1
|
Wan D, Lu T, Li C, Hu C. Glucocorticoids Rapidly Modulate Ca V1.2-Mediated Calcium Signals through Kv2.1 Channel Clusters in Hippocampal Neurons. J Neurosci 2024; 44:e0179242024. [PMID: 39299804 DOI: 10.1523/jneurosci.0179-24.2024] [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: 01/25/2024] [Revised: 08/15/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
The precise regulation of Ca2+ signals plays a crucial role in the physiological functions of neurons. Here, we investigated the rapid effect of glucocorticoids on Ca2+ signals in cultured hippocampal neurons from both female and male rats. In cultured hippocampal neurons, glucocorticoids inhibited the spontaneous somatic Ca2+ spikes generated by Kv2.1-organized Ca2+ microdomains. Furthermore, glucocorticoids rapidly reduced the cell surface expressions of Kv2.1 and CaV1.2 channels in hippocampal neurons. In HEK293 cells transfected with Kv2.1 alone, glucocorticoids significantly reduced the surface expression of Kv2.1 with little effect on K+ currents. In HEK293 cells transfected with CaV1.2 alone, glucocorticoids inhibited CaV1.2 currents but had no effect on the cell surface expression of CaV1.2. Notably, in the presence of wild-type Kv2.1, glucocorticoids caused a decrease in the surface expression of CaV1.2 channels in HEK293 cells. However, this effect was not observed in the presence of nonclustering Kv2.1S586A mutant channels. Live-cell imaging showed that glucocorticoids rapidly decreased Kv2.1 clusters on the plasma membrane. Correspondingly, Western blot results indicated a significant increase in the cytoplasmic level of Kv2.1, suggesting the endocytosis of Kv2.1 clusters. Glucocorticoids rapidly decreased the intracellular cAMP concentration and the phosphorylation level of PKA in hippocampal neurons. The PKA inhibitor H89 mimicked the effect of glucocorticoids on Kv2.1, while the PKA agonist forskolin abrogated the effect. In conclusion, glucocorticoids rapidly suppress CaV1.2-mediated Ca2+ signals in hippocampal neurons by promoting the endocytosis of Kv2.1 channel clusters through reducing PKA activity.
Collapse
Affiliation(s)
- Di Wan
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Tongchuang Lu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Chenyang Li
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Changlong Hu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China,
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| |
Collapse
|
2
|
Girotti M, Bulin SE, Carreno FR. Effects of chronic stress on cognitive function - From neurobiology to intervention. Neurobiol Stress 2024; 33:100670. [PMID: 39295772 PMCID: PMC11407068 DOI: 10.1016/j.ynstr.2024.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
Exposure to chronic stress contributes considerably to the development of cognitive impairments in psychiatric disorders such as depression, generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), post-traumatic stress disorder (PTSD), and addictive behavior. Unfortunately, unlike mood-related symptoms, cognitive impairments are not effectively treated by available therapies, a situation in part resulting from a still incomplete knowledge of the neurobiological substrates that underly cognitive domains and the difficulty in generating interventions that are both efficacious and safe. In this review, we will present an overview of the cognitive domains affected by stress with a specific focus on cognitive flexibility, behavioral inhibition, and working memory. We will then consider the effects of stress on neuronal correlates of cognitive function and the factors which may modulate the interaction of stress and cognition. Finally, we will discuss intervention strategies for treatment of stress-related disorders and gaps in knowledge with emerging new treatments under development. Understanding how cognitive impairment occurs during exposure to chronic stress is crucial to make progress towards the development of new and effective therapeutic approaches.
Collapse
Affiliation(s)
| | - Sarah E. Bulin
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr., San Antonio, TX, 78229, USA
| | - Flavia R. Carreno
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr., San Antonio, TX, 78229, USA
| |
Collapse
|
3
|
Joëls M, Karst H, Tasker JG. The emerging role of rapid corticosteroid actions on excitatory and inhibitory synaptic signaling in the brain. Front Neuroendocrinol 2024; 74:101146. [PMID: 39004314 DOI: 10.1016/j.yfrne.2024.101146] [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: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Over the past two decades, there has been increasing evidence for the importance of rapid-onset actions of corticosteroid hormones in the brain. Here, we highlight the distinct rapid corticosteroid actions that regulate excitatory and inhibitory synaptic transmission in the hypothalamus, the hippocampus, basolateral amygdala, and prefrontal cortex. The receptors that mediate rapid corticosteroid actions are located at or close to the plasma membrane, though many of the receptor characteristics remain unresolved. Rapid-onset corticosteroid effects play a role in fast neuroendocrine feedback as well as in higher brain functions, including increased aggression and anxiety, and impaired memory retrieval. The rapid non-genomic corticosteroid actions precede and complement slow-onset, long-lasting transcriptional actions of the steroids. Both rapid and slow corticosteroid actions appear to be indispensable to adapt to a continuously changing environment, and their imbalance can increase an individual's susceptibility to psychopathology.
Collapse
Affiliation(s)
- Marian Joëls
- University Medical Center Groningen, University of Groningen, the Netherlands; University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Henk Karst
- University Medical Center Utrecht, Utrecht University, the Netherlands; SILS-CNS. University of Amsterdam, the Netherlands.
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, and Southeast Louisiana Veterans Affairs Healthcare System, New Orleans, USA.
| |
Collapse
|
4
|
Uyar E, Erdinç M, Kelle İ, Erdinç L, Şeker U, Nergiz Y. The Involvement of the Serotonergic System in Ketamine and Fluoxetine Combination-induced Cognitive Impairments in Mice. Eurasian J Med 2024; 56:102-107. [PMID: 39128082 PMCID: PMC11332274 DOI: 10.5152/eurasianjmed.2024.23219] [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: 08/11/2023] [Accepted: 04/15/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Glutamatergic N-methyl-D-aspartate (NMDA) receptors play vital roles in memory formation. Changes in the activity of these receptors influence memory processes. Ketamine is a noncompetitive NMDA receptor antagonist drug with promising mood-altering and pain-reducing effects in low doses. These effects are believed to be related to altered serotonergic transmission. METHODS The present study investigated the involvement of the serotonergic system in low-dose ketamine administrations' effects on memory acquisition, consolidation, and retrieval processes. Sixty-four male BALB/c mice were used in this experiment and separated into 8t groups. Mice were treated subchronically with a selective serotonin reuptake inhibitor, fluoxetine, and a serotonin depletion agent, p-chlorophenylalanine (pCPA). A serotonin antagonist, methiothepin, and ketamine were acutely administered 60 minutes before or after the behavioral tests. A passive avoidance (PA) test measured emotional memory acquisition, consolidation, and retrieval processes. Hippocampi malondialdehyde (MDA) levels were analyzed, and histopathological examinations were performed. RESULTS Ketamine alone did not significantly affect memory encoding processes in the PA test, while the ketamine-fluoxetine combination disrupted memory consolidation. Fluoxetine negatively affected the memory acquisition process, which was normalized during the consolidation and retrieval trials. Drug applications did not significantly alter hippocampal MDA levels. In all ketamine-applied groups, histopathologic alterations were evident. CONCLUSION Low-dose ketamine administration induces neurodegeneration, and it also impairs memory functions when combined with fluoxetine, indicating increased serotonergic transmission may be involved in the memory-impairing and neurotoxic effects of ketamine.
Collapse
Affiliation(s)
- Emre Uyar
- Department of Medical Pharmacology, Uskudar University Faculty of Medicine, Istanbul, Türkiye
| | - Meral Erdinç
- Department of Medical Pharmacology, Uskudar University Faculty of Medicine, Istanbul, Türkiye
| | - İlker Kelle
- Department of Medical Pharmacology, Dicle University Faculty of Medicine, Diyarbakir, Türkiye
| | - Levent Erdinç
- Department of Biochemistry, Dicle University Faculty of Medicine, Diyarbakir, Türkiye
| | - Uğur Şeker
- Department of Histology and Embryology, Mardin Artuklu University Faculty of Medicine, Mardin, Türkiye
| | - Yusuf Nergiz
- Department of Histology and Embryology, Dicle University Faculty of Medicine, Diyarbakir, Türkiye
| |
Collapse
|
5
|
Miyako K, Kajitani N, Koga Y, Takizawa H, Boku S, Takebayashi M. Identification of the antidepressant effect of electroconvulsive stimulation-related genes in hippocampal astrocyte. J Psychiatr Res 2024; 170:318-327. [PMID: 38194849 DOI: 10.1016/j.jpsychires.2024.01.004] [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: 10/25/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/11/2024]
Abstract
Major depressive disorder (MDD) remains a significant global health concern, with limited and slow efficacy of existing antidepressants. Electroconvulsive therapy (ECT) has superior and immediate efficacy for MDD, but its action mechanism remains elusive. Therefore, the elucidation of the action mechanism of ECT is expected to lead to the development of novel antidepressants with superior and immediate efficacy. Recent studies suggest a potential role of hippocampal astrocyte in MDD and ECT. Hence, we investigated antidepressant effect of electroconvulsive stimulation (ECS), an animal model of ECT, -related genes in hippocampal astrocyte with a mouse model of MDD, in which corticosterone (CORT)-induced depression-like behaviors were recovered by ECS. In this model, both of CORT-induced depression-like behaviors and the reduction of hippocampal astrocyte were recovered by ECS. Following it, astrocytes were isolated from the hippocampus of this model and RNA-seq was performed with these isolated astrocytes. Interestingly, gene expression patterns altered by CORT were reversed by ECS. Additionally, cell proliferation-related signaling pathways were inhibited by CORT and recovered by ECS. Finally, serum and glucocorticoid kinase-1 (SGK1), a multi-functional protein kinase, was identified as a candidate gene reciprocally regulated by CORT and ECS in hippocampal astrocyte. Our findings suggest a potential role of SGK1 in the antidepressant effect of ECS via the regulation of the proliferation of astrocyte and provide new insights into the involvement of hippocampal astrocyte in MDD and ECT. Targeting SGK1 may offer a novel approach to the development of new antidepressants which can replicate superior and immediate efficacy of ECT.
Collapse
Affiliation(s)
- Kotaro Miyako
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoto Kajitani
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yusaku Koga
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuken Boku
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Minoru Takebayashi
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
6
|
Mamelak M. Depression and the Glutamate/GABA-Glutamine Cycle. Curr Neuropharmacol 2024; 23:75-84. [PMID: 39150032 PMCID: PMC11519819 DOI: 10.2174/1570159x22666240815120244] [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: 01/22/2024] [Revised: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 08/17/2024] Open
Abstract
Many features of major depressive disorder are mirrored in rodent models of psychological stress. These models have been used to examine the relationship between the activation of the hypothalamic- pituitary axis in response to stress, the development of oxidative stress and neuroinflammation, the dominance of cholinergic neurotransmission and the associated increase in REM sleep pressure. Rodent models have also provided valuable insights into the impairment of glycolysis and brain glucose utilization by the brain under stress, the resulting decrease in brain energy production and the reduction in glutamate/GABA-glutamine cycling. The rapidly acting antidepressants, scopolamine, ketamine and ECT, all raise extracellular glutamate and scopolamine and ketamine have specifically been shown to increase glutamate/GABA-glutamine cycling in men and rodents with corresponding short-term relief of depression. The nightly use of gammahydroxybutyrate (GHB) may achieve more permanent results and may even act prophylactically to prevent the development or recurrence of depression. GHB is a GABAB agonist and restores the normal balance between cholinergic and monoaminergic neurotransmission by inhibiting cholinergic neurotransmission. It relieves REM sleep pressure. GHB's metabolism generates NADPH, a key antioxidant cofactor. Its metabolism also generates succinate, the tricarboxylic acid cycle intermediate, to provide energy to the cell and to synthesize glutamate. In both animals and man, GHB increases the level of brain glutamate.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Perica MI, Luna B. Impact of stress on excitatory and inhibitory markers of adolescent cognitive critical period plasticity. Neurosci Biobehav Rev 2023; 153:105378. [PMID: 37643681 PMCID: PMC10591935 DOI: 10.1016/j.neubiorev.2023.105378] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Adolescence is a time of significant neurocognitive development. Prolonged maturation of prefrontal cortex (PFC) through adolescence has been found to support improvements in executive function. Changes in excitatory and inhibitory mechanisms of critical period plasticity have been found to be present in the PFC through adolescence, suggesting that environment may have a greater effect on development during this time. Stress is one factor known to affect neurodevelopment increasing risk for psychopathology. However, less is known about how stress experienced during adolescence could affect adolescent-specific critical period plasticity mechanisms and cognitive outcomes. In this review, we synthesize findings from human and animal literatures looking at the experience of stress during adolescence on cognition and frontal excitatory and inhibitory neural activity. Studies indicate enhancing effects of acute stress on cognition and excitation within specific contexts, while chronic stress generally dampens excitatory and inhibitory processes and impairs cognition. We propose a model of how stress could affect frontal critical period plasticity, thus potentially altering neurodevelopmental trajectories that could lead to risk for psychopathology.
Collapse
Affiliation(s)
- Maria I Perica
- Department of Psychology, University of Pittsburgh, PA, USA.
| | - Beatriz Luna
- Department of Psychology, University of Pittsburgh, PA, USA
| |
Collapse
|
8
|
Koszałka A, Lustyk K, Pytka K. Sex-dependent differences in animal cognition. Neurosci Biobehav Rev 2023; 153:105374. [PMID: 37634555 DOI: 10.1016/j.neubiorev.2023.105374] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
The differences in cognitive processes driven by biological sex are the issues that have gotten growing attention recently. Considering the increasing population suffering from various cognitive impairments and the development of therapeutic strategies, it is essential that we recognize the mechanisms responsible for discrepancies observed in male and female learning and memory functions. In this review, we discuss recent reports from preclinical studies on rodents regarding selected cognitive domains to explore the state of knowledge on sex-dependent differences and point to challenges encountered during such research. We focus on spatial, recognition, and emotional memory, as well as on executive functions, such as attention, cognitive flexibility, and working memory. This review will help to acknowledge sex-related differences in cognition and indicate some fields that lack sufficient data.
Collapse
Affiliation(s)
- Aleksandra Koszałka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688 Krakow, Poland; Jagiellonian University Medical College, Doctoral School of Medical and Health Sciences, Św. Łazarza 16, 31-530 Krakow, Poland
| | - Klaudia Lustyk
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
9
|
Karst H, Joëls M. Corticosterone rapidly reduces glutamatergic but not GABAergic transmission in the infralimbic prefrontal cortex of male mice. Steroids 2023; 198:109283. [PMID: 37487816 DOI: 10.1016/j.steroids.2023.109283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
Rapid non-genomic effects of corticosteroid hormones, affecting glutamatergic and GABAergic transmission, have been described for many limbic structures in the rodent brain. These rapid effects appear to be region specific. It is not always clear which (or even whether) corticosteroid receptor -the glucocorticoid receptor (GR) or mineralocorticoid receptor (MR)- initiate these rapid effects. In the hippocampus and amygdala membrane-associated MR, but also membrane-associated GR (in amygdala), are involved. Other studies indicate that the rapid modulation may be induced by transactivation of kinases, or other receptors, like the G-protein coupled estrogen receptor (GPER) which was recently found to bind the mineralocorticoid aldosterone. In the current study we explored, in young adult male C57Bl6 mice, possible rapid effects of corticosterone on layer 2/3 infralimbic-prefrontal cortex (IL-PFC) neurons. We show that corticosterone, via non-genomic MR activation, reduces the mEPSC -but does not affect mIPSC- frequency; we observed no effect on mEPSC or mIPSC amplitude. As a result, overall spontaneous activity in the IL-PFC is suppressed. A potential role of GPER cannot be excluded, since G-15, an antagonist of GPER, also prevented the rapid effects of corticosterone.
Collapse
Affiliation(s)
- Henk Karst
- Dept. Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands; University of Amsterdam, SILS-CNS, Amsterdam, the Netherlands.
| | - Marian Joëls
- Dept. Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
10
|
dos-Santos RC, Sweeten BLW, Stelly CE, Tasker JG. The Neuroendocrine Impact of Acute Stress on Synaptic Plasticity. Endocrinology 2023; 164:bqad149. [PMID: 37788632 PMCID: PMC11046011 DOI: 10.1210/endocr/bqad149] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023]
Abstract
Stress induces changes in nervous system function on different signaling levels, from molecular signaling to synaptic transmission to neural circuits to behavior-and on different time scales, from rapid onset and transient to delayed and long-lasting. The principal effectors of stress plasticity are glucocorticoids, steroid hormones that act with a broad range of signaling competency due to the expression of multiple nuclear and membrane receptor subtypes in virtually every tissue of the organism. Glucocorticoid and mineralocorticoid receptors are localized to each of the cellular compartments of the receptor-expressing cells-the membrane, cytosol, and nucleus. In this review, we cover the neuroendocrine effects of stress, focusing mainly on the rapid actions of acute stress-induced glucocorticoids that effect changes in synaptic transmission and neuronal excitability by modulating synaptic and intrinsic neuronal properties via activation of presumed membrane glucocorticoid and mineralocorticoid receptors. We describe the synaptic plasticity that occurs in 4 stress-associated brain structures, the hypothalamus, hippocampus, amygdala, and prefrontal cortex, in response to single or short-term stress exposure. The rapid transformative impact of glucocorticoids makes this stress signal a particularly potent effector of acute neuronal plasticity.
Collapse
Affiliation(s)
- Raoni Conceição dos-Santos
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Brook L W Sweeten
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Claire E Stelly
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
11
|
Hassanshahi A, Janahmadi M, Razavinasab M, Ranjbar H, Hosseinmardi N, Behzadi G, Kohlmeier KA, Ilaghi M, Shabani M. Preventive putative effect of agmatine on cognitive and molecular outcomes in ventral tegmental area of male offspring following physical and psychological prenatal stress. Dev Psychobiol 2023; 65:e22410. [PMID: 37607891 DOI: 10.1002/dev.22410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 08/24/2023]
Abstract
Prenatal stress (PS) results from a maternal experience of stressful events during pregnancy, which has been associated with an increased risk of behavioral disorders including substance abuse and anxiety in the offspring. PS is known to result in heightened dopamine release in the ventral tegmental area (VTA), in part through the effects of corticotropin-releasing hormone, which directly excites dopaminergic cells. It has recently been suggested that agmatine plays a role in modulating anxiety-like behaviors. In this study, we investigated whether agmatine could reduce negative cognitive outcomes in male mice prenatally exposed to psychological/physical stress, and whether this could be associated with molecular changes in VTA. Agmatine (37.5 mg/kg) was administrated 30 min prior to PS induction in pregnant Swiss mice. Male offspring were evaluated in a series of behavioral and molecular assays. Findings demonstrated that agmatine reduced the impairment in locomotor activity induced by both psychological and physical PS. Agmatine also decreased heightened conditioned place preference to morphine seen in PS offspring. Moreover, agmatine ameliorated the anxiety-like behavior and drug-seeking behavior induced by PS in the male offspring. Molecular effects were seen in VTA as the enhanced brain-derived neurotrophic factor (BDNF) induced by PS in the VTA was reduced by agmatine. Behavioral tests indicate that agmatine exerts a protective effect on PS-induced impairments in male offspring, which could be due in part to agmatine-associated molecular alterations in the VTA. Taken together, our data suggest that prenatal treatment with agmatine exerts protective effect against negative consequences of PS on the development of affective circuits in the offspring.
Collapse
Affiliation(s)
- Amin Hassanshahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moazamehosadat Razavinasab
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Hoda Ranjbar
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Narges Hosseinmardi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mehran Ilaghi
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Kim EJ, Kim JJ. Neurocognitive effects of stress: a metaparadigm perspective. Mol Psychiatry 2023; 28:2750-2763. [PMID: 36759545 PMCID: PMC9909677 DOI: 10.1038/s41380-023-01986-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Stressful experiences, both physical and psychological, that are overwhelming (i.e., inescapable and unpredictable), can measurably affect subsequent neuronal properties and cognitive functioning of the hippocampus. At the cellular level, stress has been shown to alter hippocampal synaptic plasticity, spike and local field potential activity, dendritic morphology, neurogenesis, and neurodegeneration. At the behavioral level, stress has been found to impair learning and memory for declarative (or explicit) tasks that are based on cognition, such as verbal recall memory in humans and spatial memory in rodents, while facilitating those that are based on emotion, such as differential fear conditioning in humans and contextual fear conditioning in rodents. These vertically related alterations in the hippocampus, procedurally observed after subjects have undergone stress, are generally believed to be mediated by recurrently elevated circulating hypothalamic-pituitary-adrenal (HPA) axis effector hormones, glucocorticoids, directly acting on hippocampal neurons densely populated with corticosteroid receptors. The main purposes of this review are to (i) provide a synopsis of the neurocognitive effects of stress in a historical context that led to the contemporary HPA axis dogma of basic and translational stress research, (ii) critically reappraise the necessity and sufficiency of the glucocorticoid hypothesis of stress, and (iii) suggest an alternative metaparadigm approach to monitor and manipulate the progression of stress effects at the neural coding level. Real-time analyses can reveal neural activity markers of stress in the hippocampus that can be used to extrapolate neurocognitive effects across a range of stress paradigms (i.e., resolve scaling and dichotomous memory effects issues) and understand individual differences, thereby providing a novel neurophysiological scaffold for advancing future stress research.
Collapse
Affiliation(s)
- Eun Joo Kim
- Department of Psychology, University of Washington, Seattle, WA, 98195, USA
- School of Psychology, Korea University, Seoul, 02841, Republic of Korea
| | - Jeansok J Kim
- Department of Psychology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
13
|
León BE, Peyton L, Essa H, Wieden T, Marion N, Childers WE, Abou-Gharbia M, Choi DS. A novel monobactam lacking antimicrobial activity, MC-100093, reduces sex-specific ethanol preference and depressive-like behaviors in mice. Neuropharmacology 2023; 232:109515. [PMID: 37001726 PMCID: PMC10144181 DOI: 10.1016/j.neuropharm.2023.109515] [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: 10/19/2022] [Revised: 02/24/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Several β-lactam derivatives upregulate astrocytic glutamate transporter type 1expression and are known to improve measures in models of mood and alcohol use disorders (AUD) through normalizing glutamatergic states. However, long-term, and high doses of β-lactams may cause adverse side effects for treating mood disorders and AUD. Studies suggest that MC-100093, a novel β-lactam lacking antimicrobial activity, rescues GLT1 expression. Thus, we sought to investigate whether MC-100093 improves affective behaviors and reduces voluntary ethanol drinking. We intraperitoneally administered MC-100093 (50 mg/kg) or vehicle once per day to C57BL/6J male and female mice (8-10 weeks old) over 6 days. We employed the open field test and the elevated plus maze to examine the effect of MC-100093 on anxiety-like behaviors. We assayed MC-100093's effects on depressive-like behaviors using the tail suspension and forced swim tests. Next, utilizing a separate cohort of male and female C57BL6 mice, we assessed the effects MC100093 treatment on voluntary ethanol drinking utilizing the 2-bottle choice continuous access drinking paradigm. After screening and selecting high-drinking mice, we systematically administered MC-100093 (50 mg/kg) or vehicle to the high-drinking mice over 6 days. Overall, we found that MC-100093 treatment resulted in sex-specific pharmacological effects with female mice displaying reduced innate depressive-like behaviors during the tail suspension and force swim testing juxtaposed with male treated mice who displayed no changes in tail suspension and a paradoxical increased depressive-like behavior during the forced swim testing. Additionally, we found that MC100093 treatment reduced female preference for 10% EtOH during the 2-bottle choice continuous access drinking with no effects of MC100093 treatment detected in male mice. Overall, this data suggests sex-specific regulation of innate depressive-like behavior and voluntary EtOH drinking by MC100093 treatment. Western blot analysis of the medial prefrontal cortex and hippocampus revealed no changes in male or female GLT1 protein abundance relative to GAPDH.
Collapse
Affiliation(s)
- Brandon Emanuel León
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Regenerative Sciences Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Hesham Essa
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Tia Wieden
- Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA
| | - Nicole Marion
- Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA
| | - Wayne E Childers
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, 19140, USA
| | - Magid Abou-Gharbia
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, 19140, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, 59905, USA.
| |
Collapse
|
14
|
Guo LK, Su Y, Zhang YYN, Yu H, Lu Z, Li WQ, Yang YF, Xiao X, Yan H, Lu TL, Li J, Liao YD, Kang ZW, Wang LF, Li Y, Li M, Liu B, Huang HL, Lv LX, Yao Y, Tan YL, Breen G, Everall I, Wang HX, Huang Z, Zhang D, Yue WH. Prediction of treatment response to antipsychotic drugs for precision medicine approach to schizophrenia: randomized trials and multiomics analysis. Mil Med Res 2023; 10:24. [PMID: 37269009 DOI: 10.1186/s40779-023-00459-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/05/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Choosing the appropriate antipsychotic drug (APD) treatment for patients with schizophrenia (SCZ) can be challenging, as the treatment response to APD is highly variable and difficult to predict due to the lack of effective biomarkers. Previous studies have indicated the association between treatment response and genetic and epigenetic factors, but no effective biomarkers have been identified. Hence, further research is imperative to enhance precision medicine in SCZ treatment. METHODS Participants with SCZ were recruited from two randomized trials. The discovery cohort was recruited from the CAPOC trial (n = 2307) involved 6 weeks of treatment and equally randomized the participants to the Olanzapine, Risperidone, Quetiapine, Aripiprazole, Ziprasidone, and Haloperidol/Perphenazine (subsequently equally assigned to one or the other) groups. The external validation cohort was recruited from the CAPEC trial (n = 1379), which involved 8 weeks of treatment and equally randomized the participants to the Olanzapine, Risperidone, and Aripiprazole groups. Additionally, healthy controls (n = 275) from the local community were utilized as a genetic/epigenetic reference. The genetic and epigenetic (DNA methylation) risks of SCZ were assessed using the polygenic risk score (PRS) and polymethylation score, respectively. The study also examined the genetic-epigenetic interactions with treatment response through differential methylation analysis, methylation quantitative trait loci, colocalization, and promoter-anchored chromatin interaction. Machine learning was used to develop a prediction model for treatment response, which was evaluated for accuracy and clinical benefit using the area under curve (AUC) for classification, R2 for regression, and decision curve analysis. RESULTS Six risk genes for SCZ (LINC01795, DDHD2, SBNO1, KCNG2, SEMA7A, and RUFY1) involved in cortical morphology were identified as having a genetic-epigenetic interaction associated with treatment response. The developed and externally validated prediction model, which incorporated clinical information, PRS, genetic risk score (GRS), and proxy methylation level (proxyDNAm), demonstrated positive benefits for a wide range of patients receiving different APDs, regardless of sex [discovery cohort: AUC = 0.874 (95% CI 0.867-0.881), R2 = 0.478; external validation cohort: AUC = 0.851 (95% CI 0.841-0.861), R2 = 0.507]. CONCLUSIONS This study presents a promising precision medicine approach to evaluate treatment response, which has the potential to aid clinicians in making informed decisions about APD treatment for patients with SCZ. Trial registration Chinese Clinical Trial Registry ( https://www.chictr.org.cn/ ), 18. Aug 2009 retrospectively registered: CAPOC-ChiCTR-RNC-09000521 ( https://www.chictr.org.cn/showproj.aspx?proj=9014 ), CAPEC-ChiCTR-RNC-09000522 ( https://www.chictr.org.cn/showproj.aspx?proj=9013 ).
Collapse
Affiliation(s)
- Liang-Kun Guo
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Yi Su
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, 100096, China
| | - Yu-Ya-Nan Zhang
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Hao Yu
- Department of Psychiatry, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhe Lu
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Wen-Qiang Li
- Henan Key Lab of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435001, Henan, China
| | - Yong-Feng Yang
- Henan Key Lab of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435001, Henan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Hao Yan
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Tian-Lan Lu
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Jun Li
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Yun-Dan Liao
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Zhe-Wei Kang
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Li-Fang Wang
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Yue Li
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, WC2R 2LS, UK
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Hai-Liang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, 02141, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Lu-Xian Lv
- Henan Key Lab of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435001, Henan, China
| | - Yin Yao
- Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yun-Long Tan
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, 100096, China
| | - Gerome Breen
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, WC2R 2LS, UK
| | - Ian Everall
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, WC2R 2LS, UK
| | - Hong-Xing Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory for Neuroscience for Ministry of Education, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Dai Zhang
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China.
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| | - Wei-Hua Yue
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, 100191, China.
- NHC Key Laboratory of Mental Health and Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder (2018RU006), Chinese Academy of Medical Sciences, Beijing, 100191, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
15
|
Yang L, Shi LJ, Shen SY, Yang JY, Lv SS, Wang ZC, Huang Q, Xu WD, Yu J, Zhang YQ. Toward Antifragility: Social Defeat Stress Enhances Learning and Memory in Young Mice Via Hippocampal Synaptosome Associated Protein 25. Psychol Sci 2023; 34:616-632. [PMID: 37040450 DOI: 10.1177/09567976231160098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023] Open
Abstract
Social adversity not only causes severe psychological diseases but also may improve people's ability to learn and grow. However, the beneficial effects of social adversity are often ignored. In this study, we investigated whether and how social adversity affects learning and memory in a mouse social defeat stress (SDS) model. A total of 652 mice were placed in experimental groups of six to 23 mice each. SDS enhanced spatial, novelty, and fear memory with increased synaptosome associated protein 25 (SNAP-25) level and dendritic spine density in hippocampal neurons among young but not middle-aged mice. Chemogenetic inhibition of hippocampal CaMK2A+ neurons blocked SDS-induced enhancement of learning or memory. Knockdown of SNAP-25 or blockade of N-methyl-D-aspartate (NMDA) receptor subunit GluN2B in the hippocampus prevented SDS-induced learning memory enhancement in an emotion-independent manner. These findings suggest that social adversity promotes learning and memory ability in youths and provide a neurobiological foundation for biopsychological antifragility.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Li-Jun Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Shi-Yu Shen
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University
| | - Jing-Yan Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Su-Su Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Zhe-Chen Wang
- Department of Psychology, School of Social Development and Public Policy, Fudan University
- School of Psychology, The University of Queensland
| | - Qian Huang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| | - Wen-Dong Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
- Department of Hand Surgery, Huashan Hospital, Fudan University
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University
| |
Collapse
|
16
|
Campos-Cardoso R, Novaes LS, Godoy LD, Dos Santos NB, Perfetto JG, Lazarini-Lopes W, Garcia-Cairasco N, Padovan CM, Munhoz CD. The resilience of adolescent male rats to acute stress-induced delayed anxiety is age-related and glucocorticoid release-dependent. Neuropharmacology 2023; 226:109385. [PMID: 36603798 DOI: 10.1016/j.neuropharm.2022.109385] [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: 06/06/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 01/03/2023]
Abstract
Studies investigated how stressful experiences modulate physiological and behavioral responses and the consequences of stress-induced corticosterone release in anxiety-like behavior. Adolescence is crucial to brain maturation, and several neurobiological changes in this period lead individuals to increased susceptibility or resilience to aversive situations. Despite the effects of stress in adults, information about adolescents' responses to acute stress is lacking. We aimed to understand how adolescence affects acute stress responses. Male adolescent rats (30 days old) were 2 h restrained, and anxiety-like behaviors were measured immediately or 10 days after stress in the elevated plus-maze (EPM) and the light-dark box (LDB) tests. To verify the importance of CORT modulation in stress-induced anxiety, another group of rats was treated, 30 min before restraint, with metyrapone to blunt the stress-induced CORT peak and tested immediately after stress. To show that stress effects on behavior were age-dependent, another set of rats was tested in two different periods - early adolescence (30 days old) and mid-adolescence (40 days old) and were treated or not with metyrapone before the stress session and tested immediately or ten days later in the LDB test. Only early adolescent male rats were resilient to delayed anxiety-like behavior in EPM and LDB tests. Metyrapone treatment increased the rats' exploration immediately and ten days after stress. These data suggest a specific age at which adolescent rats are resilient to the delayed effects of acute restraint stress and that the metyrapone treatment has long-term behavioral consequences.
Collapse
Affiliation(s)
- Rodrigo Campos-Cardoso
- Department of Pharmacology, Universidade de São Paulo, Instituto de Ciências Biomédicas, Brazil; Department of Neurosciences and Behavioral Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Leonardo Santana Novaes
- Department of Pharmacology, Universidade de São Paulo, Instituto de Ciências Biomédicas, Brazil
| | - Lívea Dornela Godoy
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Juliano Genaro Perfetto
- Department of Pharmacology, Universidade de São Paulo, Instituto de Ciências Biomédicas, Brazil
| | - Willian Lazarini-Lopes
- Department of Neurosciences and Behavioral Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Cláudia Maria Padovan
- Department of Neurosciences and Behavioral Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; Department of Psychology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Carolina Demarchi Munhoz
- Department of Pharmacology, Universidade de São Paulo, Instituto de Ciências Biomédicas, Brazil.
| |
Collapse
|
17
|
Karanikas E. The immune-stress/endocrine-redox-metabolic nature of psychosis' etiopathology; focus on the intersystemic pathways interactions. Neurosci Lett 2023; 794:137011. [PMID: 36513162 DOI: 10.1016/j.neulet.2022.137011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/26/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
The evidence supporting the involvement of a number of systems in the neurobiological etiopathology of psychosis has recently grown exponentially. Indeed, the focus of research has changed from measuring solely neurotransmitters to estimating parameters from fields like immunity, stress/endocrine, redox, and metabolism. Yet, little is known regarding the exact role of each one of these fields on the formation of not only the brain neuropathological substrate in psychosis but also the associated general systemic pathology, in terms of causality directions. Research has shown deviations in the levels and/or function of basic effector molecules of the aforementioned fields namely cytokines, pro-/anti- oxidants, glucocorticoids, catecholamines, glucose, and lipids metabolites as well as kynurenines, in psychosis. Yet the evidence regarding their impact on neurotransmitters is minimal and the findings concerning these systems' interactions in the psychotic context are even more dispersed. The present review aims to draw holistically the frame of the hitherto known "players" in the field of psychosis' cellular pathobiology, with a particular focus on their in-between interactions.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Department of Psychiatry, 424 General Military Hospital, Thessaloniki, Greece.
| |
Collapse
|
18
|
Witztum J, Singh A, Zhang R, Johnson M, Liston C. An automated platform for Assessing Working Memory and prefrontal circuit function. Neurobiol Stress 2023; 24:100518. [PMID: 36970451 PMCID: PMC10033752 DOI: 10.1016/j.ynstr.2023.100518] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 01/02/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Working memory is a process for actively maintaining and updating task-relevant information, despite interference from competing inputs, and is supported in part by sustained activity in prefrontal cortical pyramidal neurons and coordinated interactions with inhibitory interneurons, which may serve to regulate interference. Chronic stress has potent effects on working memory performance, possibly by interfering with these interactions or by disrupting long-range inputs from key upstream brain regions. Still, the mechanisms by which chronic stress disrupts working memory are not well understood, due in part to a need for scalable, easy-to-implement behavioral assays that are compatible with two-photon calcium imaging and other tools for recording from large populations of neurons. Here, we describe the development and validation of a platform that was designed specifically for automated, high-throughput assessments of working memory and simultaneous two-photon imaging in chronic stress studies. This platform is relatively inexpensive and easy to build; fully automated and scalable such that one investigator can test relatively large cohorts of animals concurrently; fully compatible with two-photon imaging, yet also designed to mitigate head-fixation stress; and can be easily adapted for other behavioral paradigms. Our validation data confirm that mice could be trained to perform a delayed response working memory task with relatively high-fidelity over the course of ∼15 days. Two-photon imaging data validate the feasibility of recording from large populations of cells during working memory tasks performance and characterizing their functional properties. Activity patterns in >70% of medial prefrontal cortical neurons were modulated by at least one task feature, and a majority of cells were engaged by multiple task features. We conclude with a brief literature review of the circuit mechanisms supporting working memory and their disruption in chronic stress states-highlighting directions for future research enabled by this platform.
Collapse
|
19
|
Su Y, Wang B, Ye J, Wang Y, Cui Y, Chen C, Ruan N, Hu Z, Li L, Liu H, Xie H. Dexmedetomidine improves the acute stress reactivity of male rat through interventions of serum- and glucocorticoid-inducible kinase 1 and nNOS in the bed nucleus of the stria terminalis. Biochem Biophys Res Commun 2023; 638:155-162. [PMID: 36459879 DOI: 10.1016/j.bbrc.2022.11.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 10/24/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Moderate acute stress responses are beneficial for adaptation and maintenance of homeostasis. Exposure of male rat to stress induces effects in the bed nucleus of the stria terminalis (BNST), for it can be activated by the same stimuli that induce activation of the hypothalamic-pituitary-adrenal axis. However, the underlying mechanism of the BNST on male stress reactivity remains unclear. In this study, we explored whether systematic administration of dexmedetomidine (DEXM) altered the acute stress reactivity through its effect on the BNST. Male Sprague-Dawley rats in the stress (STRE) group, DEXM group, and the DEXM + GSK-650394 (GSK, an antagonist of serum- and glucocorticoid-inducible kinase 1 (SGK1)) group, except those in the vehicle (VEH) group, underwent 1-h restraint plus water-immersion (RPWI) exposure. All the rats proceeded the open field test (OFT) 24 h before RPWI and 1 h after RPWI. After the second OFT, the rats received VEH, DEXM (75 μg/kg i.p.), or were pretreated with GSK (2 μM i.p.) 0.5 h ahead of DEXM respectively. The third OFT was conducted 6 h after drug administration and then the rats were sacrificed. The rats that experienced RPWI showed dramatically elevated serum corticosterone (CORT), multiplied neuronal nitric oxide synthase (nNOS) and SGK1 in the BNST, and terrible OFT behavior. We discovered when the nNOS and SGK1 were decreased in the rat BNST through DEXM treatment, the serum CORT was reduced and the OFT manifestation was ameliorated, whereas these were restrained by GSK application. Our results reveal that modest interventions to SGK1 and nNOS in the BNST improve the male rat reactivity to acute stress, and DEXM was one modulator of these effects.
Collapse
Affiliation(s)
- Ying Su
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Benfu Wang
- Department of Anesthesiology, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jianwen Ye
- Department of Anesthesiology, The Second Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Yixuan Wang
- Department of Anesthesiology, The Second Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Yanhua Cui
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunjiang Chen
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Naqi Ruan
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiyan Hu
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Li
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huacheng Liu
- Zhejiang Province Key Laboratory of Anesthesiology, Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Hong Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
20
|
Demin KA, Krotova NA, Ilyin NP, Galstyan DS, Kolesnikova TO, Strekalova T, de Abreu MS, Petersen EV, Zabegalov KN, Kalueff AV. Evolutionarily conserved gene expression patterns for affective disorders revealed using cross-species brain transcriptomic analyses in humans, rats and zebrafish. Sci Rep 2022; 12:20836. [PMID: 36460699 PMCID: PMC9718822 DOI: 10.1038/s41598-022-22688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Widespread, debilitating and often treatment-resistant, depression and other stress-related neuropsychiatric disorders represent an urgent unmet biomedical and societal problem. Although animal models of these disorders are commonly used to study stress pathogenesis, they are often difficult to translate across species into valuable and meaningful clinically relevant data. To address this problem, here we utilized several cross-species/cross-taxon approaches to identify potential evolutionarily conserved differentially expressed genes and their sets. We also assessed enrichment of these genes for transcription factors DNA-binding sites down- and up- stream from their genetic sequences. For this, we compared our own RNA-seq brain transcriptomic data obtained from chronically stressed rats and zebrafish with publicly available human transcriptomic data for patients with major depression and their respective healthy control groups. Utilizing these data from the three species, we next analyzed their differential gene expression, gene set enrichment and protein-protein interaction networks, combined with validated tools for data pooling. This approach allowed us to identify several key brain proteins (GRIA1, DLG1, CDH1, THRB, PLCG2, NGEF, IKZF1 and FEZF2) as promising, evolutionarily conserved and shared affective 'hub' protein targets, as well as to propose a novel gene set that may be used to further study affective pathogenesis. Overall, these approaches may advance cross-species brain transcriptomic analyses, and call for further cross-species studies into putative shared molecular mechanisms of affective pathogenesis.
Collapse
Affiliation(s)
- Konstantin A Demin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| | - Nataliya A Krotova
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | | | | | | | | | | | - Allan V Kalueff
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia.
- Institute of Neurosciences and Medicine, Novosibirsk, Russia.
- Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
21
|
Gómez de San José N, Goossens J, Al Shweiki MR, Halbgebauer S, Oeckl P, Steinacker P, Danzer KM, Graf H, Schönfeldt-Lecuona C, Belbin O, Lleó A, Vanmechelen E, Otto M. Glutamate receptor 4 as a fluid biomarker for the diagnosis of psychiatric disorders. J Psychiatr Res 2022; 156:390-397. [PMID: 36323141 DOI: 10.1016/j.jpsychires.2022.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/02/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
Psychiatric disorders are widely underreported diseases, especially in their early stages. So far, there is no fluid biomarker to confirm the diagnosis of these disorders. Proteomics data suggest the synaptic protein glutamate receptor 4 (GluR4), part of the AMPA receptor, as a potential diagnostic biomarker of major depressive disorder (MDD). A novel sandwich ELISA was established and analytically validated to detect GluR4 in cerebrospinal fluid (CSF) samples. A total of 85 subjects diagnosed with MDD (n = 36), bipolar disorder (BD, n = 12), schizophrenia (SCZ, n = 12) and neurological controls (CON, n = 25) were analysed. The data exhibited a significant correlation (r = 0.74; CI:0.62 to 0.82; p < 0.0001) with the antibody-free multiple reaction monitoring (MRM) mass spectrometry (MS) data. CSF GluR4 levels were lower in MDD (p < 0.002) and BD (p = 0.012) than in CON. Moreover, subjects with SCZ described a trend towards lower levels than CON (p = 0.13). The novel GluR4 ELISA may favour the clinical application of this protein as a potential diagnostic biomarker of psychiatric disorders and may facilitate the understanding of the pathophysiological mechanisms behind these disorders.
Collapse
Affiliation(s)
| | | | | | - Steffen Halbgebauer
- Department of Neurology, University of Ulm, 89075, Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany.
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, 89075, Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany.
| | - Petra Steinacker
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany.
| | - Karin M Danzer
- Department of Neurology, University of Ulm, 89075, Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany.
| | - Heiko Graf
- Department of Psychiatry and Psychotherapy III, University of Ulm, 89075, Ulm, Germany.
| | | | - Olivia Belbin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
| | | | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany.
| |
Collapse
|
22
|
Jiang Z, Chen C, Weiss GL, Fu X, Stelly CE, Sweeten BLW, Tirrell PS, Pursell I, Stevens CR, Fisher MO, Begley JC, Harrison LM, Tasker JG. Stress-induced glucocorticoid desensitizes adrenoreceptors to gate the neuroendocrine response to somatic stress in male mice. Cell Rep 2022; 41:111509. [PMID: 36261014 PMCID: PMC9635929 DOI: 10.1016/j.celrep.2022.111509] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/05/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Noradrenergic afferents to hypothalamic corticotropin releasing hormone (CRH) neurons provide a major excitatory drive to the hypothalamic-pituitary-adrenal (HPA) axis via α1 adrenoreceptor activation. Noradrenergic afferents are recruited preferentially by somatic, rather than psychological, stress stimuli. Stress-induced glucocorticoids feed back onto the hypothalamus to negatively regulate the HPA axis, providing a critical autoregulatory constraint that prevents glucocorticoid overexposure and neuropathology. Whether negative feedback mechanisms target stress modality-specific HPA activation is not known. Here, we describe a desensitization of the α1 adrenoreceptor activation of the HPA axis following acute stress in male mice that is mediated by rapid glucocorticoid regulation of adrenoreceptor trafficking in CRH neurons. Glucocorticoid-induced α1 receptor trafficking desensitizes the HPA axis to a somatic but not a psychological stressor. Our findings demonstrate a rapid glucocorticoid suppression of adrenergic signaling in CRH neurons that is specific to somatic stress activation, and they reveal a rapid, stress modality-selective glucocorticoid negative feedback mechanism.
Collapse
Affiliation(s)
- Zhiying Jiang
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Chun Chen
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Grant L Weiss
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Xin Fu
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Claire E Stelly
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Brook L W Sweeten
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Parker S Tirrell
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - India Pursell
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Carly R Stevens
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Marc O Fisher
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - John C Begley
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA
| | - Laura M Harrison
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Jeffrey G Tasker
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
23
|
Harrison LM, Tasker JG. Multiplexed Membrane Signaling by Glucocorticoids. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 26:100390. [PMID: 38075196 PMCID: PMC10703063 DOI: 10.1016/j.coemr.2022.100390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
Glucocorticoids exert pleiotropic effects either by a relatively slow mechanism involving binding to cytosolic/nuclear receptors and regulation of gene expression or by rapid activation of a putative membrane receptor and membrane signal transduction. Rapid glucocorticoid actions are initiated at the membrane and recruit intracellular signaling pathways that engage multiple downstream cellular targets, including lipid and gas intercellular messengers, membrane neurotransmitter receptor trafficking, nuclear glucocorticoid receptor activation and trafficking, and more. Thus, membrane glucocorticoid signaling diverges into a multiplexed array of signaling pathways to simultaneously regulate highly diverse cellular functions, giving these steroid hormones a broad range of rapid regulatory capabilities. In this review, we provide a brief overview of the growing body of knowledge of the cell signaling mechanisms of rapid glucocorticoid actions in the brain.
Collapse
Affiliation(s)
- Laura M Harrison
- Department of Cell and Molecular Biology, Tulane Brain Institute, Tulane University, New Orleans, LA 70118
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane Brain Institute, Tulane University, New Orleans, LA 70118
| |
Collapse
|
24
|
Harnett NG, Finegold KE, Lebois LAM, van Rooij SJH, Ely TD, Murty VP, Jovanovic T, Bruce SE, House SL, Beaudoin FL, An X, Zeng D, Neylan TC, Clifford GD, Linnstaedt SD, Germine LT, Bollen KA, Rauch SL, Haran JP, Storrow AB, Lewandowski C, Musey PI, Hendry PL, Sheikh S, Jones CW, Punches BE, Kurz MC, Swor RA, Hudak LA, Pascual JL, Seamon MJ, Harris E, Chang AM, Pearson C, Peak DA, Domeier RM, Rathlev NK, O'Neil BJ, Sergot P, Sanchez LD, Miller MW, Pietrzak RH, Joormann J, Barch DM, Pizzagalli DA, Sheridan JF, Harte SE, Elliott JM, Kessler RC, Koenen KC, McLean SA, Nickerson LD, Ressler KJ, Stevens JS. Structural covariance of the ventral visual stream predicts posttraumatic intrusion and nightmare symptoms: a multivariate data fusion analysis. Transl Psychiatry 2022; 12:321. [PMID: 35941117 PMCID: PMC9360028 DOI: 10.1038/s41398-022-02085-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 01/16/2023] Open
Abstract
Visual components of trauma memories are often vividly re-experienced by survivors with deleterious consequences for normal function. Neuroimaging research on trauma has primarily focused on threat-processing circuitry as core to trauma-related dysfunction. Conversely, limited attention has been given to visual circuitry which may be particularly relevant to posttraumatic stress disorder (PTSD). Prior work suggests that the ventral visual stream is directly related to the cognitive and affective disturbances observed in PTSD and may be predictive of later symptom expression. The present study used multimodal magnetic resonance imaging data (n = 278) collected two weeks after trauma exposure from the AURORA study, a longitudinal, multisite investigation of adverse posttraumatic neuropsychiatric sequelae. Indices of gray and white matter were combined using data fusion to identify a structural covariance network (SCN) of the ventral visual stream 2 weeks after trauma. Participant's loadings on the SCN were positively associated with both intrusion symptoms and intensity of nightmares. Further, SCN loadings moderated connectivity between a previously observed amygdala-hippocampal functional covariance network and the inferior temporal gyrus. Follow-up MRI data at 6 months showed an inverse relationship between SCN loadings and negative alterations in cognition in mood. Further, individuals who showed decreased strength of the SCN between 2 weeks and 6 months had generally higher PTSD symptom severity over time. The present findings highlight a role for structural integrity of the ventral visual stream in the development of PTSD. The ventral visual stream may be particularly important for the consolidation or retrieval of trauma memories and may contribute to efficient reactivation of visual components of the trauma memory, thereby exacerbating PTSD symptoms. Potentially chronic engagement of the network may lead to reduced structural integrity which becomes a risk factor for lasting PTSD symptoms.
Collapse
Affiliation(s)
- Nathaniel G Harnett
- Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | | | - Lauren A M Lebois
- Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Sanne J H van Rooij
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Timothy D Ely
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Vishnu P Murty
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI, USA
| | - Steven E Bruce
- Department of Psychological Sciences, University of Missouri - St. Louis, St. Louis, MO, USA
| | - Stacey L House
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Francesca L Beaudoin
- Department of Emergency Medicine & Department of Health Services, Policy, and Practice, The Alpert Medical School of Brown University, Rhode Island Hospital and The Miriam Hospital, Providence, RI, USA
| | - Xinming An
- Institute for Trauma Recovery, Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Donglin Zeng
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Thomas C Neylan
- Departments of Psychiatry and Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Gari D Clifford
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery, Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura T Germine
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Institute for Technology in Psychiatry, McLean Hospital, Belmont, MA, USA
- The Many Brains Project, Belmont, MA, USA
| | - Kenneth A Bollen
- Department of Psychology and Neuroscience & Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott L Rauch
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Institute for Technology in Psychiatry, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, McLean Hospital, Belmont, MA, USA
| | - John P Haran
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alan B Storrow
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Paul I Musey
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Phyllis L Hendry
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, USA
| | - Sophia Sheikh
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, USA
| | - Christopher W Jones
- Department of Emergency Medicine, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Brittany E Punches
- Department of Emergency Medicine, Ohio State University College of Medicine, Columbus, OH, USA
- Ohio State University College of Nursing, Columbus, OH, USA
| | - Michael C Kurz
- Department of Emergency Medicine, University of Alabama School of Medicine, Birmingham, AL, USA
- Department of Surgery, Division of Acute Care Surgery, University of Alabama School of Medicine, Birmingham, AL, USA
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert A Swor
- Department of Emergency Medicine, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Lauren A Hudak
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jose L Pascual
- Department of Surgery, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark J Seamon
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Anna M Chang
- Department of Emergency Medicine, Jefferson University Hospitals, Philadelphia, PA, USA
| | - Claire Pearson
- Department of Emergency Medicine, Wayne State University, Ascension St. John Hospital, Detroit, MI, USA
| | - David A Peak
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Robert M Domeier
- Department of Emergency Medicine, Saint Joseph Mercy Hospital, Ypsilanti, MI, USA
| | - Niels K Rathlev
- Department of Emergency Medicine, University of Massachusetts Medical School-Baystate, Springfield, MA, USA
| | - Brian J O'Neil
- Department of Emergency Medicine, Wayne State University, Detroit Receiving Hospital, Detroit, MI, USA
| | - Paulina Sergot
- Department of Emergency Medicine, McGovern Medical School, University of Texas Health, Houston, TX, USA
| | - Leon D Sanchez
- Department of Emergency Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Emergency Medicine, Harvard Medical School, Boston, MA, USA
| | - Mark W Miller
- National Center for PTSD, Behavioral Science Division, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Robert H Pietrzak
- National Center for PTSD, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Jutta Joormann
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Deanna M Barch
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Diego A Pizzagalli
- Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - John F Sheridan
- Division of Biosciences, Ohio State University College of Dentistry, Columbus, OH, USA
- Institute for Behavioral Medicine Research, OSU Wexner Medical Center, Columbus, OH, USA
| | - Steven E Harte
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine-Rheumatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James M Elliott
- Kolling Institute, University of Sydney, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Northern Sydney Local Health District, New South Wales, Australia
- Physical Therapy & Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Samuel A McLean
- Department of Emergency Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute for Trauma Recovery, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lisa D Nickerson
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Imaging Center, McLean Hospital, Belmont, MA, USA
| | - Kerry J Ressler
- Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Jennifer S Stevens
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
25
|
Gonda X, Dome P, Erdelyi-Hamza B, Krause S, Elek LP, Sharma SR, Tarazi FI. Invisible wounds: Suturing the gap between the neurobiology, conventional and emerging therapies for posttraumatic stress disorder. Eur Neuropsychopharmacol 2022; 61:17-29. [PMID: 35716404 DOI: 10.1016/j.euroneuro.2022.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/26/2022]
Abstract
A sharp increase in the prevalence of neuropsychiatric disorders, including major depression, anxiety, substance use disorders and posttraumatic stress disorder (PTSD) has occurred due to the traumatic nature of the persisting COVID-19 global pandemic. PTSD is estimated to occur in up to 25% of individuals following exposure to acute or chronic trauma, and the pandemic has inflicted both forms of trauma on much of the population through both direct physiological attack as well as an inherent upheaval to our sense of safety. However, despite significant advances in our ability to define and apprehend the effects of traumatic events, the neurobiology and neuroanatomical circuitry of PTSD, one of the most severe consequences of traumatic exposure, remains poorly understood. Furthermore, the current psychotherapies or pharmacological options for treatment have limited efficacy, durability, and low adherence rates. Consequently, there is a great need to better understand the neurobiology and neuroanatomy of PTSD and develop novel therapies that extend beyond the current limited treatments. This review summarizes the neurobiological and neuroanatomical underpinnings of PTSD and discusses the conventional and emerging psychotherapies, pharmacological and combined psychopharmacological therapies, including the use of psychedelic-assisted psychotherapies and neuromodulatory interventions, for the improved treatment of PTSD and the potential for their wider applications in other neuropsychiatric disorders resulting from traumatic exposure.
Collapse
Affiliation(s)
- Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Hungary; NAP-2-SE New Antidepressant Target Research Group, Semmelweis University, Hungary; International Centre for Education and Research in Neuropsychiatry, Samara State Medical University, Russia.
| | - Peter Dome
- Department of Psychiatry and Psychotherapy, Semmelweis University, Hungary; National Institute of Mental Health, Neurology and Neurosurgery - Nyiro Gyula Hospital, Hungary
| | - Berta Erdelyi-Hamza
- Department of Psychiatry and Psychotherapy, Semmelweis University, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Hungary
| | - Sandor Krause
- National Institute of Mental Health, Neurology and Neurosurgery - Nyiro Gyula Hospital, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Hungary; Department of Pharmacodynamics, Semmelweis University, Hungary
| | - Livia Priyanka Elek
- Department of Psychiatry and Psychotherapy, Semmelweis University, Hungary; Department of Clinical Psychology, Semmelweis University, Hungary
| | - Samata R Sharma
- Department of Psychiatry, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Frank I Tarazi
- Department of Psychiatry and Neuroscience, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
26
|
Thomas R, Hernandez A, Benavides DR, Li W, Tan C, Umfress A, Plattner F, Chakraborti A, Pozzo-Miller L, Taylor SS, Bibb JA. Integrated regulation of PKA by fast and slow neurotransmission in the nucleus accumbens controls plasticity and stress responses. J Biol Chem 2022; 298:102245. [PMID: 35835216 PMCID: PMC9386499 DOI: 10.1016/j.jbc.2022.102245] [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: 04/19/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cortical glutamate and midbrain dopamine neurotransmission converge to mediate striatum-dependent behaviors, while maladaptations in striatal circuitry contribute to mental disorders. However, the crosstalk between glutamate and dopamine signaling has not been entirely elucidated. Here we uncover a molecular mechanism by which glutamatergic and dopaminergic signaling integrate to regulate cAMP-dependent protein kinase (PKA) via phosphorylation of the PKA regulatory subunit, RIIβ. Using a combination of biochemical, pharmacological, neurophysiological, and behavioral approaches, we find that glutamate-dependent reduction in cyclin-dependent kinase 5 (Cdk5)-dependent RIIβ phosphorylation alters the PKA holoenzyme autoinhibitory state to increase PKA signaling in response to dopamine. Furthermore, we show that disruption of RIIβ phosphorylation by Cdk5 enhances cortico-ventral striatal synaptic plasticity. In addition, we demonstrate that acute and chronic stress in rats inversely modulate RIIβ phosphorylation and ventral striatal infusion of a small interfering peptide that selectively targets RIIβ regulation by Cdk5 improves behavioral response to stress. We propose this new signaling mechanism integrating ventral striatal glutamate and dopamine neurotransmission is important to brain function, may contribute to neuropsychiatric conditions, and serves as a possible target for the development of novel therapeutics for stress-related disorders.
Collapse
Affiliation(s)
- Rachel Thomas
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adan Hernandez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, Santiago de Querétaro, Querétaro, México; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David R Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Li
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alan Umfress
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Florian Plattner
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ayanabha Chakraborti
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Susan S Taylor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - James A Bibb
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
27
|
Okura A, Inoue K, Sakuma E, Takase H, Ueki T, Mase M. SGK1 in Schwann cells is a potential molecular switch involved in axonal and glial regeneration during peripheral nerve injury. Biochem Biophys Res Commun 2022; 607:158-165. [DOI: 10.1016/j.bbrc.2022.03.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/23/2022] [Indexed: 12/29/2022]
|
28
|
Ventral hippocampal NMDA receptors mediate the effects of nicotine on stress-induced anxiety/exploratory behaviors in rats. Neurosci Lett 2022; 780:136649. [DOI: 10.1016/j.neulet.2022.136649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 03/19/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022]
|
29
|
Lee MT, Peng WH, Kan HW, Wu CC, Wang DW, Ho YC. Neurobiology of Depression: Chronic Stress Alters the Glutamatergic System in the Brain-Focusing on AMPA Receptor. Biomedicines 2022; 10:biomedicines10051005. [PMID: 35625742 PMCID: PMC9138646 DOI: 10.3390/biomedicines10051005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/25/2022] Open
Abstract
Major depressive disorder (MDD) is a common neuropsychiatric disorder affecting the mood and mental well-being. Its pathophysiology remains elusive due to the complexity and heterogeneity of this disorder that affects millions of individuals worldwide. Chronic stress is frequently cited as the one of the risk factors for MDD. To date, the conventional monoaminergic theory (serotonin, norepinephrine, and/or dopamine dysregulation) has received the most attention in the treatment of MDD, and all available classes of antidepressants target these monoaminergic systems. However, the contributions of other neurotransmitter systems in MDD have been widely reported. Emerging preclinical and clinical findings reveal that maladaptive glutamatergic neurotransmission might underlie the pathophysiology of MDD, thus revealing its critical role in the neurobiology of MDD and as the therapeutic target. Aiming beyond the monoaminergic hypothesis, studies of the neurobiological mechanisms underlying the stress-induced impairment of AMPA (a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-glutamatergic neurotransmission in the brain could provide novel insights for the development of a new generation of antidepressants without the detrimental side effects. Here, the authors reviewed the recent literature focusing on the role of AMPA-glutamatergic neurotransmission in stress-induced maladaptive responses in emotional and mood-associated brain regions, including the hippocampus, amygdala, prefrontal cortex, nucleus accumbens and periaqueductal gray.
Collapse
Affiliation(s)
- Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Wei-Hao Peng
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (W.-H.P.); (H.-W.K.)
| | - Hung-Wei Kan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (W.-H.P.); (H.-W.K.)
| | - Cheng-Chun Wu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-C.W.); (D.-W.W.)
| | - Deng-Wu Wang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-C.W.); (D.-W.W.)
- Department of Psychiatry, E-Da Hospital, Kaohsiung City 82445, Taiwan
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-C.W.); (D.-W.W.)
- Correspondence:
| |
Collapse
|
30
|
Sala N, Paoli C, Bonifacino T, Mingardi J, Schiavon E, La Via L, Milanese M, Tornese P, Datusalia AK, Rosa J, Facchinetti R, Frumento G, Carini G, Salerno Scarzella F, Scuderi C, Forti L, Barbon A, Bonanno G, Popoli M, Musazzi L. Acute Ketamine Facilitates Fear Memory Extinction in a Rat Model of PTSD Along With Restoring Glutamatergic Alterations and Dendritic Atrophy in the Prefrontal Cortex. Front Pharmacol 2022; 13:759626. [PMID: 35370690 PMCID: PMC8968915 DOI: 10.3389/fphar.2022.759626] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022] Open
Abstract
Stress represents a major risk factor for psychiatric disorders, including post-traumatic stress disorder (PTSD). Recently, we dissected the destabilizing effects of acute stress on the excitatory glutamate system in the prefrontal cortex (PFC). Here, we assessed the effects of single subanesthetic administration of ketamine (10 mg/kg) on glutamate transmission and dendritic arborization in the PFC of footshock (FS)-stressed rats, along with changes in depressive, anxious, and fear extinction behaviors. We found that ketamine, while inducing a mild increase of glutamate release in the PFC of naïve rats, blocked the acute stress-induced enhancement of glutamate release when administered 24 or 72 h before or 6 h after FS. Accordingly, the treatment with ketamine 6 h after FS also reduced the stress-dependent increase of spontaneous excitatory postsynaptic current (sEPSC) amplitude in prelimbic (PL)-PFC. At the same time, ketamine injection 6 h after FS was found to rescue apical dendritic retraction of pyramidal neurons induced by acute stress in PL-PFC and facilitated contextual fear extinction. These results show rapid effects of ketamine in animals subjected to acute FS, in line with previous studies suggesting a therapeutic action of the drug in PTSD models. Our data are consistent with a mechanism of ketamine involving re-establishment of synaptic homeostasis, through restoration of glutamate release, and structural remodeling of dendrites.
Collapse
Affiliation(s)
- Nathalie Sala
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Caterina Paoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.,School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Jessica Mingardi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emanuele Schiavon
- Department of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Paolo Tornese
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Ashok K Datusalia
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, India
| | - Jessica Rosa
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.,Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Roberta Facchinetti
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Giulia Frumento
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Giulia Carini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Lia Forti
- Department of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
31
|
Sanacora G, Yan Z, Popoli M. The stressed synapse 2.0: pathophysiological mechanisms in stress-related neuropsychiatric disorders. Nat Rev Neurosci 2022; 23:86-103. [PMID: 34893785 DOI: 10.1038/s41583-021-00540-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 12/25/2022]
Abstract
Stress is a primary risk factor for several neuropsychiatric disorders. Evidence from preclinical models and clinical studies of depression have revealed an array of structural and functional maladaptive changes, whereby adverse environmental factors shape the brain. These changes, observed from the molecular and transcriptional levels through to large-scale brain networks, to the behaviours reveal a complex matrix of interrelated pathophysiological processes that differ between sexes, providing insight into the potential underpinnings of the sex bias of neuropsychiatric disorders. Although many preclinical studies use chronic stress protocols, long-term changes are also induced by acute exposure to traumatic stress, opening a path to identify determinants of resilient versus susceptible responses to both acute and chronic stress. Epigenetic regulation of gene expression has emerged as a key player underlying the persistent impact of stress on the brain. Indeed, histone modification, DNA methylation and microRNAs are closely involved in many aspects of the stress response and reveal the glutamate system as a key player. The success of ketamine has stimulated a whole line of research and development on drugs directly or indirectly targeting glutamate function. However, the challenge of translating the emerging understanding of stress pathophysiology into effective clinical treatments remains a major challenge.
Collapse
Affiliation(s)
- Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Department of Pharmaceutical Sciences, University of Milano, Milan, Italy.
| |
Collapse
|
32
|
Lee J, Kwon H, Cho E, Jeon J, Lee IK, Cho WS, Park SJ, Lee S, Kim DH, Jung JW. Hydrangea macrophylla and Thunberginol C Attenuate Stress-Induced Anxiety in Mice. Antioxidants (Basel) 2022; 11:antiox11020234. [PMID: 35204117 PMCID: PMC8868050 DOI: 10.3390/antiox11020234] [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: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 12/19/2022] Open
Abstract
Stress is an important neurological input for successful life. However, chronic stress and stress hormones could be a cause of various neurological disorders including anxiety disorders. Therefore, there have been many efforts to find effective materials for curing stress-induced neurological disorders. In this study, we examined the effect of Hydrangea macrophylla (HM) on corticosterone-induced neurotoxicity, stress-induced anxiety in mice and suggested a possible active ingredient of HM. HM protected cortical neurons against neurotoxicity of corticosterone (CORT), a stress hormone. HM also blocked CORT-induced hippocampal synaptic deficit via regulating Akt signaling. Oral administration of HM improved chronic restraint stress-induced anxiety in Elevated Plus maze test along with reduction of plasma corticosterone and TNF-α levels. Moreover, HM reduced stress-induced neuroinflammation and oxidative stress. Thunberginol C, an active ingredient of HM, also prevented CORT-induced neuronal cell death and restraint stress-induced anxiety. Moreover, thunberginol C reduced plasma TNF-α level and neuroinflammation and oxidative stress. Collectively, HM could be a good candidate for preventing stress-induced neurological disorders and thunberginol C may be an active ingredient of HM for this purpose.
Collapse
Affiliation(s)
- Jihye Lee
- Division of Endocrinology, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Huiyoung Kwon
- Department of Health Sciences, The Graduate School of Dong-A University, Dong-A University, Busan 49315, Korea; (H.K.); (W.-S.C.)
| | - Eunbi Cho
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul 05029, Korea; (E.C.); (J.J.)
| | - Jieun Jeon
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul 05029, Korea; (E.C.); (J.J.)
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu 41944, Korea;
| | - Wan-Seob Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Dong-A University, Busan 49315, Korea; (H.K.); (W.-S.C.)
| | - Se Jin Park
- Department of Food Biotechnology and Environmental Science, School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Korea;
| | - Seungheon Lee
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea;
| | - Dong Hyun Kim
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul 05029, Korea; (E.C.); (J.J.)
- Correspondence: (D.H.K.); (J.W.J.)
| | - Ji Wook Jung
- Department of Herbal Medicinal Pharmacology, College of Herbal Bio-Industry, Daegu Haany University, Kyungsan 38610, Korea
- Correspondence: (D.H.K.); (J.W.J.)
| |
Collapse
|
33
|
Kemp GM, Altimimi HF, Nho Y, Heir R, Klyczek A, Stellwagen D. Sustained TNF signaling is required for the synaptic and anxiety-like behavioral response to acute stress. Mol Psychiatry 2022; 27:4474-4484. [PMID: 36104437 PMCID: PMC9734040 DOI: 10.1038/s41380-022-01737-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 12/14/2022]
Abstract
Acute stress triggers plasticity of forebrain synapses as well as behavioral changes. Here we reveal that Tumor Necrosis Factor α (TNF) is a required downstream mediator of the stress response in mice, necessary for stress-induced synaptic potentiation in the ventral hippocampus and for an increase in anxiety-like behaviour. Acute stress is sufficient to activate microglia, triggering the long-term release of TNF. Critically, on-going TNF signaling specifically in the ventral hippocampus is necessary to sustain both the stress-induced synaptic and behavioral changes, as these could be reversed hours after induction by antagonizing TNF signaling. This demonstrates that TNF maintains the synaptic and behavioral stress response in vivo, making TNF a potential novel therapeutic target for stress disorders.
Collapse
Affiliation(s)
- Gina M. Kemp
- grid.63984.300000 0000 9064 4811Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC Canada
| | - Haider F. Altimimi
- grid.63984.300000 0000 9064 4811Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC Canada
| | - Yoonmi Nho
- grid.63984.300000 0000 9064 4811Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC Canada
| | - Renu Heir
- grid.63984.300000 0000 9064 4811Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC Canada
| | - Adam Klyczek
- grid.63984.300000 0000 9064 4811Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC, Canada.
| |
Collapse
|
34
|
Yan Z, Rein B. Mechanisms of synaptic transmission dysregulation in the prefrontal cortex: pathophysiological implications. Mol Psychiatry 2022; 27:445-465. [PMID: 33875802 PMCID: PMC8523584 DOI: 10.1038/s41380-021-01092-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/13/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
The prefrontal cortex (PFC) serves as the chief executive officer of the brain, controlling the highest level cognitive and emotional processes. Its local circuits among glutamatergic principal neurons and GABAergic interneurons, as well as its long-range connections with other brain regions, have been functionally linked to specific behaviors, ranging from working memory to reward seeking. The efficacy of synaptic signaling in the PFC network is profundedly influenced by monoaminergic inputs via the activation of dopamine, adrenergic, or serotonin receptors. Stress hormones and neuropeptides also exert complex effects on the synaptic structure and function of PFC neurons. Dysregulation of PFC synaptic transmission is strongly linked to social deficits, affective disturbance, and memory loss in brain disorders, including autism, schizophrenia, depression, and Alzheimer's disease. Critical neural circuits, biological pathways, and molecular players that go awry in these mental illnesses have been revealed by integrated electrophysiological, optogenetic, biochemical, and transcriptomic studies of PFC. Novel epigenetic mechanism-based strategies are proposed as potential avenues of therapeutic intervention for PFC-involved diseases. This review provides an overview of PFC network organization and synaptic modulation, as well as the mechanisms linking PFC dysfunction to the pathophysiology of neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. Insights from the preclinical studies offer the potential for discovering new medical treatments for human patients with these brain disorders.
Collapse
Affiliation(s)
- Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | | |
Collapse
|
35
|
Ha GE, Cheong E. Chronic Restraint Stress Decreases the Excitability of Hypothalamic POMC Neuron and Increases Food Intake. Exp Neurobiol 2021; 30:375-386. [PMID: 34983879 PMCID: PMC8752322 DOI: 10.5607/en21037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 11/19/2022] Open
Abstract
Stress activates the hypothalamic-pituitary-adrenal system, and induces the release of glucocorticoids, stress hormones, into circulation. Many studies have shown that stress affects feeding behavior, however, the underlying circuitry and molecular mechanisms are not fully understood. The balance between orexigenic (simulating appetite) and anorexigenic (loss of appetite) signals reciprocally modulate feeding behavior. It is suggested that proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons in the arcuate nucleus (ARC) of the hypothalamus are the first-order neurons that respond to the circulating signals of hunger and satiety. Here, we examined a chronic restraint stress model and observed an increase in food intake, which was not correlated with anhedonia. We investigated whether stress affects the properties of POMC and NPY neurons and found that chronic restraint stress reduced the excitatory inputs onto POMC neurons and increased the action potential threshold. Therefore, our study suggests that chronic stress modulates the intrinsic excitability and excitatory inputs in POMC neurons, leading to changes in feeding behavior.
Collapse
Affiliation(s)
- Go Eun Ha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
36
|
Tse YC, Nath M, Larosa A, Wong TP. Opposing Changes in Synaptic and Extrasynaptic N-Methyl-D-Aspartate Receptor Function in Response to Acute and Chronic Restraint Stress. Front Mol Neurosci 2021; 14:716675. [PMID: 34690693 PMCID: PMC8531402 DOI: 10.3389/fnmol.2021.716675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
A pertinent mechanism by which stress impacts learning and memory is through stress-induced plastic changes in glutamatergic transmission in the hippocampus. For instance, acute stress has been shown to alter the expression, binding, and function of the ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR). However, the consequences of chronic stress, which could lead to various stress-related brain disorders, on NMDAR function remain unclear. While most studies on NMDARs focused on these receptors in synapses (synaptic NMDARs or sNMDARs), emerging findings have revealed functional roles of NMDARs outside synapses (extrasynaptic NMDARs or exNMDARs) that are distinct from those of sNMDARs. Using a restraint stress paradigm in adult rats, the objective of the current study is to examine whether sNMDARs and exNMDARs in the hippocampus are differentially regulated by acute and chronic stress. We examined sNMDAR and exNMDAR function in dorsal CA1 hippocampal neurons from brain slices of adult rats that were acutely (1 episode) or chronically (21 daily episodes) stressed by restraint (30 min). We found that acute stress increases sNMDAR but suppresses exNMDAR function. Surprisingly, we only observed a reduction in exNMDAR function after chronic stress. Taken together, our findings suggest that sNMDARs and exNMDARs may be differentially regulated by acute and chronic stress. Most importantly, the observed suppression in exNMDAR function by both acute and chronic stress implies crucial but overlooked roles of hippocampal exNMDARs in stress-related disorders.
Collapse
Affiliation(s)
- Yiu Chung Tse
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada
| | - Moushumi Nath
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Amanda Larosa
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
37
|
Lesuis SL, Brosens N, Immerzeel N, van der Loo RJ, Mitrić M, Bielefeld P, Fitzsimons CP, Lucassen PJ, Kushner SA, van den Oever MC, Krugers HJ. Glucocorticoids Promote Fear Generalization by Increasing the Size of a Dentate Gyrus Engram Cell Population. Biol Psychiatry 2021; 90:494-504. [PMID: 34503674 DOI: 10.1016/j.biopsych.2021.04.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Traumatic experiences, such as conditioned threat, are coded as enduring memories that are frequently subject to generalization, which is characterized by (re-) expression of fear in safe environments. However, the neurobiological mechanisms underlying threat generalization after a traumatic experience and the role of stress hormones in this process remain poorly understood. METHODS We examined the influence of glucocorticoid hormones on the strength and specificity of conditioned fear memory at the level of sparsely distributed dentate gyrus (DG) engram cells in male mice. RESULTS We found that elevating glucocorticoid hormones after fear conditioning induces a generalized contextual fear response. This was accompanied by a selective and persistent increase in the excitability and number of activated DG granule cells. Selective chemogenetic suppression of these sparse cells in the DG prevented glucocorticoid-induced fear generalization and restored contextual memory specificity, while leaving expression of auditory fear memory unaffected. CONCLUSIONS These results implicate the sparse ensemble of DG engram cells as a critical cellular substrate underlying fear generalization induced by glucocorticoid stress hormones.
Collapse
Affiliation(s)
- Sylvie L Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Niek Brosens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Nathalie Immerzeel
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Miodrag Mitrić
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pascal Bielefeld
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlos P Fitzsimons
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Doyle MA, Bali V, Eagle AL, Stark AR, Fallon B, Neve RL, Robison AJ, Mazei-Robison MS. Serum- and glucocorticoid-inducible kinase 1 activity in ventral tegmental area dopamine neurons regulates cocaine conditioned place preference but not cocaine self-administration. Neuropsychopharmacology 2021; 46:1574-1583. [PMID: 34007042 PMCID: PMC8280171 DOI: 10.1038/s41386-021-01032-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023]
Abstract
Drugs of abuse regulate the activity of the mesolimbic dopamine (DA) system, and drug-induced changes in ventral tegmental area (VTA) cellular activity and gene regulation are linked to behavioral outputs associated with addiction. Previous work from our lab determined that VTA serum- and glucocorticoid-inducible kinase 1 (SGK1) transcription and catalytic activity were increased by repeated cocaine administration; however, it was unknown if these biochemical changes contributed to cocaine-elicited behaviors. Using transgenic and viral-mediated manipulations, we investigated the role of VTA SGK1 catalytic activity in regulating cocaine conditioned place preference and self-administration. We showed intra-VTA infusion of a catalytically inactive SGK1 mutant (K127Q) significantly decreased cocaine conditioned place preference (CPP). Further, we found that K127Q expression in VTA DA neurons significantly decreased cocaine CPP, while this same manipulation in VTA GABA neurons had no effect. However, blunted VTA DA SGK1 catalytic activity did not alter cocaine self-administration. Altogether, these studies identify the specific VTA cells critical for SGK1-mediated effects on cocaine CPP but not self-administration.
Collapse
Affiliation(s)
- Marie A. Doyle
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA
| | - Vedrana Bali
- grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| | - Andrew L. Eagle
- grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| | - Ali R. Stark
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA
| | - Barbara Fallon
- grid.17088.360000 0001 2150 1785Pharmacology and Toxicology Department, Michigan State University, East Lansing, USA
| | - Rachael L. Neve
- grid.32224.350000 0004 0386 9924Gene Technology Core, Massachusetts General Hospital, Boston, USA
| | - A. J. Robison
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA ,grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| | - Michelle S. Mazei-Robison
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA ,grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| |
Collapse
|
39
|
Floriou-Servou A, von Ziegler L, Waag R, Schläppi C, Germain PL, Bohacek J. The Acute Stress Response in the Multiomic Era. Biol Psychiatry 2021; 89:1116-1126. [PMID: 33722387 DOI: 10.1016/j.biopsych.2020.12.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/13/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022]
Abstract
Studying the stress response is a major pillar of neuroscience research not only because stress is a daily reality but also because the exquisitely fine-tuned bodily changes triggered by stress are a neuroendocrinological marvel. While the genome-wide changes induced by chronic stress have been extensively studied, we know surprisingly little about the complex molecular cascades triggered by acute stressors, the building blocks of chronic stress. The acute stress (or fight-or-flight) response mobilizes organismal energy resources to meet situational demands. However, successful stress coping also requires the efficient termination of the stress response. Maladaptive coping-particularly in response to severe or repeated stressors-can lead to allostatic (over)load, causing wear and tear on tissues, exhaustion, and disease. We propose that deep molecular profiling of the changes triggered by acute stressors could provide molecular correlates for allostatic load and predict healthy or maladaptive stress responses. We present a theoretical framework to interpret multiomic data in light of energy homeostasis and activity-dependent gene regulation, and we review the signaling cascades and molecular changes rapidly induced by acute stress in different cell types in the brain. In addition, we review and reanalyze recent data from multiomic screens conducted mainly in the rodent hippocampus and amygdala after acute psychophysical stressors. We identify challenges surrounding experimental design and data analysis, and we highlight promising new research directions to better understand the stress response on a multiomic level.
Collapse
Affiliation(s)
- Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Christa Schläppi
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Pierre-Luc Germain
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland; Laboratory of Statistical Bioinformatics, Department for Molecular Life Sciences, University of Zürich, Zürich, Switzerland.
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland.
| |
Collapse
|
40
|
Stress, memory, and implications for major depression. Behav Brain Res 2021; 412:113410. [PMID: 34116119 DOI: 10.1016/j.bbr.2021.113410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 06/04/2021] [Accepted: 06/06/2021] [Indexed: 12/22/2022]
Abstract
The stress response comprises a phylogenetically conserved set of cognitive, physiological, and behavioral responses that evolved as a survival strategy. In this context, the memory of stressful events would be adaptive as it could avoid re-exposure to an adverse event, otherwise the event would be facilitated in positively stressful or non-distressful conditions. However, the interaction between stress and memory comprises complex responses, some of them which are not yet completely understood, and which depend on several factors such as the memory system that is recruited, the nature and duration of the stressful event, as well as the timing in which this interaction takes place. In this narrative review, we briefly discuss the mechanisms of the stress response, the main memory systems, and its neural correlates. Then, we show how stress, through the action of its biochemical mediators, influences memory systems and mnemonic processes. Finally, we make use of major depressive disorder to explore the possible implications of non-adaptive interactions between stress and memory to psychiatric disorders, as well as possible roles for memory studies in the field of psychiatry.
Collapse
|
41
|
Chu SF, Zhang Z, Zhou X, He WB, Yang B, Cui LY, He HY, Wang ZZ, Chen NH. Low corticosterone levels attenuate late life depression and enhance glutamatergic neurotransmission in female rats. Acta Pharmacol Sin 2021; 42:848-860. [PMID: 33028984 PMCID: PMC8149629 DOI: 10.1038/s41401-020-00536-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022] Open
Abstract
Sustained elevation of corticosterone (CORT) is one of the common causes of aging and major depression disorder. However, the role of elevated CORT in late life depression (LLD) has not been elucidated. In this study, 18-month-old female rats were subjected to bilateral adrenalectomy or sham surgery. Their CORT levels in plasma were adjusted by CORT replacement and the rats were divided into high-level CORT (H-CORT), low-level CORT (L-CORT), and Sham group. We showed that L-CORT rats displayed attenuated depressive symptoms and memory defects in behavioral tests as compared with Sham or H-CORT rats. Furthermore, we showed that glutamatergic transmission was enhanced in L-CORT rats, evidenced by enhanced population spike amplitude (PSA) recorded from the dentate gyrus of hippocampus in vivo and increased glutamate release from hippocampal synaptosomes caused by high frequency stimulation or CORT exposure. Intracerebroventricular injection of an enzymatic glutamate scavenger system, glutamic-pyruvic transmine (GPT, 1 μM), significantly increased the PSA in Sham rats, suggesting that extracelluar accumulation of glutamate might be the culprit of impaired glutamatergic transmission, which was dependent on the uptake by Glt-1 in astrocytes. We revealed that hippocampal Glt-1 expression level in the L-CORT rats was much higher than in Sham and H-CORT rats. In a gradient neuron-astrocyte coculture, we found that the expression of Glt-1 was decreased with the increase of neural percentage, suggesting that impairment of Glt-1 might result from the high level of CORT contributed neural damage. In sham rats, administration of DHK that inhibited Glt-1 activity induced significant LLD symptoms, whereas administration of RIL that promoted glutamate uptake significantly attenuated LLD. All of these results suggest that glutamatergic transmission impairment is one of important pathogenesis in LLD induced by high level of CORT, which provide promising clues for the treatment of LLD.
Collapse
Affiliation(s)
- Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xin Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Bo Yang
- Department of Pharmacy, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin, 300300, China
| | - Li-Yuan Cui
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hong-Yuan He
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
42
|
Cooper JA, Nuutinen MR, Lawlor VM, DeVries BAM, Barrick EM, Hossein S, Cole DJ, Leonard CV, Hahn EC, Teer AP, Shields GS, Slavich GM, Ongur D, Jensen JE, Du F, Pizzagalli DA, Treadway MT. Reduced adaptation of glutamatergic stress response is associated with pessimistic expectations in depression. Nat Commun 2021; 12:3166. [PMID: 34039978 PMCID: PMC8155144 DOI: 10.1038/s41467-021-23284-9] [Citation(s) in RCA: 13] [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: 05/18/2020] [Accepted: 04/19/2021] [Indexed: 01/20/2023] Open
Abstract
Stress is a significant risk factor for the development of major depressive disorder (MDD), yet the underlying mechanisms remain unclear. Preclinically, adaptive and maladaptive stress-induced changes in glutamatergic function have been observed in the medial prefrontal cortex (mPFC). Here, we examine stress-induced changes in human mPFC glutamate using magnetic resonance spectroscopy (MRS) in two healthy control samples and a third sample of unmedicated participants with MDD who completed the Maastricht acute stress task, and one sample of healthy control participants who completed a no-stress control manipulation. In healthy controls, we find that the magnitude of mPFC glutamate response to the acute stressor decreases as individual levels of perceived stress increase. This adaptative glutamate response is absent in individuals with MDD and is associated with pessimistic expectations during a 1-month follow-up period. Together, this work shows evidence for glutamatergic adaptation to stress that is significantly disrupted in MDD.
Collapse
Affiliation(s)
| | | | | | | | - Elyssa M Barrick
- Center for Depression, Anxiety and Stress Research, McLean Hospital/Harvard Medical School, Belmont, MA, USA
| | - Shabnam Hossein
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Daniel J Cole
- Department of Psychology, Emory University, Atlanta, GA, USA
| | | | - Evan C Hahn
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Andrew P Teer
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Grant S Shields
- Department of Psychological Science, University of Arkansas, Fayetteville, AR, USA
| | - George M Slavich
- Cousins Center for Psychoneuroimmunology and Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Dost Ongur
- Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA, USA
| | - J Eric Jensen
- McLean Imaging Center, McLean Hospital/Harvard Medical School, Belmont, MA, USA
| | - Fei Du
- McLean Imaging Center, McLean Hospital/Harvard Medical School, Belmont, MA, USA
| | - Diego A Pizzagalli
- Center for Depression, Anxiety and Stress Research, McLean Hospital/Harvard Medical School, Belmont, MA, USA
- Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA, USA
- McLean Imaging Center, McLean Hospital/Harvard Medical School, Belmont, MA, USA
| | - Michael T Treadway
- Department of Psychology, Emory University, Atlanta, GA, USA.
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA.
| |
Collapse
|
43
|
Liu B, Li N, He Z, Zhang X, Duan G. Emerging Role of Serum Glucocorticoid-Regulated Kinase 1 in Pathological Pain. Front Mol Neurosci 2021; 14:683527. [PMID: 34093127 PMCID: PMC8177009 DOI: 10.3389/fnmol.2021.683527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/22/2021] [Indexed: 11/28/2022] Open
Abstract
Currently, the management of acute and chronic pain in clinical practice remains unsatisfactory due to the existence of limited effective treatments, and novel therapeutic strategies for pathological pain are urgently needed. In the past few decades, the role of serum and glucocorticoid-inducible kinase 1 (SGK1) in the development of pain and diurnal rhythms has been implicated in numerous studies. The expression levels of SGK1 mRNA and protein were found to be elevated in the spinal cord and brain in various pathological pain models. Blocking SGK1 significantly attenuated pain-like responses and the development of pathological pain. These studies provide strong evidence that SGK1 plays a role in the development of various types of pathological pain and that targeting SGK1 may be a novel therapeutic strategy for pain management. In this review article, we provide evidence from animal models for the potential role of SGK1 in the regulation of pathological pain caused by inflammation, nerve injury, psychiatric disorders, and chronic opioid exposure.
Collapse
Affiliation(s)
- Baowen Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ningbo Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhigang He
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangyou Duan
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Sanguino‐Gómez J, Buurstede JC, Abiega O, Fitzsimons CP, Lucassen PJ, Eggen BJL, Lesuis SL, Meijer OC, Krugers HJ. An emerging role for microglia in stress‐effects on memory. Eur J Neurosci 2021; 55:2491-2518. [PMID: 33724565 PMCID: PMC9373920 DOI: 10.1111/ejn.15188] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/13/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Stressful experiences evoke, among others, a rapid increase in brain (nor)epinephrine (NE) levels and a slower increase in glucocorticoid hormones (GCs) in the brain. Microglia are key regulators of neuronal function and contain receptors for NE and GCs. These brain cells may therefore potentially be involved in modulating stress effects on neuronal function and learning and memory. In this review, we discuss that stress induces (1) an increase in microglial numbers as well as (2) a shift toward a pro‐inflammatory profile. These microglia have (3) impaired crosstalk with neurons and (4) disrupted glutamate signaling. Moreover, microglial immune responses after stress (5) alter the kynurenine pathway through metabolites that impair glutamatergic transmission. All these effects could be involved in the impairments in memory and in synaptic plasticity caused by (prolonged) stress, implicating microglia as a potential novel target in stress‐related memory impairments.
Collapse
Affiliation(s)
| | - Jacobus C. Buurstede
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Oihane Abiega
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems Section Molecular Neurobiology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
- Program in Neurosciences and Mental Health Hospital for Sick Children Toronto ON Canada
| | - Onno C. Meijer
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
45
|
Wu J, Liu C, Zhang L, He B, Shi WP, Shi HL, Qin C. Chronic restraint stress impairs cognition via modulating HDAC2 expression. Transl Neurosci 2021; 12:154-163. [PMID: 33986954 PMCID: PMC8090798 DOI: 10.1515/tnsci-2020-0168] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
Background To investigate the effects of chronic restraint stress on cognition and the probable molecular mechanism in mice. Methods In the current work, a restraining tube was used as a way to induce chronic stress in mice. The protein levels were determined with ELISA and western blot. A series of behavior tests, including the Morris water maze, elevated plus maze, open field test, and novel object recognition test, were also performed to examine the anxiety and the ability of learning and memory. Moreover, murine neuroblastoma N2a cells were used to confirm the findings from mice under chronic stress. Results Decreased synaptic functions were impaired in chronic stress with the downregulation of PSD95, GluR-1, the neurotrophic factor BDNF, and immediate-onset genes Arc and Egr. Chronic restraint decreased the histone acetylation level in hippocampal neurons while HDAC2 was increased and was co-localized with glucocorticoid receptors. Moreover, chronic stress inhibited the PI3K/AKT signaling pathway and induced energy metabolism dysfunctions. Conclusion This work examining the elevated levels of HDAC2 in the hippocampus may provide new insights and targets for drug development for treating many neurodegenerative diseases.
Collapse
Affiliation(s)
- Jie Wu
- Pathology Department, Comparative Medical Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Panjiayuan Nanli No. 5, Beijing, 100021, People's Republic of China.,Department of Pathology, Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, Shandong province, 266003, People's Republic of China
| | - Cui Liu
- Comparative Medical Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Panjiayuan Nanli No. 5, Beijing, 100021, People's Republic of China
| | - Ling Zhang
- Comparative Medical Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Panjiayuan Nanli No. 5, Beijing, 100021, People's Republic of China
| | - Bing He
- Department of Pathology, Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, Shandong province, 266003, People's Republic of China
| | - Wei-Ping Shi
- Department of Pathology, Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, Shandong province, 266003, People's Republic of China
| | - Hai-Lei Shi
- Department of Pathology, Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, Shandong province, 266003, People's Republic of China
| | - Chuan Qin
- Comparative Medical Center, Peking Union Medical College (PUMC) and Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Panjiayuan Nanli No. 5, Beijing, 100021, People's Republic of China
| |
Collapse
|
46
|
Oligophrenin-1 moderates behavioral responses to stress by regulating parvalbumin interneuron activity in the medial prefrontal cortex. Neuron 2021; 109:1636-1656.e8. [PMID: 33831348 DOI: 10.1016/j.neuron.2021.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 02/09/2021] [Accepted: 03/10/2021] [Indexed: 12/28/2022]
Abstract
Ample evidence indicates that individuals with intellectual disability (ID) are at increased risk of developing stress-related behavioral problems and mood disorders, yet a mechanistic explanation for such a link remains largely elusive. Here, we focused on characterizing the syndromic ID gene oligophrenin-1 (OPHN1). We find that Ophn1 deficiency in mice markedly enhances helpless/depressive-like behavior in the face of repeated/uncontrollable stress. Strikingly, Ophn1 deletion exclusively in parvalbumin (PV) interneurons in the prelimbic medial prefrontal cortex (PL-mPFC) is sufficient to induce helplessness. This behavioral phenotype is mediated by a diminished excitatory drive onto Ophn1-deficient PL-mPFC PV interneurons, leading to hyperactivity in this region. Importantly, suppressing neuronal activity or RhoA/Rho-kinase signaling in the PL-mPFC reverses helpless behavior. Our results identify OPHN1 as a critical regulator of adaptive behavioral responses to stress and shed light onto the mechanistic links among OPHN1 genetic deficits, mPFC circuit dysfunction, and abnormalities in stress-related behaviors.
Collapse
|
47
|
Park JC, Jeon YJ, Jang YS, Cho J, Choi DH, Han JS. SGK1 knockdown in the medial prefrontal cortex reduces resistance to stress-induced memory impairment. Eur Neuropsychopharmacol 2021; 45:29-34. [PMID: 33735826 DOI: 10.1016/j.euroneuro.2021.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/07/2021] [Accepted: 02/15/2021] [Indexed: 01/02/2023]
Abstract
Down-regulation of serum and glucocorticoid-regulated kinase1 (SGK1) expression has been reported in the postmortem prefrontal cortex (PFC) of subjects with post-traumatic stress disorder. Furthermore, experimental treatments that reduce SGK1 function in the medial prefrontal cortex (mPFC) cause depressive-like behaviors and synaptic dysfunction. Therefore, we examined the effect of SGK1 down-regulation in the mPFC on resistance to stress-induced cognitive impairment. Rats with viral-mediated knockdown of SGK1 in the mPFC were subjected to either a brief 20-min restraint plus 20 intermittent tail shocks or a prolonged 60-min restraint plus 60 intermittent tail shocks, after which their performance in an object recognition task was assessed. Recognition memory remained intact in control rats following the brief stress, but was impaired in rats with SGK1 knockdown in the mPFC. Prolonged stress impaired recognition memory in both control rats and rats with SGK1 knockdown. Our findings indicate that altered mPFC SGK1 signaling is a potential mechanism for resistance to stress-induced cognitive impairment.
Collapse
Affiliation(s)
- Jung-Cheol Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Yong-Jae Jeon
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Yoon-Sun Jang
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Jeiwon Cho
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Dong-Hee Choi
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Republic of Korea; Department of Medical Science, Konkuk University School of Medicine, Konkuk University, Seoul 05029, Republic of Korea.
| | - Jung-Soo Han
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
48
|
Velli A, Iordanidou C, Asimi T, Vynichaki MI, Cholevas A, Mantouka AI, Nassens L, Chalkiadaki K, Sidiropoulou K. Sexual dimorphic effects of restraint stress on prefrontal cortical function are mediated by glucocorticoid receptor activation. Eur J Neurosci 2021; 55:2754-2765. [PMID: 33759255 DOI: 10.1111/ejn.15203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/04/2023]
Abstract
Stress, a major regulator and precipitating factor of cognitive and emotional disorders, differentially manifests between males and females. Our aim was to investigate the mechanisms underlying the sexual dimorphic effects of acute restraint stress (RS) on males and females on the function of the prefrontal cortex (PFC). Adult male and female mice were subjected to RS or left in their home-cage (NR), and then tested in the light-dark test followed by the temporal order object recognition (TOR) task. Female mice exhibited increased anxiety-like levels, whereas male mice only showed deficits in the TOR task. When the behavioural tests were conducted 24 hr following restraint stress (RS24), only the reduced performance in the TOR task in male mice persisted. In a different cohort, evoked field excitatory postsynaptic potentials (fEPSPs) were recorded in layer II of acute PFC slices, immediately or 24 hr after RS. Long-term potentiation (LTP) was significantly reduced in RS and RS24 male, but not female, compared with their respective NR group. LTP in PFC slices incubated with corticosterone showed significantly reduced LTP only in males. To determine whether glucocorticoid signalling is implicated in the RS-induced behavioural effects, a different cohort of mice was administered mifepristone, a corticosterone receptor antagonist. Mifepristone administration 1 hr before RS prevented the effects of RS on the TOR task in males, but not anxiety. In conclusion, RS has differential effects on recency memory and anxiety, in males and females, which are partly mediated by the effects of corticosterone signalling on synaptic plasticity.
Collapse
Affiliation(s)
- Angeliki Velli
- Department of Biology, University of Crete, Rethimno, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklio, Greece
| | | | - Theodora Asimi
- Department of Biology, University of Crete, Rethimno, Greece
| | | | | | | | - Liesje Nassens
- Department of Biology, University of Crete, Rethimno, Greece
| | | | - Kyriaki Sidiropoulou
- Department of Biology, University of Crete, Rethimno, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklio, Greece
| |
Collapse
|
49
|
Jaszczyk A, Juszczak GR. Glucocorticoids, metabolism and brain activity. Neurosci Biobehav Rev 2021; 126:113-145. [PMID: 33727030 DOI: 10.1016/j.neubiorev.2021.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022]
Abstract
The review integrates different experimental approaches including biochemistry, c-Fos expression, microdialysis (glutamate, GABA, noradrenaline and serotonin), electrophysiology and fMRI to better understand the effect of elevated level of glucocorticoids on the brain activity and metabolism. The available data indicate that glucocorticoids alter the dynamics of neuronal activity leading to context-specific changes including both excitation and inhibition and these effects are expected to support the task-related responses. Glucocorticoids also lead to diversification of available sources of energy due to elevated levels of glucose, lactate, pyruvate, mannose and hydroxybutyrate (ketone bodies), which can be used to fuel brain, and facilitate storage and utilization of brain carbohydrate reserves formed by glycogen. However, the mismatch between carbohydrate supply and utilization that is most likely to occur in situations not requiring energy-consuming activities lead to metabolic stress due to elevated brain levels of glucose. Excessive doses of glucocorticoids also impair the production of energy (ATP) and mitochondrial oxidation. Therefore, glucocorticoids have both adaptive and maladaptive effects consistently with the concept of allostatic load and overload.
Collapse
Affiliation(s)
- Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzebiec, 36a Postepu str., Poland
| | - Grzegorz R Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzebiec, 36a Postepu str., Poland.
| |
Collapse
|
50
|
Kaur M, Sanches M. Experimental Therapeutics in Treatment-Resistant Major Depressive Disorder. J Exp Pharmacol 2021; 13:181-196. [PMID: 33658867 PMCID: PMC7917305 DOI: 10.2147/jep.s259302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Treatment-Resistant Depression (TRD) patients remain a challenging sub-division of patients with Major Depressive Disorder (MDD) in day to day clinical practice. As with any diagnostic condition, comprehensive evaluation, exclusion of other psychiatric conditions, assessment of co-morbid medical and psychiatric illnesses and psychosocial stressors are the keys to appropriate diagnosis and subsequent management. There are various management options available for the treatment of MDD, however, about 30% of the patients fail to achieve full remission of symptoms despite multiple trials and belong to this sub-category of MDD. This article brings forth discussion of other non-conventional medication and non-medication treatment alternatives that merit exploration of their efficacy in TRD. Many of the proposed novel medications and other treatment modalities such as Deep Brain Stimulation, exercise, yoga are already used for other medical and psychiatric disorders and have some evidence suggesting their potential benefits in TRD in conjunction with conventional medications or even as monotherapy. Nevertheless, more research is needed in this direction to establish effectiveness.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Behavioral Medicine, Cone Health, Greensboro, NC, USA
| | - Marsal Sanches
- UT Health Center of Excellence on Mood Disorders, Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, Houston, TX, USA
| |
Collapse
|