropin-releasing hormone (CRH) system has been implicated in stress-related psychopathologies such as depression and anxiety. Although most studies have linked CRH/CRH receptor 1 signaling to aversive, stress-like behavior, recent work has revealed a crucial role for distinct CRH circuits in maintaining positive emotional valence and appetitive responses under baseline conditions. Here we addressed whether deletion of CRH, specifically from GABAergic forebrain neurons (Crh CKO-GABA mice) differentially affects general behavior under baseline and chronic stress conditions. Expression mapping in Crh CK O-GABA mice revealed absence of Crh in GABAergic neurons of the cortex and limbic regions including the hippocampus, central nucleus of the amygdala and the bed nucleus of the stria terminals, but not in the paraventricular nucleus of hypothalamus. Consequently, conditional CRH knockout animals exhibited no alterations in circadian and stress-induced corticosterone release compared to controls. Under baseline conditions, absence of Crh from forebrain GABAergic neurons resulted in social interaction deficits but had no effect on other behavioral measures including locomotion, anxiety, immobility in the forced swim test, acoustic startle response and fear conditioning. Interestingly, following exposure to chronic social defeat stress, Crh CKO-GABA mice displayed a resilient phenotype, which was accompanied by a dampened, stress-induced expression of immediate early genes c-fos and zif268 in several brain regions. Collectively our data reveals the requirement of GABAergic CRH circuits in maintaining appropriate social behavior in naïve animals and further supports the ability of CRH to promote divergent behavioral states under baseline and severe stress conditions.
Collapse
Affiliation(s)
- Nina Dedic
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States
| | - Claudia Kühne
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Karina S Gomes
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Laboratory of Neuropsychopharmacology, Paulista State University, Araraquara, Brazil
| | - Jakob Hartmann
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States.,Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States
| | - Mathias V Schmidt
- Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
34
|
Williams S, Ghosh C. Neurovascular glucocorticoid receptors and glucocorticoids: implications in health, neurological disorders and drug therapy. Drug Discov Today 2019; 25:89-106. [PMID: 31541713 DOI: 10.1016/j.drudis.2019.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/12/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Abstract
Glucocorticoid receptors (GRs) are ubiquitous transcription factors widely studied for their role in controlling events related to inflammation, stress and homeostasis. Recently, GRs have reemerged as crucial targets of investigation in neurological disorders, with a focus on pharmacological strategies to direct complex mechanistic GR regulation and improve therapy. In the brain, GRs control functions necessary for neurovascular integrity, including responses to stress, neurological changes mediated by the hypothalamic-pituitary-adrenal axis and brain-specific responses to corticosteroids. Therefore, this review will examine GR regulation at the neurovascular interface in normal and pathological conditions, pharmacological GR modulation and glucocorticoid insensitivity in neurological disorders.
Collapse
Affiliation(s)
- Sherice Williams
- Brain Physiology Laboratory/Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chaitali Ghosh
- Brain Physiology Laboratory/Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine and Biomedical Engineering at Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
35
|
Scheimann JR, Moloney RD, Mahbod P, Morano RL, Fitzgerald M, Hoskins O, Packard BA, Cotella EM, Hu YC, Herman JP. Conditional deletion of glucocorticoid receptors in rat brain results in sex-specific deficits in fear and coping behaviors. eLife 2019; 8:44672. [PMID: 31329100 PMCID: PMC6645713 DOI: 10.7554/elife.44672] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/03/2019] [Indexed: 01/16/2023] Open
Abstract
Glucocorticoid receptors (GR) have diverse functions relevant to maintenance of homeostasis and adaptation to environmental challenges. Understanding the importance of tissue-specific GR function in physiology and behavior has been hampered by near-ubiquitous localization in brain and body. Here we use CRISPR/Cas9 gene editing to create a conditional GR knockdown in Sprague Dawley rats. To test the impact of cell- and region-specific GR knockdown on physiology and behavior, we targeted GR knockdown to output neurons of the prelimbic cortex. Prelimbic knockdown of GR in females caused deficits in acquisition and extinction of fear memory during auditory fear conditioning, whereas males exhibited enhanced active-coping behavior during forced swim. Our data support the utility of this conditional knockdown rat to afford high-precision knockdown of GR across a variety of contexts, ranging from neuronal depletion to circuit-wide manipulations, leveraging the behavioral tractability and enhanced brain size of the rat as a model organism.
Collapse
Affiliation(s)
- Jessie R Scheimann
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Rachel D Moloney
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Parinaz Mahbod
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Rachel L Morano
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Maureen Fitzgerald
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Olivia Hoskins
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Benjamin A Packard
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Evelin M Cotella
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, United States.,Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - James P Herman
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| |
Collapse
|
36
|
Pham TH, Gardier AM. Fast-acting antidepressant activity of ketamine: highlights on brain serotonin, glutamate, and GABA neurotransmission in preclinical studies. Pharmacol Ther 2019; 199:58-90. [DOI: 10.1016/j.pharmthera.2019.02.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
|
37
|
Malikowska-Racia N, Salat K. Recent advances in the neurobiology of posttraumatic stress disorder: A review of possible mechanisms underlying an effective pharmacotherapy. Pharmacol Res 2019; 142:30-49. [PMID: 30742899 DOI: 10.1016/j.phrs.2019.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/24/2019] [Accepted: 02/01/2019] [Indexed: 12/24/2022]
Abstract
Recent progress in the field of neurobiology supported by clinical evidence gradually reveals the mystery of human brain functioning. So far, many psychiatric disorders have been described in great detail, although there are still plenty of cases that are misunderstood. These include posttraumatic stress disorder (PTSD), which is a unique disease that combines a wide range of neurobiological changes, which involve disturbances of the hypothalamic-pituitary-adrenal gland axis, hyperactivation of the amygdala complex, and attenuation of some hippocampal and cortical functions. Such multiplicity results in differential symptomatology, including elevated anxiety, nightmares, fear retrieval episodes that may trigger delusions and hallucinations, sleep disturbances, and many others that strongly interfere with the quality of the patient's life. Because of widespread neurological changes and the disease manifestation, the pharmacotherapy of PTSD remains unclear and requires a multidimensional approach and involvement of polypharmacotherapy. Hopefully, more and more neuroscientists and clinicians will study PTSD, which will provide us with new information that would possibly accelerate establishment of well-tolerated and effective pharmacotherapy. In this review, we have focused on neurobiological changes regarding PTSD, addressing the most disturbed brain structures and neurotransmissions, as well as discussing in detail the recently taken and novel therapeutic paths.
Collapse
Affiliation(s)
- Natalia Malikowska-Racia
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland.
| | - Kinga Salat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland
| |
Collapse
|
38
|
The mineralocorticoid receptor is essential for stress axis regulation in zebrafish larvae. Sci Rep 2018; 8:18081. [PMID: 30591705 PMCID: PMC6308233 DOI: 10.1038/s41598-018-36681-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/26/2018] [Indexed: 11/08/2022] Open
Abstract
The mineralocorticoid receptor (MR) in mammals mediates the effects of aldosterone in regulating fluid balance and potassium homeostasis. While MR signalling is essential for survival in mammals, there is no evidence that MR has any physiological role in ray-finned fish. Teleosts lack aldosterone and emerging evidence suggest that cortisol mediates ion and fluid regulation by activating glucocorticoid receptor (GR) signalling. Consequently, a physiological role for MR signalling, despite its conserved and ancient origin, is still lacking. We tested the hypothesis that a key physiological role for MR signalling in fish is the regulation of stress axis activation and function. Using either MR or GR knockout zebrafish, our results reveal distinct and complementary role for these receptors in stress axis function. GR-/- mutants were hypercortisolemic and failed to elicit a cortisol stress response, while MR-/- mutants showed a delayed, but sustained cortisol response post-stressor. Both these receptors are involved in stress-related behaviour, as the loss of either receptors abolished the glucocorticoid-mediated larval hyperactivity to a light stimulus. Overall, the results underscore a key physiological role for MR signalling in ray-finned fishes, and we propose that the regulation of the highly conserved stress axis as the original function of this receptor.
Collapse
|
39
|
Scheimann JR, Mahbod P, Morano R, Frantz L, Packard B, Campbell K, Herman JP. Deletion of Glucocorticoid Receptors in Forebrain GABAergic Neurons Alters Acute Stress Responding and Passive Avoidance Behavior in Female Mice. Front Behav Neurosci 2018; 12:325. [PMID: 30627088 PMCID: PMC6309161 DOI: 10.3389/fnbeh.2018.00325] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/10/2018] [Indexed: 01/16/2023] Open
Abstract
The glucocorticoid receptor (GR) is critically involved in regulation of stress responses [inhibition of the hypothalamic-pituitary-adrenal (HPA) axis], emotional behavior and cognition via interactions with forebrain corticolimbic circuity. Work to date has largely focused on GR actions in forebrain excitatory neurons; however, recent studies suggest a potential role mediated by interneurons. Here, we targeted GR deletion in forebrain GABAergic neurons, including the cortical interneurons, using a Dlx5/6-Cre driver line to test the role of forebrain interneuronal GR in HPA axis regulation and behavior. Our data indicate that GR deletion in GABAergic neurons causes elevated corticosterone stress responsiveness and decreased cross-over latencies in a passive avoidance task in females, but not males. Dlx5/6-Cre driven gene deletion caused loss of GR in interneurons in the prefrontal cortex (PFC) and hippocampus, but also in select diencephalic GABAergic neurons (including the reticular thalamic nucleus and dorsomedial hypothalamus). Our data suggest that GR signaling in interneurons is differentially important in females, which may have implications for GR-directed therapies for stress-related affective disease states.
Collapse
Affiliation(s)
- Jessie R Scheimann
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Parinaz Mahbod
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Rachel Morano
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Lindsey Frantz
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Ben Packard
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
40
|
Desrumaux CM, Mansuy M, Lemaire S, Przybilski J, Le Guern N, Givalois L, Lagrost L. Brain Vitamin E Deficiency During Development Is Associated With Increased Glutamate Levels and Anxiety in Adult Mice. Front Behav Neurosci 2018; 12:310. [PMID: 30618663 PMCID: PMC6297247 DOI: 10.3389/fnbeh.2018.00310] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022] Open
Abstract
Vitamin E, the most important lipophilic radical scavenging antioxidant in vivo, has a pivotal role in brain. In an earlier study, we observed that adult mice with a defect in the gene encoding plasma phospholipid transfer protein (PLTP) display a moderate reduction in cerebral vitamin E levels, and exacerbated anxiety despite normal locomotion and memory functions. Here we sought to determine whether dietary vitamin E supplementation can modulate neurotransmitter levels and alleviate the increased anxiety phenotype of PLTP-deficient (PLTP−/−) mice. To address this question, a vitamin E-enriched diet was used, and two complementary approches were implemented: (i) “early supplementation”: neurotransmitter levels and anxiety were assessed in 6 months old PLTP−/− mice born from vitamin E-supplemented parents; and (ii) “late supplementation”: neurotransmitter levels and anxiety were assessed in 6 months old PLTP−/− mice fed a vitamin E-enriched diet from weaning. Our results show for the first time that an inadequate supply of vitamin E during development, due to moderate maternal vitamin E deficiency, is associated with reduced brain vitamin E levels at birth and irreversible alterations in brain glutamate levels. They also suggest this deficiency is associated with increased anxiety at adulthood. Thus, the present study leads to conclude on the importance of the micronutrient vitamin E during pregnancy.
Collapse
Affiliation(s)
- Catherine M Desrumaux
- INSERM, U1198, Team "Environmental Impacts in Alzheimer's Disease and Related Disorders" (EiAlz), Montpellier, France.,Faculty of Sciences, Université Montpellier, Montpellier, France.,EPHE, Paris, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Marine Mansuy
- INSERM, U1198, Team "Environmental Impacts in Alzheimer's Disease and Related Disorders" (EiAlz), Montpellier, France.,Faculty of Sciences, Université Montpellier, Montpellier, France.,EPHE, Paris, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Stéphanie Lemaire
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,University Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,Hôpital du Bocage, Dijon, France
| | - Justine Przybilski
- INSERM, U1198, Team "Environmental Impacts in Alzheimer's Disease and Related Disorders" (EiAlz), Montpellier, France.,Faculty of Sciences, Université Montpellier, Montpellier, France.,EPHE, Paris, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Naig Le Guern
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,University Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | - Laurent Givalois
- INSERM, U1198, Team "Environmental Impacts in Alzheimer's Disease and Related Disorders" (EiAlz), Montpellier, France.,Faculty of Sciences, Université Montpellier, Montpellier, France.,EPHE, Paris, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Laurent Lagrost
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,University Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| |
Collapse
|
41
|
Tertil M, Skupio U, Barut J, Dubovyk V, Wawrzczak-Bargiela A, Soltys Z, Golda S, Kudla L, Wiktorowska L, Szklarczyk K, Korostynski M, Przewlocki R, Slezak M. Glucocorticoid receptor signaling in astrocytes is required for aversive memory formation. Transl Psychiatry 2018; 8:255. [PMID: 30487639 PMCID: PMC6261947 DOI: 10.1038/s41398-018-0300-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/15/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022] Open
Abstract
Stress elicits the release of glucocorticoids (GCs) that regulate energy metabolism and play a role in emotional memory. Astrocytes express glucocorticoid receptors (GR), but their contribution to cognitive effects of GC's action in the brain is unknown. To address this question, we studied how astrocyte-specific elimination of GR affects animal behavior known to be regulated by stress. Mice with astrocyte-specific ablation of GR presented impaired aversive memory expression in two different paradigms of Pavlovian learning: contextual fear conditioning and conditioned place aversion. These mice also displayed compromised regulation of genes encoding key elements of the glucose metabolism pathway upon GR stimulation. In particular, we identified that the glial, but not the neuronal isoform of a crucial stress-response molecule, Sgk1, undergoes GR-dependent regulation in vivo and demonstrated the involvement of SGK1 in regulation of glucose uptake in astrocytes. Together, our results reveal astrocytes as a central element in GC-dependent formation of aversive memory and suggest their relevance for stress-induced alteration of brain glucose metabolism. Consequently, astrocytes should be considered as a cellular target of therapies of stress-induced brain diseases.
Collapse
Affiliation(s)
- Magdalena Tertil
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Urszula Skupio
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Justyna Barut
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Valentyna Dubovyk
- Team Brain Microcircuits in Psychiatric Diseases, BioMed X Innovation Center, Heidelberg, 69120 Germany
| | - Agnieszka Wawrzczak-Bargiela
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Zbigniew Soltys
- 0000 0001 2162 9631grid.5522.0Department of Neuroanatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Cracow, 30-387 Poland
| | - Slawomir Golda
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Lucja Kudla
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Lucja Wiktorowska
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Klaudia Szklarczyk
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Michal Korostynski
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Ryszard Przewlocki
- 0000 0001 1958 0162grid.413454.3Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343 Poland
| | - Michal Slezak
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Cracow, 31-343, Poland. .,Team Brain Microcircuits in Psychiatric Diseases, BioMed X Innovation Center, Heidelberg, 69120, Germany.
| |
Collapse
|
42
|
Meijer OC, Buurstede JC, Schaaf MJM. Corticosteroid Receptors in the Brain: Transcriptional Mechanisms for Specificity and Context-Dependent Effects. Cell Mol Neurobiol 2018; 39:539-549. [PMID: 30291573 PMCID: PMC6469829 DOI: 10.1007/s10571-018-0625-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022]
Abstract
Corticosteroid hormones act in the brain to support adaptation to stress via binding to mineralocorticoid and glucocorticoid receptors (MR and GR). These receptors act in large measure as transcription factors. Corticosteroid effects can be highly divergent, depending on the receptor type, but also on brain region, cell type, and physiological context. These differences ultimately depend on differential interactions of MR and GR with other proteins, which determine ligand binding, nuclear translocation, and transcriptional activities. In this review, we discuss established and potential mechanisms that confer receptor and cell type-specific effects of the MR and GR-mediated transcriptional effects in the brain.
Collapse
Affiliation(s)
- Onno C Meijer
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands. .,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
| | - J C Buurstede
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Marcel J M Schaaf
- Department of Animal Sciences and Health (M.J.M.S.), Institute of Biology, Leiden University, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
43
|
Salivary kynurenic acid response to psychological stress: inverse relationship to cortical glutamate in schizophrenia. Neuropsychopharmacology 2018; 43:1706-1711. [PMID: 29728648 PMCID: PMC6006286 DOI: 10.1038/s41386-018-0072-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 12/22/2022]
Abstract
Frontal glutamatergic synapses are thought to be critical for adaptive, long-term stress responses. Prefrontal cortices, including the anterior cingulate cortex (ACC) contribute to stress perception and regulation, and are involved in top-down regulation of peripheral glucocorticoid and inflammatory responses to stress. Levels of kynurenic acid (KYNA) in saliva increase in response to psychological stress, and this stress-induced effect may be abnormal in people with schizophrenia. Here we test the hypothesis that ACC glutamatergic functioning may contribute to the stress-induced salivary KYNA response in schizophrenia. In 56 patients with schizophrenia and 58 healthy controls, our results confirm that levels of KYNA in saliva increase following psychological stress. The magnitude of the effect correlated negatively with proton magnetic resonance spectroscopy (MRS) glutamate + glutamine (r = -.31, p = .017) and glutamate (r = -0.27, p = .047) levels in the ACC in patients but not in the controls (all p ≥ .45). Although, a causal relationship cannot be ascertained in this cross-sectional study, these findings suggest a potentially meaningful link between central glutamate levels and kynurenine pathway response to stress in individuals with schizophrenia.
Collapse
|
44
|
Wei J, Zhong P, Qin L, Tan T, Yan Z. Chemicogenetic Restoration of the Prefrontal Cortex to Amygdala Pathway Ameliorates Stress-Induced Deficits. Cereb Cortex 2018; 28:1980-1990. [PMID: 28498919 PMCID: PMC6018994 DOI: 10.1093/cercor/bhx104] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/27/2017] [Indexed: 01/11/2023] Open
Abstract
Corticosteroid stress hormones exert a profound impact on cognitive and emotional processes. Understanding the neuronal circuits that are altered by chronic stress is important for counteracting the detrimental effects of stress in a brain region- and cell type-specific manner. Using the chemogenetic tool, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), which enables the remote, noninvasive and long-lasting modulation of cellular activity and signal transduction in discrete neuronal populations in vivo, we sought to identify the specific pathways that play an essential role in stress responses. We found that prolonged severe stress induced the diminished glutamatergic projection from pyramidal neurons in prefrontal cortex (PFC) to GABAergic interneurons in basolateral amygdala (BLA), leading to the loss of feedforward inhibition and ensuing hyperexcitability of BLA principal neurons, which caused a variety of behavioral abnormalities. Activating PFC pyramidal neurons with hM3D(Gq) DREADD restored the functional connection between PFC and BLA in stressed animals, resulting in the rescue of recognition memory, normalization of locomotor activity and reduction of aggressive behaviors. Inhibiting BLA principal neurons directly with hM4D(Gi) DREADD also blocked BLA hyperactivity and aggressive behaviors in stressed animals. These results have offered an effective avenue to counteract the stress-induced disruption of circuitry homeostasis.
Collapse
Affiliation(s)
- Jing Wei
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Luye Qin
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Tao Tan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| |
Collapse
|
45
|
Dedic N, Kühne C, Jakovcevski M, Hartmann J, Genewsky AJ, Gomes KS, Anderzhanova E, Pöhlmann ML, Chang S, Kolarz A, Vogl AM, Dine J, Metzger MW, Schmid B, Almada RC, Ressler KJ, Wotjak CT, Grinevich V, Chen A, Schmidt MV, Wurst W, Refojo D, Deussing JM. Chronic CRH depletion from GABAergic, long-range projection neurons in the extended amygdala reduces dopamine release and increases anxiety. Nat Neurosci 2018; 21:803-807. [PMID: 29786085 DOI: 10.1038/s41593-018-0151-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/10/2018] [Indexed: 12/12/2022]
Abstract
The interplay between corticotropin-releasing hormone (CRH) and the dopaminergic system has predominantly been studied in addiction and reward, while CRH-dopamine interactions in anxiety are scarcely understood. We describe a new population of CRH-expressing, GABAergic, long-range-projecting neurons in the extended amygdala that innervate the ventral tegmental area and alter anxiety following chronic CRH depletion. These neurons are part of a distinct CRH circuit that acts anxiolytically by positively modulating dopamine release.
Collapse
Affiliation(s)
- Nina Dedic
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Claudia Kühne
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jakob Hartmann
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - Andreas J Genewsky
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Karina S Gomes
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Laboratory of Neuropsychopharmacology, Paulista State University, Araraquara, Brazil
| | - Elmira Anderzhanova
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Max L Pöhlmann
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Simon Chang
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Adam Kolarz
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Annette M Vogl
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Julien Dine
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Michael W Metzger
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bianca Schmid
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rafael C Almada
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - Carsten T Wotjak
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides, German Cancer Research Center, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mathias V Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Munich, Germany.,Technische Universität München, Chair of Developmental Genetics, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Munich, Munich, Germany
| | - Damian Refojo
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Instituto de Investigacion en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
46
|
Dedic N, Pöhlmann ML, Richter JS, Mehta D, Czamara D, Metzger MW, Dine J, Bedenk BT, Hartmann J, Wagner KV, Jurik A, Almli LM, Lori A, Moosmang S, Hofmann F, Wotjak CT, Rammes G, Eder M, Chen A, Ressler KJ, Wurst W, Schmidt MV, Binder EB, Deussing JM. Cross-disorder risk gene CACNA1C differentially modulates susceptibility to psychiatric disorders during development and adulthood. Mol Psychiatry 2018; 23:533-543. [PMID: 28696432 PMCID: PMC5822460 DOI: 10.1038/mp.2017.133] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022]
Abstract
Single-nucleotide polymorphisms (SNPs) in CACNA1C, the α1C subunit of the voltage-gated L-type calcium channel Cav1.2, rank among the most consistent and replicable genetics findings in psychiatry and have been associated with schizophrenia, bipolar disorder and major depression. However, genetic variants of complex diseases often only confer a marginal increase in disease risk, which is additionally influenced by the environment. Here we show that embryonic deletion of Cacna1c in forebrain glutamatergic neurons promotes the manifestation of endophenotypes related to psychiatric disorders including cognitive decline, impaired synaptic plasticity, reduced sociability, hyperactivity and increased anxiety. Additional analyses revealed that depletion of Cacna1c during embryonic development also increases the susceptibility to chronic stress, which suggest that Cav1.2 interacts with the environment to shape disease vulnerability. Remarkably, this was not observed when Cacna1c was deleted in glutamatergic neurons during adulthood, where the later deletion even improved cognitive flexibility, strengthened synaptic plasticity and induced stress resilience. In a parallel gene × environment design in humans, we additionally demonstrate that SNPs in CACNA1C significantly interact with adverse life events to alter the risk to develop symptoms of psychiatric disorders. Overall, our results further validate Cacna1c as a cross-disorder risk gene in mice and humans, and additionally suggest a differential role for Cav1.2 during development and adulthood in shaping cognition, sociability, emotional behavior and stress susceptibility. This may prompt the consideration for pharmacological manipulation of Cav1.2 in neuropsychiatric disorders with developmental and/or stress-related origins.
Collapse
Affiliation(s)
- N Dedic
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - M L Pöhlmann
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J S Richter
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - D Mehta
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - D Czamara
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia
| | - M W Metzger
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J Dine
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - B T Bedenk
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J Hartmann
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - K V Wagner
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A Jurik
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - L M Almli
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - A Lori
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - S Moosmang
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - F Hofmann
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - C T Wotjak
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - G Rammes
- Clinic of Anaesthesiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - M Eder
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A Chen
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - K J Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - W Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - M V Schmidt
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - E B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - J M Deussing
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
47
|
Abstract
Emotion-related responses, such as fear and anxiety, are important behavioral phenomena in most animal species, as well as in humans. However, the underlying mechanisms of fear and anxiety in animals and in humans are still largely unknown, and anxiety disorders continue to represent a large unmet medical need in the human clinic. Animal models may speed up discovery of these mechanisms and may also lead to betterment of human health. Herein, we report the identification of a chemokine-like gene family, samdori (sam), and present functional characterization of sam2. We observed increased anxiety-related responses in both zebrafish and mouse knockout models. Taken together, these results support a crucial and evolutionarily conserved role of sam2 in regulating anxiety-like behavior. Emotional responses, such as fear and anxiety, are fundamentally important behavioral phenomena with strong fitness components in most animal species. Anxiety-related disorders continue to represent a major unmet medical need in our society, mostly because we still do not fully understand the mechanisms of these diseases. Animal models may speed up discovery of these mechanisms. The zebrafish is a highly promising model organism in this field. Here, we report the identification of a chemokine-like gene family, samdori (sam), and present functional characterization of one of its members, sam2. We show exclusive mRNA expression of sam2 in the CNS, predominantly in the dorsal habenula, telencephalon, and hypothalamus. We found knockout (KO) zebrafish to exhibit altered anxiety-related responses in the tank, scototaxis and shoaling assays, and increased crh mRNA expression in their hypothalamus compared with wild-type fish. To investigate generalizability of our findings to mammals, we developed a Sam2 KO mouse and compared it to wild-type littermates. Consistent with zebrafish findings, homozygous KO mice exhibited signs of elevated anxiety. We also found bath application of purified SAM2 protein to increase inhibitory postsynaptic transmission onto CRH neurons of the paraventricular nucleus. Finally, we identified a human homolog of SAM2, and were able to refine a candidate gene region encompassing SAM2, among 21 annotated genes, which is associated with intellectual disability and autism spectrum disorder in the 12q14.1 deletion syndrome. Taken together, these results suggest a crucial and evolutionarily conserved role of sam2 in regulating mechanisms associated with anxiety.
Collapse
|
48
|
Abstract
Glucocorticoids via the glucocorticoid receptor (GR) have effects on a variety of cell types, eliciting important physiological responses via changes in gene expression and signaling. Although decades of research have illuminated the mechanism of how this important steroid receptor controls gene expression using
in vitro and cell culture–based approaches, how GR responds to changes in external signals
in vivo under normal and pathological conditions remains elusive. The goal of this review is to highlight recent work on GR action in fat cells and liver to affect metabolism
in vivo and the role GR ligands and receptor phosphorylation play in calibrating signaling outputs by GR in the brain in health and disease. We also suggest that both the brain and fat tissue communicate to affect physiology and behavior and that understanding this “brain-fat axis” will enable a more complete understanding of metabolic diseases and inform new ways to target them.
Collapse
Affiliation(s)
- Michael J Garabedian
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Sciences, 450 East 29th Street, Room 324, New York, NY, 10016, USA
| | - Charles A Harris
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Freddy Jeanneteau
- Departments of Physiology and Neuroscience, Institute of Functional Genomics, INSERM U1191, CNRS UMR5203, University of Montpellier, 34094 Montpellier, France
| |
Collapse
|
49
|
Acute Stress Suppresses Synaptic Inhibition and Increases Anxiety via Endocannabinoid Release in the Basolateral Amygdala. J Neurosci 2017; 36:8461-70. [PMID: 27511017 DOI: 10.1523/jneurosci.2279-15.2016] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 06/25/2016] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Stress and glucocorticoids stimulate the rapid mobilization of endocannabinoids in the basolateral amygdala (BLA). Cannabinoid receptors in the BLA contribute to anxiogenesis and fear-memory formation. We tested for rapid glucocorticoid-induced endocannabinoid regulation of synaptic inhibition in the rat BLA. Glucocorticoid application to amygdala slices elicited a rapid, nonreversible suppression of spontaneous, but not evoked, GABAergic synaptic currents in BLA principal neurons; the effect was also seen with a membrane-impermeant glucocorticoid, but not with intracellular glucocorticoid application, implicating a membrane-associated glucocorticoid receptor. The glucocorticoid suppression of GABA currents was not blocked by antagonists of nuclear corticosteroid receptors, or by inhibitors of gene transcription or protein synthesis, but was blocked by inhibiting postsynaptic G-protein activity, suggesting a postsynaptic nongenomic steroid signaling mechanism that stimulates the release of a retrograde messenger. The rapid glucocorticoid-induced suppression of inhibition was prevented by blocking CB1 receptors and 2-arachidonoylglycerol (2-AG) synthesis, and it was mimicked and occluded by CB1 receptor agonists, indicating it was mediated by the retrograde release of the endocannabinoid 2-AG. The rapid glucocorticoid effect in BLA neurons in vitro was occluded by prior in vivo acute stress-induced, or prior in vitro glucocorticoid-induced, release of endocannabinoid. Acute stress also caused an increase in anxiety-like behavior that was attenuated by blocking CB1 receptor activation and inhibiting 2-AG synthesis in the BLA. Together, these findings suggest that acute stress causes a long-lasting suppression of synaptic inhibition in BLA neurons via a membrane glucocorticoid receptor-induced release of 2-AG at GABA synapses, which contributes to stress-induced anxiogenesis. SIGNIFICANCE STATEMENT We provide a cellular mechanism in the basolateral amygdala (BLA) for the rapid stress regulation of anxiogenesis in rats. We demonstrate a nongenomic glucocorticoid induction of long-lasting suppression of synaptic inhibition that is mediated by retrograde endocannabinoid release at GABA synapses. The rapid glucocorticoid-induced endocannabinoid suppression of synaptic inhibition is initiated by a membrane-associated glucocorticoid receptor in BLA principal neurons. We show that acute stress increases anxiety-like behavior via an endocannabinoid-dependent mechanism centered in the BLA. The stress-induced endocannabinoid modulation of synaptic transmission in the BLA contributes, therefore, to the stress regulation of anxiety, and may play a role in anxiety disorders of the amygdala.
Collapse
|
50
|
Amygdalar Endothelin-1 Regulates Pyramidal Neuron Excitability and Affects Anxiety. Sci Rep 2017; 7:2316. [PMID: 28539637 PMCID: PMC5443782 DOI: 10.1038/s41598-017-02583-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/13/2017] [Indexed: 11/24/2022] Open
Abstract
An abnormal neuronal activity in the amygdala is involved in the pathogenesis of anxiety disorders. However, little is known about the mechanisms. High-anxiety mice and low-anxiety mice, representing the innate extremes of anxiety-related behaviors, were first grouped according to their anxiety levels in the elevated plus maze test. We found that the mRNA for endothelin-1 (ET1) and ET1 B-type receptors (ETBRs) in the amygdala was down-regulated in high-anxiety mice compared with low-anxiety mice. Knocking down basolateral amygdala (BLA) ET1 expression enhanced anxiety-like behaviors, whereas over-expressing ETBRs, but not A-type receptors (ETARs), had an anxiolytic effect. The combined down-regulation of ETBR and ET1 had no additional anxiogenic effect compared to knocking down the ETBR gene alone, suggesting that BLA ET1 acts through ETBRs to regulate anxiety-like behaviors. To explore the mechanism underlying this phenomenon further, we verified that most of the ET1 and the ET1 receptors in the BLA were expressed in pyramidal neurons. The ET1–ETBR signaling pathway decreased the firing frequencies and threshold currents for the action potentials of BLA pyramidal neurons but did not alter BLA synaptic neurotransmission. Together, these results indicate that amygdalar ET1-ETBR signaling could attenuate anxiety-like behaviors by directly decreasing the excitability of glutamatergic neurons.
Collapse
|