1
|
Wallsten B, Gligor AH, Gonzalez AE, Ramos JD, Baratta MV, Sorg BA. Response of parvalbumin interneurons and perineuronal nets in rat medial prefrontal cortex and lateral amygdala to stressor controllability. Brain Res 2024; 1848:149351. [PMID: 39592089 DOI: 10.1016/j.brainres.2024.149351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024]
Abstract
Behavioral control over a stressor limits the impact of the stressor being experienced and produces enduring changes that reduce the effects of future stressors. In rats, these stress-buffering effects of control (escapable stress, ES) require activation of the medial prefrontal cortex (mPFC) and prevent the typical amygdala-dependent behavioral outcomes of uncontrollable stress (inescapable stress, IS). Parvalbumin (PV) interneurons regulate output of excitatory neurons, and most mPFC PV neurons are surrounded by perineuronal nets (PNNs), which regulate firing. We exposed male rats to a single session of ES, IS, or no stress and measured c-Fos expression within PV/PNN-containing cells in mPFC subregions (prelimbic, PL; infralimbic, IL) and in the lateral amygdala. We also measured the number and intensity of PNNs. Within PL and IL PV/PNN cells, both ES and IS increased c-Fos intensity in PV/PNN, non-PV, and non-PNN cells. Within the IL, only ES increased the number of c-Fos-expressing PV/PNN-labeled cells. In the lateral amygdala, only ES increased c-Fos intensity within PV cells and PV/PNN cells. Thus, PV neurons in the IL and lateral amygdala may represent an important substrate by which behavioral control buffers against the amygdala-dependent behavioral outcomes typically observed after uncontrollable stress.
Collapse
Affiliation(s)
- Brittani Wallsten
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Abigail H Gligor
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Angela E Gonzalez
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States; Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Program in Neuroscience, Washington State University, Vancouver, WA 98686, United States
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, United States
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States; Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Program in Neuroscience, Washington State University, Vancouver, WA 98686, United States.
| |
Collapse
|
2
|
Hu L, Qiu MJ, Fan WJ, Wang WE, Liu SH, Liu XQ, Liu SW, Shen ZJ, Zheng YF, Liu GC, Jia ZY, Wang XQ, Fang N. Characterization of GABAergic marker expression in prefrontal cortex in dexamethasone induced depression/anxiety model. Front Endocrinol (Lausanne) 2024; 15:1433026. [PMID: 39483976 PMCID: PMC11524930 DOI: 10.3389/fendo.2024.1433026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background The pivotal responsibility of GABAergic interneurons is inhibitory neurotransmission; in this way, their significance lies in regulating the maintenance of excitation/inhibition (E/I) balance in cortical circuits. An abundance of glucocorticoids (GCs) exposure results in a disorder of GABAergic interneurons in the prefrontal cortex (PFC); the relationship between this status and an enhanced vulnerability to neuropsychiatric ailments, like depression and anxiety, has been identified, but this connection is still poorly understood because systematic and comprehensive research is lacking. Here, we aim to investigate the impact of dexamethasone (DEX, a GC receptor agonist) on GABAergic interneurons in the PFC of eight-week-old adult male mice. Methods A double-blind study was conducted where thirty-two mice were treated subcutaneously either saline or DEX (0.2 mg/10 ml per kg of body weight) dissolved in saline daily for 21 days. Weight measurements were taken at five-day intervals to assess the emotional changes in mice as well as the response to DEX treatment. Following the 21-day regimen of DEX injections, mice underwent examinations for depression/anxiety-like behaviours and GABAergic marker expression in PFC. Results In a depression/anxiety model generated by chronic DEX treatment, we found that our DEX procedure did trigger depression/anxiety-like behaviors in mice. Furthermore, DEX treatment reduced the expression levels of a GABA-synthesizing enzyme (GAD67), Reelin, calcium-binding proteins (parvalbumin and calretinin) and neuropeptides co-expressed in GABAergic neurons (somatostatin, neuropeptide Y and vasoactive intestinal peptide) in the PFC were reduced after 21 days of DEX treatment; these reductions were accompanied by decreases in brain size and cerebral cortex thickness. Conclusion Our results indicate that a reduction in the number of GABAergic interneurons may result in deficiencies in cortical inhibitory neurotransmission, potentially causing an E/I imbalance in the PFC; this insight suggests a potential breakthrough strategy for the treatment of depression and anxiety.
Collapse
Affiliation(s)
- Ling Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ming-Jing Qiu
- Neurological Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen-Juan Fan
- Luohe Medical College, Henan Province Engineering Research Center of Nutrition and Health, Luohe, China
| | - Wan-Er Wang
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Shao-Hao Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xiao-Qi Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Shi-Wei Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Ze-Jin Shen
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Ya-Fei Zheng
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Guang-Chao Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Zi-Yi Jia
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xiao-Qing Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Na Fang
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
3
|
Omoumi S, Rashidy‐Pour A, Seyedinia SA, Tarahomi P, Sedaghat K, Vafaei AA, Raise‐Abdullahi P. Systemic corticosterone enhances fear memory extinction in rats: Involvement of the infralimbic medial prefrontal cortex GABA A and GABA B receptors. Brain Behav 2024; 14:e70043. [PMID: 39344296 PMCID: PMC11440020 DOI: 10.1002/brb3.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/27/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE The infralimbic (IL) subregion of the medial prefrontal cortex (mPFC) regulates the extinction of conditioned fear memory. Glucocorticoid and gamma-aminobutyric acid (GABA) receptors are expressed in the mPFC and are also critical in fear extinction. This study investigated the possible interactive effects of the glucocorticoids and GABAergic system in the IL on the regulation of fear extinction. METHOD The rats were trained using an auditory fear conditioning task during which they received three conditioned stimuli (tones, 30 s, 4 kHz, 80 dB), co-terminated with the three unconditioned stimuli (footshock, 0.8 mA, 1 s). Extinction testing was conducted over 3 days (Ext 1-3). Thirty minutes before the first extinction trial (Ext 1), the rats received bicuculline (BIC, 1 mg/kg/2 mL, intraperitoneal [i.p.]) as a GABAA receptor antagonist or CGP55845 (CGP, 0.1 mg/kg/2 ML, i.p.) as a GABAB receptor antagonist followed by systemic injection of corticosterone (CORT, 3 mg/kg/2 ML, i.p.). Furthermore, separate groups of rats received a bilateral intra-IL injection of BIC (100 ng/0.3 µL/side) or CGP (10 ng/0.3 µL/side) followed by a systemic injection of CORT (3 mg/kg/2 ML, i.p.) before the first extinction trial (Ext 1). The extracellular signal-regulated kinase (ERK1) and cAMP response element-binding (CREB) activity in the IL was examined by Western blot analysis after Ext 1. FINDING The results indicated that systemic CORT injection facilitated fear extinction and increased the expression of ERK1 but not CREB in the IL. Both systemic and intra-IL co-injection of BIC or CGP blocked the effects of CORT on fear extinction and ERK1 expression. CONCLUSION These findings suggest that glucocorticoids and the GABAergic system may modulate fear extinction through the ERK pathway in the IL.
Collapse
Affiliation(s)
- Samira Omoumi
- Research Center of PhysiologySemnan University of Medical SciencesSemnanIran
- Department of Physiology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Ali Rashidy‐Pour
- Research Center of PhysiologySemnan University of Medical SciencesSemnanIran
- Department of Physiology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Seyed Ali Seyedinia
- Research Center of PhysiologySemnan University of Medical SciencesSemnanIran
- Student Research Committee, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Parnia Tarahomi
- Research Center of PhysiologySemnan University of Medical SciencesSemnanIran
- Student Research Committee, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Katayoun Sedaghat
- Research Center of PhysiologySemnan University of Medical SciencesSemnanIran
- Department of Physiology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Abbas Ali Vafaei
- Research Center of PhysiologySemnan University of Medical SciencesSemnanIran
- Department of Physiology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | | |
Collapse
|
4
|
Miller CN, Li Y, Beier KT, Aoto J. Acute stress causes sex-dependent changes to ventral subiculum synapses, circuitry, and anxiety-like behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606264. [PMID: 39131353 PMCID: PMC11312572 DOI: 10.1101/2024.08.02.606264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Experiencing a single severe stressor is sufficient to drive sexually dimorphic psychiatric disease development. The ventral subiculum (vSUB) emerges as a site where stress may induce sexually dimorphic adaptations due to its sex-specific organization and pivotal role in stress integration. Using a 1-hr acute restraint stress model, we uncover that stress causes a net decrease in vSUB activity in females that is potent, long-lasting, and driven by adrenergic receptor signaling. By contrast, males exhibit a net increase in vSUB activity that is transient and driven by corticosterone signaling. We further identified sex-dependent changes in vSUB output to the bed nucleus of the stria terminalis and in anxiety-like behavior in response to stress. These findings reveal striking changes in psychiatric disease-relevant brain regions and behavior following stress with sex-, cell-type, and synapse-specificity that contribute to our understanding of sex-dependent adaptations that may shape stress-related psychiatric disease risk.
Collapse
Affiliation(s)
- Carley N Miller
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yuan Li
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA 92697
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA 92697
- Department of Biomedical Engineering, University of California, Irvine, CA, USA 92697
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA 92697
| | - Jason Aoto
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
5
|
Choudhary D, Sasibhushana RB, Shankaranarayana Rao BS, Srikumar BN. Mifepristone blocks the anxiolytic- and antidepressant-like effects of allopregnanolone in male rats. Int J Neurosci 2024; 134:839-848. [PMID: 36469636 DOI: 10.1080/00207454.2022.2153047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Allopregnanolone (3α, 5α-tetrahydroprogesterone) is an inhibitory neurosteroid synthesized from progesterone via 5α-reductase activity in the brain and has anxiolytic, antidepressant, sedative, anticonvulsant, and analgesic activity. Altered levels of allopregnanolone cause anxiety, depression, premenstrual syndrome, and psychiatric disorders. Although allopregnanolone exerts most of its actions by modulating GABAA receptor, NMDA receptor, BDNF expression, and PXR activity, a recent study showed its effects are blocked by mifepristone on lordosis behavior which indicates the involvement of progestin or glucocorticoid receptors in the effects of allopregnanolone since mifepristone blocks both these receptors. However, whether these receptors are involved in acute anxiolytic or antidepressant-like effects is unknown. METHODS Adult male Wistar rats were used to study whether the prior administration of mifepristone would alter the effects of allopregnanolone in the elevated plus maze (EPM) and forced swim test (FST) was evaluated. RESULTS 10 mg/Kg dose of allopregnanolone increased percent open arm entries in the EPM, whereas 3 mg/Kg dose of allopregnanolone decreased percent immobility in the FST. Mifepristone administration resulted in a U-shaped response in the FST (with 1 mg/Kg, s.c., decreasing the immobility time) without significantly impacting the behavior in the EPM. In combination studies, mifepristone blocked the anxiolytic and antidepressant effects of allopregnanolone. CONCLUSION The current study provides evidence for the first time that progestin or glucocorticoid receptors are involved in the acute anxiolytic and antidepressant effects of allopregnanolone. Understanding the mechanism of action of allopregnanolone will help us design better therapeutic strategies to treat neuropsychiatric diseases such as depression and anxiety.
Collapse
Affiliation(s)
- Divya Choudhary
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
- Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, India
| | - R B Sasibhushana
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - B S Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - B N Srikumar
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| |
Collapse
|
6
|
Stephan M, Papiol S, Zhang M, Song J, Frommeyer SM, Haupt H, Jensen N, Kannaiyan N, Gupta R, Schuler P, Picklmann P, McCarthy M, Schulte E, Landen M, Falkai P, Scheuss V, Schulze T, Zhang W, Rossner MJ. Modulation of Neuronal Excitability and Plasticity by BHLHE41 Conveys Lithium Non-Responsiveness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.25.605130. [PMID: 39372797 PMCID: PMC11451663 DOI: 10.1101/2024.07.25.605130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Many bipolar disorder (BD) patients are non-responsive to lithium. The mechanisms underlying lithium (non-)responsiveness are largely unknown. By using gene-set enrichment analysis methods, we found that core clock gene-sets are significantly associated with lithium response. Among the top hits was BHLHE41, a modulator of the molecular clock and homeostatic sleep. Since BHLHE41 and its paralog BHLHE40 are functionally redundant, we assessed chronic lithium response in double-knockout mutant mice (DKO). We demonstrated that DKOs are non-responsive to lithium's effect in various behavioral tasks. Cellular assays and patch clamp recordings revealed lowered excitability and reduced lithium-response in prefrontal cortical layer 2/3 DKO neurons and on hippocampal long-term potentiation. Single-cell RNA sequencing identified that lithium deregulated mitochondrial respiration, cation channel and postsynapse associated gene-sets specifically in upper layer excitatory neurons. Our findings show that lithium acts in a highly cell-specific way on neuronal metabolism and excitability and modulates synaptic plasticity depending on BHLHE40/41.
Collapse
Affiliation(s)
- Marius Stephan
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Sergi Papiol
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Spain
| | - Mingyue Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Muenster, Germany
| | - Jie Song
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Samuel M Frommeyer
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Muenster, Germany
| | - Helen Haupt
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Muenster, Germany
| | - Niels Jensen
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
| | | | - Rajinder Gupta
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
| | - Philipp Schuler
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
| | - Pia Picklmann
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
| | - Michael McCarthy
- VA San Diego Healthcare System, CA, USA
- Department of Psychiatry, Center for Circadian Biology, University of California San Diego, San Diego, CA, USA
| | - Eva Schulte
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany
- Institute of Human Genetics, University Hospital, Faculty of Medicine, University of Bonn, Bonn, Germany
- Department of Psychiatry, University Hospital, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Mikael Landen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
| | - Volker Scheuss
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
- MSH Medical School, Hamburg, Germany
| | - Thomas Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Muenster, Germany
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, Molecular and Behavioral Neurobiology, LMU University Hospital, LMU Munich, Germany
- Systasy Bioscience GmbH, Munich, Germany
| |
Collapse
|
7
|
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
|
8
|
Martinez CA, Pantazopoulos H, Gisabella B, Stephens ET, Garteiser J, Del Arco A. Choice impulsivity after repeated social stress is associated with increased perineuronal nets in the medial prefrontal cortex. Sci Rep 2024; 14:7093. [PMID: 38528075 DOI: 10.1038/s41598-024-57599-6] [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: 01/12/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
Repeated stress can predispose to substance abuse. However, behavioral and neurobiological adaptations that link stress to substance abuse remain unclear. This study investigates whether intermittent social defeat (ISD), a stress protocol that promotes drug-seeking behavior, alters intertemporal decision-making and cortical inhibitory function in the medial prefrontal cortex (mPFC). Male long evans rats were trained in a delay discounting task (DDT) where rats make a choice between a fast (1 s) small reward (1 sugar pellet) and a large reward (3 sugar pellets) that comes with a time delay (10 s or 20 s). A decreased preference for delayed rewards was used as an index of choice impulsivity. Rats were exposed to ISD and tested in the DDT 24 h after each stress episode, and one- and two-weeks after the last stress episode. Immunohistochemistry was performed in rat's brains to evaluate perineuronal nets (PNNs) and parvalbumin GABA interneurons (PV) labeling as markers of inhibitory function in mPFC. ISD significantly decreased the preference for delayed large rewards in low impulsive, but not high impulsive, animals. ISD also increased the density of PNNs in the mPFC. These results suggest that increased choice impulsivity and cortical inhibition predispose animals to seek out rewards after stress.
Collapse
Affiliation(s)
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Emily T Stephens
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jacob Garteiser
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alberto Del Arco
- HESRM, School of Applied Sciences, University of Mississippi, Oxford, MS, USA.
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
9
|
Levi UI, Bintu MM, Daniella OC, Oyenike OAF, Agbonu AO, Adedamola AM, Ndidi E, Saka SF, Gela BV, Mbagwu SI, Edem EE, Olukayode OJ, James C. Neurobehavioral deficits, histoarchitectural alterations, parvalbumin neuronal damage and glial activation in the brain of male Wistar rat exposed to Landfill leachate. J Chem Neuroanat 2024; 136:102377. [PMID: 38176474 DOI: 10.1016/j.jchemneu.2023.102377] [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: 09/05/2023] [Revised: 11/28/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024]
Abstract
Concerns about inappropriate disposal of waste into unsanitary municipal solid waste landfills around the world have been on the increase, and this poses a public health challenge due to leachate production. The neurotoxic effect of Gwagwalada landfill leachate (GLL) was investigated in male adult Wistar rats. Rats were exposed to a 10% concentration of GLL for 21 days. The control group received tap water for the same period of the experiment. Our results showed that neurobehavior, absolute body and brain weights and brain histomorphology as well as parvalbumin interneurons were severely altered, with consequent astrogliosis and microgliosis after 21 days of administrating GLL. Specifically, there was severe loss and shrinkage of Purkinje cells, with their nucleus, and severe diffused vacuolations of the white matter tract of GLL-exposed rat brains. There was severe cell loss in the granular layer of the cerebellum resulting in a reduced thickness of the layer. Also, there was severe loss of dendritic arborization of the Purkinje cells in GLL-exposed rat brains, and damage as well as reduced populations of parvalbumin-containing fast-spiking GABAergic interneurons in various regions of the brain. In conclusion, data from the present study demonstrated the detrimental effects of Gwagwalada landfill leachate on the brain which may be implicated in neuropsychological conditions.
Collapse
Affiliation(s)
- Usende Ifukibot Levi
- Department of Veterinary Anatomy, University of Abuja, Nigeria; Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA.
| | - Mofio M Bintu
- Department of Biological Sciences, University of Abuja, Abuja, Nigeria
| | | | | | - Adikpe Oluwa Agbonu
- Department of Veterinary Physiology and Biochemistry, University of Abuja, Abuja, Nigeria
| | | | - Enefe Ndidi
- Department of Veterinary Physiology and Biochemistry, University of Abuja, Abuja, Nigeria
| | - Sanni Fatimah Saka
- Department of Veterinary Physiology and Biochemistry, University of Abuja, Abuja, Nigeria
| | - Beselia V Gela
- Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA; Department of Physiology and Pharmacology, Petre Schotadze Tbilisi Medical Academy, Tbilisi, Georgia
| | - Smart I Mbagwu
- Dept of Anatomy, Faculty of Basic Medical Sciences, Nnamdi Azikiwe University, Nigeria
| | - Edem Ekpenyong Edem
- Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Nigeria
| | | | - Connor James
- Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA
| |
Collapse
|
10
|
Fan Z, Gong X, Xu H, Qu Y, Li B, Li L, Yan Y, Wu L, Yan C. Hippocampal parvalbumin and perineuronal nets: Possible involvement in anxiety-like behavior in rats. Hippocampus 2024; 34:156-165. [PMID: 38100162 DOI: 10.1002/hipo.23595] [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: 07/25/2023] [Revised: 11/26/2023] [Accepted: 12/03/2023] [Indexed: 02/20/2024]
Abstract
The excitatory-inhibitory imbalance has been considered an important mechanism underlying stress-related psychiatric disorders. In the present study, rats were exposed to 6 days of inescapable foot shock (IFS) to induce stress. The open field test and elevated plus maze test showed that IFS-exposed rats exhibited increased anxiety-like behavior. Immunofluorescence showed that IFS rats had a decreased density of GAD67-immunoreactive interneurons in the dorsal hippocampal CA1 region, while no significant change in the density of CaMKIIα-immunoreactive glutamatergic neurons was seen. We investigated the expression of different interneuron subtype markers, including parvalbumin (PV), somatostatin (SST), and calretinin (CR), and noted a marked decline in the density of PV-immunoreactive interneurons in the dorsal CA1 region of IFS rats. The perineuronal net (PNN) is a specialized extracellular matrix structure primarily around PV interneurons. We used Wisteria floribunda agglutinin lectin to label the PNNs and observed that IFS rats had an increased proportion of PNN-coated PV-positive interneurons in CA1. The number of PSD95-positive excitatory synaptic puncta on the soma of PNN-free PV-positive interneurons was significantly higher than that of PNN-coated PV-positive interneurons. Our findings suggest that the effect of IFS on the hippocampal GABAergic interneurons could be cell-type-specific. Loss of PV phenotype in the dorsal hippocampal CA1 region may contribute to anxiety in rats. The dysregulated PV-PNN relationship in CA1 after traumatic stress exposure might represent one of the neurobiological correlates of the observed anxiety-like behavior.
Collapse
Affiliation(s)
- Zhixin Fan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiayu Gong
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanfang Xu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Qu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bozhi Li
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanxin Li
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Yan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lili Wu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Can Yan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
11
|
Noel ES, Chen A, Peña YA, Honeycutt JA. Early life adversity drives sex-dependent changes in 5-mC DNA methylation of parvalbumin cells in the prefrontal cortex in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578313. [PMID: 38352518 PMCID: PMC10862911 DOI: 10.1101/2024.01.31.578313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Early life adversity (ELA) can result in increased risk for developing affective disorders, such as anxiety or depression, later in life, with women showing increased risk. Interactions between an individual's genes and their environment play key roles in producing, as well as mitigating, later life neuropathology. Our current understanding of the underlying epigenomic drivers of ELA associated anxiety and depression are limited, and this stems in part from the complexity of underlying biochemical processes associated with how early experiences shapes later life behavior. Epigenetic alterations, or experience-driven modifications to DNA, can be leveraged to understand the interplay between genes and the environment. The present study characterized DNA methylation patterning, assessed via evaluation of 5-methylcytosine (5-mC), following ELA in a Sprague Dawley rat model of ELA induced by early caregiver deprivation. This study utilized maternal separation to investigate sex- and age-specific outcomes of ELA on epigenetic patterning in parvalbumin (PV)-containing interneurons in the prefrontal cortex (PFC), a subpopulation of inhibitory neurons which are associated with ELA and affective dysfunction. While global analysis of 5-mC methylation and CpG site specific pyrosequencing of the PV promoter, Pvalb, showed no obvious effects of ELA, when analyses were restricted to assessing 5-mC intensity in colocalized PV cells, there were significant sex and age dependent effects. We found that ELA leads sex-specific changes in PV cell counts, and that cell counts can be predicted by 5-mC intensity, with males and females showing distinct patterns of methylation and PV outcomes. ELA also produced sex-specific effects in corticosterone reactivity, with juvenile females showing a blunted stress hormone response compared to controls. Overall, ELA led to a sex-specific developmental shift in PV profile, which is comparable to profiles that are seen at a later developmental timepoint, and this shift may be mediated in part by epigenomic alterations driven by altered DNA methylation.
Collapse
Affiliation(s)
- Emma S Noel
- Program in Biochemistry, Brunswick, ME 04011 USA
| | - Alissa Chen
- Program in Neuroscience, Brunswick, ME 04011 USA
| | | | - Jennifer A Honeycutt
- Program in Neuroscience, Brunswick, ME 04011 USA
- Department of Psychology Bowdoin College, Brunswick, ME 04011 USA
| |
Collapse
|
12
|
Brosens N, Lesuis SL, Rao-Ruiz P, van den Oever MC, Krugers HJ. Shaping Memories Via Stress: A Synaptic Engram Perspective. Biol Psychiatry 2023:S0006-3223(23)01720-1. [PMID: 37977215 DOI: 10.1016/j.biopsych.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/09/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Stress modulates the activity of various memory systems and can thereby guide behavioral interaction with the environment in an adaptive or maladaptive manner. At the cellular level, a large body of evidence indicates that (nor)adrenaline and glucocorticoid release induced by acute stress exposure affects synapse function and synaptic plasticity, which are critical substrates for learning and memory. Recent evidence suggests that memories are supported in the brain by sparsely distributed neurons within networks, termed engram cell ensembles. While the physiological and molecular effects of stress on the synapse are increasingly well characterized, how these synaptic modifications shape the multiscale dynamics of engram cell ensembles is still poorly understood. In this review, we discuss and integrate recent information on how acute stress affects synapse function and how this may alter engram cell ensembles and their synaptic connectivity to shape memory strength and memory precision. We provide a mechanistic framework of a synaptic engram under stress and put forward outstanding questions that address knowledge gaps in our understanding of the mechanisms that underlie stress-induced memory modulation.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| | - Sylvie L Lesuis
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Cellular and Cognitive Neuroscience group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Jeanneteau F. Fast signaling by glucocorticoids shapes neural representations of behaviors. Steroids 2023; 199:109294. [PMID: 37549777 DOI: 10.1016/j.steroids.2023.109294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Glucocorticoids are stress hormones that play central roles in the immediate and slower adaptive responses of the brain and body to new behavioral experience. The exact mechanisms by which the rapid and slow processes underlying glucocorticoid mnemonic effects unfold are under intensive scrutiny. It is possible that glucocorticoids rapidly modify memory representations in the brain by interfering with synaptic functions between inhibitory and excitatory neurons in a timing and context dependent manner. In particular, activity-dependent trans-synaptic messengers appear to have all the necessary attributes to engage in the rapid signaling by glucocorticoids and regulate the brain and behaviors. Novel frameworks for the treatment of stress-related disorders could emerge from a better characterization of the dynamic interplay between the rapid and slow signaling components by glucocorticoids on large-scale brain networks. Here I present some of the exact factors that could help reach this objective.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle , Université de Montpellier, INSERM, CNRS, 141 rue de la Cardonille, 34090, Montpellier, France.
| |
Collapse
|
14
|
Aghighi F, Salami M, Talaei SA. Effect of postnatal environmental enrichment on LTP induction in the CA1 area of hippocampus of prenatally traffic noise-stressed female rats. AIMS Neurosci 2023; 10:269-281. [PMID: 38188003 PMCID: PMC10767064 DOI: 10.3934/neuroscience.2023021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 01/09/2024] Open
Abstract
Early-life stress negatively alters mammalian brain programming. Environmental enrichment (EE) has beneficial effects on brain structure and function. This study aimed to evaluate the effects of postnatal environmental enrichment on long-term potentiation (LTP) induction in the hippocampal CA1 area of prenatally stressed female rats. The pregnant Wistar rats were housed in a standard animal room and exposed to traffic noise stress 2 hours/day during the third week of pregnancy. Their offspring either remained intact (ST) or received enrichment (SE) for a month starting from postnatal day 21. The control groups either remained intact (CO) or received enrichment (CE). Basic field excitatory post-synaptic potentials (fEPSPs) were recorded in the CA1 area; then, LTP was induced by high-frequency stimulation. Finally, the serum levels of corticosterone were measured. Our results showed that while the prenatal noise stress decreased the baseline responses of the ST rats when compared to the control rats (P < 0.001), the postnatal EE increased the fEPSPs of both the CE and SE animals when compared to the respective controls. Additionally, high-frequency stimulation (HFS) induced LTP in the fEPSPs of the CO rats (P < 0.001) and failed to induce LTP in the fEPSPs of the ST animals. The enriched condition caused increased potentiation of post-HFS responses in the controls (P < 0.001) and restored the disrupted synaptic plasticity of the CA1 area in the prenatally stressed rats. Likewise, the postnatal EE decreased the elevated serum corticosterone of prenatally stressed offspring (P < 0.001). In conclusion, the postnatal EE restored the stress induced impairment of synaptic plasticity in rats' female offspring.
Collapse
Affiliation(s)
| | | | - Sayyed Alireza Talaei
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I. R. Iran
| |
Collapse
|
15
|
Radahmadi M, Salehifard K, Reisi P. In vivo synaptic potency, short-term and long-term plasticity at the hippocampal Schaffer collateral-CA1 synapses: Role of different light-dark cycles in male rats. Brain Res 2023; 1817:148514. [PMID: 37499734 DOI: 10.1016/j.brainres.2023.148514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The changes in the light-dark(L/D) cycle could modify cellular mechanisms in some brain regions. The present study compared the effects of various L/D cycles on invivo synaptic potency, short-term and long-term plasticity in the hippocampal CA1 area, adrenal glands weight(AGWs), corticosterone (CORT) levels, and body weight differences(BWD) in male rats. Male rats were assigned into different L/D cycle groups: L4/D20, L8/D16, L12/D12(control), L16/D8, and L20/D4. The slope, amplitude, and the area under curve(AUC) related to the field excitatory postsynaptic potentials(fEPSPs) were assessed, using the input-output(I/O) functions, paired-pulse(PP) responses at different interpulse intervals, and after the induction of long-term potentiation(LTP) in the hippocampal CA1 area. Also, the CORT levels, AGWs, and BWDs were measured in all groups. The slope, amplitude, and AUC of fEPSP in the I/O functions, all three phases of PP, before and after the LTP induction, were significantly decreased in all experimental groups, especially in the L20/D4 and L4/D20 groups. As such, the CORT levels and AGWs were significantly increased in all experimental groups, especially in the L20/D4 group. Overall, the uncommon L/D cycles (minimum and particularly maximum durations of light) significantly reduced the cellular mechanism of learning and memory. Also, downtrends were observed in synaptic potency, as well as short-term and long-term plasticity. The changes in PP with high interpulse intervals, or activity of GABAB receptors, were more significant than the changes in other PP phases with different L/D durations. Additionally, the CORT levels, adrenal glands, and body weight gain occurred time-independently concerning different L/D lengths.
Collapse
Affiliation(s)
- Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Yamada J, Maeda S, Tojo M, Hayashida M, Iinuma KM, Jinno S. Altered regulation of oligodendrocytes associated with parvalbumin neurons in the ventral hippocampus underlies fear generalization in male mice. Neuropsychopharmacology 2023; 48:1668-1679. [PMID: 37277574 PMCID: PMC10516901 DOI: 10.1038/s41386-023-01611-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/07/2023]
Abstract
Fear generalization is a neurobiological process by which an organism interprets a novel stimulus as threatening because of its similarity to previously learned fear-inducing stimuli. Because recent studies have suggested that the communication between oligodendrocyte precursor cells (OPCs) and parvalbumin (PV)-expressing GABAergic neurons (PV neurons) may play critical roles in stress-related disorders, we examined the involvement of these cells in fear generalization. We first tested the behavioral characteristics of mouse models for conventional fear conditioning (cFC) and modified FC (mFC) with severe electric foot shocks and found that fear generalization was observed in mice treated with mFC but not in mice treated with cFC. The expression levels of genes related to OPCs, oligodendrocytes (OLs), and myelin in the ventral hippocampus were lower in mFC mice than in cFC mice. The densities of OPCs and OLs were decreased in the ventral hippocampus of mFC mice compared to cFC mice. The myelination ratios of PV neurons in the ventral hippocampus were lower in mFC mice than in cFC mice. The chemogenetic activation of PV neurons in the ventral hippocampus of mFC mice reduced fear generalization. The expression levels of genes related to OPCs, OLs, and myelin were recovered following the activation of PV neurons. Finally, the myelination ratios of PV neurons were increased after the activation of PV neurons. Our results suggest that altered regulation of OLs specifically associated with axons of PV neurons in the ventral hippocampus may underlie the generalization of remote fear memory following severe stress exposure.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Miori Tojo
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Miyuki Hayashida
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
17
|
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
|
18
|
Begega A, Jove CI, López M, Moreno RD. Impact of environmental enrichment on the GABAergic neurons and glucocorticoid receptors in the hippocampus and nucleus accumbens of Wistar rats: pro-resilient effects. Brain Res Bull 2023; 200:110699. [PMID: 37406885 DOI: 10.1016/j.brainresbull.2023.110699] [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] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023]
Abstract
The unpredictable chronic mild stress (UCMS) model has been used to induce depressive-like symptoms in animal models. Our work aims to evaluate the impact of environmental enrichment on male Wistar rats in an animal model for depression. For this purpose, we aim to assess changes in GR and GABAergic (PV+) density in cerebral regions related to cognitive-affective processes associated with depressive disorder, such as the dorsal- ventral hippocampus and accumbens nuclei. Three groups of rats were used: UCMs (unpredictable chronic mild stress), EE+ UCMs (enrichment + stress) and CONT (behavioral tests only). Hedonic responses elicited by sucrose solution were examined by licking behavior analysis; the anxiety level was evaluated using the elevated zero maze and the forced swimming (passive coping) tests. The environmental enrichment reduced the effects of chronic stress, promoting greater resilience. Thus, the UCMs group showed an anhedonia response, more anxiety and immobility behavior than either the control or the EE+ UCMs groups. Regarding immunochemistry results, there was a reduction in GABAergic activity coupled with increased activation of GR in UCMs in the dorsal hippocampus, but there were no differences between groups in the ventral hippocampus. These results suggest environmental enrichment could enhance greater resilience, reducing the vulnerability of the subjects to develop disorders such as depression and anxiety.
Collapse
Affiliation(s)
- Azucena Begega
- Laboratory of Neuroscience. Faculty of Psychology. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain; Institute of Neuroscience of Principado Asturias, INEUROPA. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain.
| | - Claudia I Jove
- Laboratory of Neuroscience. Faculty of Psychology. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain
| | - Matías López
- Institute of Neuroscience of Principado Asturias, INEUROPA. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain; Basic Psychology Area. Faculty of Psychology. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain
| | - Román-Darío Moreno
- Faculty of Education and Psychology. University Francisco de Vitoria, Pozuelo de Alarcón, 28223. Madrid, Spain
| |
Collapse
|
19
|
Li X, Du ZJ, Xu JN, Liang ZM, Lin S, Chen H, Li SJ, Li XW, Yang JM, Gao TM. mGluR5 in hippocampal CA1 pyramidal neurons mediates stress-induced anxiety-like behavior. Neuropsychopharmacology 2023; 48:1164-1174. [PMID: 36797374 PMCID: PMC10267178 DOI: 10.1038/s41386-023-01548-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/18/2023] [Accepted: 02/06/2023] [Indexed: 02/18/2023]
Abstract
Pharmacological manipulation of mGluR5 has showed that mGluR5 is implicated in the pathophysiology of anxiety and mGluR5 has been proposed as a potential drug target for anxiety disorders. Nevertheless, the mechanism underlying the mGluR5 involvement in stress-induced anxiety-like behavior remains largely unknown. Here, we found that chronic restraint stress induced anxiety-like behavior and decreased the expression of mGluR5 in hippocampal CA1. Specific knockdown of mGluR5 in hippocampal CA1 pyramidal neurons produced anxiety-like behavior. Furthermore, both chronic restraint stress and mGluR5 knockdown impaired inhibitory synaptic inputs in hippocampal CA1 pyramidal neurons. Notably, positive allosteric modulator of mGluR5 rescued stress-induced anxiety-like behavior and restored the inhibitory synaptic inputs. These findings point to an essential role for mGluR5 in hippocampal CA1 pyramidal neurons in mediating stress-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuo-Jun Du
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun-Nan Xu
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhi-Man Liang
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Song Lin
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hao Chen
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shu-Ji Li
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Rubinstein MR, Burgueño AL, Quiroga S, Wald MR, Genaro AM. Current Understanding of the Roles of Gut-Brain Axis in the Cognitive Deficits Caused by Perinatal Stress Exposure. Cells 2023; 12:1735. [PMID: 37443769 PMCID: PMC10340286 DOI: 10.3390/cells12131735] [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: 04/17/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.
Collapse
Affiliation(s)
- Mara Roxana Rubinstein
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| | | | | | | | - Ana María Genaro
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| |
Collapse
|
21
|
Shi HJ, Wang S, Wang XP, Zhang RX, Zhu LJ. Hippocampus: Molecular, Cellular, and Circuit Features in Anxiety. Neurosci Bull 2023; 39:1009-1026. [PMID: 36680709 PMCID: PMC10264315 DOI: 10.1007/s12264-023-01020-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/13/2022] [Indexed: 01/22/2023] Open
Abstract
Anxiety disorders are currently a major psychiatric and social problem, the mechanisms of which have been only partially elucidated. The hippocampus serves as a major target of stress mediators and is closely related to anxiety modulation. Yet so far, its complex anatomy has been a challenge for research on the mechanisms of anxiety regulation. Recent advances in imaging, virus tracking, and optogenetics/chemogenetics have permitted elucidation of the activity, connectivity, and function of specific cell types within the hippocampus and its connected brain regions, providing mechanistic insights into the elaborate organization of the hippocampal circuitry underlying anxiety. Studies of hippocampal neurotransmitter systems, including glutamatergic, GABAergic, cholinergic, dopaminergic, and serotonergic systems, have contributed to the interpretation of the underlying neural mechanisms of anxiety. Neuropeptides and neuroinflammatory factors are also involved in anxiety modulation. This review comprehensively summarizes the hippocampal mechanisms associated with anxiety modulation, based on molecular, cellular, and circuit properties, to provide tailored targets for future anxiety treatment.
Collapse
Affiliation(s)
- Hu-Jiang Shi
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shuang Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xin-Ping Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui-Xin Zhang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China.
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| |
Collapse
|
22
|
Gandhi T, Canepa CR, Adeyelu TT, Adeniyi PA, Lee CC. Neuroanatomical Alterations in the CNTNAP2 Mouse Model of Autism Spectrum Disorder. Brain Sci 2023; 13:891. [PMID: 37371370 DOI: 10.3390/brainsci13060891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is associated with neurodevelopmental alterations, including atypical forebrain cellular organization. Mutations in several ASD-related genes often result in cerebral cortical anomalies, such as the abnormal developmental migration of excitatory pyramidal cells and the malformation of inhibitory neuronal circuitry. Notably here, mutations in the CNTNAP2 gene result in ectopic superficial cortical neurons stalled in lower cortical layers and alterations to the balance of cortical excitation and inhibition. However, the broader circuit-level implications of these findings have not been previously investigated. Therefore, we assessed whether ectopic cortical neurons in CNTNAP2 mutant mice form aberrant connections with higher-order thalamic nuclei, potentially accounting for some autistic behaviors, such as repetitive and hyperactive behaviors. Furthermore, we assessed whether the development of parvalbumin-positive (PV) cortical interneurons and their specialized matrix support structures, called perineuronal nets (PNNs), were altered in these mutant mice. We found alterations in both ectopic neuronal connectivity and in the development of PNNs, PV neurons and PNNs enwrapping PV neurons in various sensory cortical regions and at different postnatal ages in the CNTNAP2 mutant mice, which likely lead to some of the cortical excitation/inhibition (E/I) imbalance associated with ASD. These findings suggest neuroanatomical alterations in cortical regions that underlie the emergence of ASD-related behaviors in this mouse model of the disorder.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70806, USA
| | - Cade R Canepa
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70806, USA
| | - Tolulope T Adeyelu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70806, USA
| | - Philip A Adeniyi
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70806, USA
| | - Charles C Lee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70806, USA
| |
Collapse
|
23
|
Gandhi T, Liu CC, Adeyelu TT, Canepa CR, Lee CC. Behavioral regulation by perineuronal nets in the prefrontal cortex of the CNTNAP2 mouse model of autism spectrum disorder. Front Behav Neurosci 2023; 17:1114789. [PMID: 36998537 PMCID: PMC10043266 DOI: 10.3389/fnbeh.2023.1114789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
Autism spectrum disorders (ASDs) arise from altered development of the central nervous system, and manifest behaviorally as social interaction deficits and restricted and repetitive behaviors. Alterations to parvalbumin (PV) expressing interneurons have been implicated in the neuropathological and behavioral deficits in autism. In addition, perineuronal nets (PNNs), specialized extracellular matrix structures that enwrap the PV-expressing neurons, also may be altered, which compromises neuronal function and susceptibility to oxidative stress. In particular, the prefrontal cortex (PFC), which regulates several core autistic traits, relies on the normal organization of PNNs and PV-expressing cells, as well as other neural circuit elements. Consequently, we investigated whether PNNs and PV-expressing cells were altered in the PFC of the CNTNAP2 knockout mouse model of ASD and whether these contributed to core autistic-like behaviors in this model system. We observed an overexpression of PNNs, PV-expressing cells, and PNNs enwrapping PV-expressing cells in adult CNTNAP2 mice. Transient digestion of PNNs from the prefrontal cortex (PFC) by injection of chondroitinase ABC in CNTNAP2 mutant mice rescued some of the social interaction deficits, but not the restricted and repetitive behaviors. These findings suggest that the neurobiological regulation of PNNs and PVs in the PFC contribute to social interaction behaviors in neurological disorders including autism.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Chin-Chi Liu
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Tolulope T. Adeyelu
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Cade R. Canepa
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
24
|
Omoumi S, Rashidy-Pour A, Seyedinia SA, Tarahomi P, Vafaei AA, Raise-Abdullahi P. Corticosterone injection into the infralimbic prefrontal cortex enhances fear memory extinction: Involvement of GABA receptors and the extracellular signal-regulated kinase. Physiol Behav 2023; 265:114156. [PMID: 36918107 DOI: 10.1016/j.physbeh.2023.114156] [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: 01/29/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023]
Abstract
This study investigated the interactive effect of glucocorticoid and Gamma-aminobutyric acid (GABA) receptors in the Infralimbic (IL) cortex on fear extinction in rats' auditory fear conditioning task (AFC). Animals received 3 conditioning trial tones (conditioned stimulus, 30 s, 4 kHz, 80 dB) co-terminated with a footshock (unconditioned stimulus, 0.8 mA, 1 s). Extinction testing was conducted over 3 days (Ext 1-3) after conditioning. Intra-IL injection of corticosterone (CORT, 20 ng/0.3 µl/side) was performed 15 min before the first extinction trial (Ext 1) which attenuated auditory fear expression in subsequent extinction trials (Ext 1-3), demonstrating fear memory extinction enhancement. Co-injection of the GABAA agonist muscimol (250 ng/0.3 µl/side) or the GABAB agonist baclofen (250 ng/0.3 µl/side) 15 min before corticosterone, did not significantly affect the facilitative effects of corticosterone on fear extinction. However, co-injection of the GABAA antagonist bicuculline (BIC, 100 ng/0.3 µl/side) or the GABAB antagonist CGP35348 (CGP, 100 ng/0.3 µl/side) 15 min before corticosterone, blocked the facilitative effects of corticosterone on fear extinction. Moreover, extracellular signal-regulated kinase (ERK) and cAMP response element-binding (CREB) in the IL were examined by Western blotting analysis after the first extinction trial (Ext 1) in some groups. Intra-IL injection of corticosterone increased the ERK activity but not CREB. Co-injection of the bicuculline or CGP35348 blocked the enhancing effect of corticosterone on ERK expression in the IL. Glucocorticoid receptors (GRs) activation in the IL cortex by corticosterone increased ERK activity and facilitated fear extinction. GABAA or GABAB antagonists decreased ERK activity and inhibited corticosterone's effect. GRs and GABA receptors in the IL cortex jointly modulate the fear extinction processes via the ERK pathway. This pre-clinical animal study may highlight GRs and GABA interactions in the IL cortex modulating fear memory processes in fear-related disorders such as post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Samira Omoumi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed Ali Seyedinia
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Student Research Committee, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Parnia Tarahomi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Student Research Committee, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | | |
Collapse
|
25
|
Jeon YS, Jeong D, Kweon H, Kim JH, Kim CY, Oh Y, Lee YH, Kim CH, Kim SG, Jeong JW, Kim E, Lee SH. Adolescent Parvalbumin Expression in the Left Orbitofrontal Cortex Shapes Sociability in Female Mice. J Neurosci 2023; 43:1555-1571. [PMID: 36717231 PMCID: PMC10008055 DOI: 10.1523/jneurosci.0918-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 12/27/2022] [Accepted: 01/05/2023] [Indexed: 01/31/2023] Open
Abstract
The adolescent social experience is essential for the maturation of the prefrontal cortex in mammalian species. However, it still needs to be determined which cortical circuits mature with such experience and how it shapes adult social behaviors in a sex-specific manner. Here, we examined social-approaching behaviors in male and female mice after postweaning social isolation (PWSI), which deprives social experience during adolescence. We found that the PWSI, particularly isolation during late adolescence, caused an abnormal increase in social approaches (hypersociability) only in female mice. We further found that the PWSI female mice showed reduced parvalbumin (PV) expression in the left orbitofrontal cortex (OFCL). When we measured neural activity in the female OFCL, a substantial number of neurons showed higher activity when mice sniffed other mice (social sniffing) than when they sniffed an object (object sniffing). Interestingly, the PWSI significantly reduced both the number of activated neurons and the activity level during social sniffing in female mice. Similarly, the CRISPR/Cas9-mediated knockdown of PV in the OFCL during late adolescence enhanced sociability and reduced the social sniffing-induced activity in adult female mice via decreased excitability of PV+ neurons and reduced synaptic inhibition in the OFCL Moreover, optogenetic activation of excitatory neurons or optogenetic inhibition of PV+ neurons in the OFCL enhanced sociability in female mice. Our data demonstrate that the adolescent social experience is critical for the maturation of PV+ inhibitory circuits in the OFCL; this maturation shapes female social behavior via enhancing social representation in the OFCL SIGNIFICANCE STATEMENT Adolescent social isolation often changes adult social behaviors in mammals. Yet, we do not fully understand the sex-specific effects of social isolation and the brain areas and circuits that mediate such changes. Here, we found that adolescent social isolation causes three abnormal phenotypes in female but not male mice: hypersociability, decreased PV+ neurons in the left orbitofrontal cortex (OFCL), and decreased socially evoked activity in the OFCL Moreover, parvalbumin (PV) deletion in the OFCL in vivo caused the same phenotypes in female mice by increasing excitation compared with inhibition within the OFCL Our data suggest that adolescent social experience is required for PV maturation in the OFCL, which is critical for evoking OFCL activity that shapes social behaviors in female mice.
Collapse
Affiliation(s)
- Yi-Seon Jeon
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Daun Jeong
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Hanseul Kweon
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, KAIST, Daejeon 34141, Korea
| | - Jae-Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Choong Yeon Kim
- School of Electrical Engineering, KAIST, Daejeon 34141, Korea
| | - Youngbin Oh
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Young-Ho Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Chan Hyuk Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Sang-Gyu Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Jae-Woong Jeong
- School of Electrical Engineering, KAIST, Daejeon 34141, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, KAIST, Daejeon 34141, Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, KAIST, Daejeon 34141, Korea
| |
Collapse
|
26
|
Woodward E, Rangel-Barajas C, Ringland A, Logrip ML, Coutellier L. Sex-Specific Timelines for Adaptations of Prefrontal Parvalbumin Neurons in Response to Stress and Changes in Anxiety- and Depressive-Like Behaviors. eNeuro 2023; 10:ENEURO.0300-22.2023. [PMID: 36808099 PMCID: PMC9997696 DOI: 10.1523/eneuro.0300-22.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 02/22/2023] Open
Abstract
Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.
Collapse
Affiliation(s)
- Emma Woodward
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Claudia Rangel-Barajas
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Amanda Ringland
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
| | - Marian L Logrip
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Laurence Coutellier
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
27
|
Woodward EM, Ringland A, Ackerman J, Coutellier L. Prepubertal ovariectomy confers resilience to stress-induced anxiety in adult female mice. Psychoneuroendocrinology 2023; 148:105997. [PMID: 36470154 PMCID: PMC9898172 DOI: 10.1016/j.psyneuen.2022.105997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
The increased vulnerability to stress-induced neuropsychiatric disorders in women, including anxiety disorders, does not emerge until pubertal onset, suggesting a role for ovarian hormones in organizing sex-specific vulnerability to anxiety. Parvalbumin (PV) interneurons in the prefrontal cortex are a potential target for these ovarian hormones. PV+ interneurons undergo maturation during the adolescent period and have been shown to be sensitive to stress and to mediate stress-induced anxiety in female mice. To test the idea that ovarian hormones at puberty are necessary for the acquisition of sensitivity to stress, hypothetically driving the response of PV+ interneurons to stress, we performed ovariectomy or sham surgery before pubertal onset in female mice. These mice then were exposed to four weeks of unpredictable chronic mild stress in adulthood. We then assessed anxiety-like behavior and PV/FosB colocalization in the medial PFC. Additionally, we assessed stress-induced anxiety-like behavior in female mice following ovariectomy in adulthood to determine if puberty is a sensitive period for ovarian hormones in mediating vulnerability to stress. We found that prepubertal ovariectomy protects against the development of anxiety-like behavior in adulthood, an effect not found following ovariectomy in adulthood. This effect may be independent of ovarian hormones on prefrontal PV+ interneurons response to stress.
Collapse
Affiliation(s)
- Emma M Woodward
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Amanda Ringland
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA; Department of Psychology, Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA.
| | - Jennifer Ackerman
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA; Department of Psychology, Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA.
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA; Department of Psychology, Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
28
|
Jeanneteau F, Meijer OC, Moisan MP. Structural basis of glucocorticoid receptor signaling bias. J Neuroendocrinol 2023; 35:e13203. [PMID: 36221223 DOI: 10.1111/jne.13203] [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: 05/21/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
Dissociation between the healthy and toxic effects of cortisol, a major stress-responding hormone has been a widely used strategy to develop anti-inflammatory glucocorticoids with fewer side effects. Such strategy falls short when treating brain disorders as timing and activity state within large-scale neuronal networks determine the physiological and behavioral specificity of cortisol response. Advances in structural molecular dynamics posit the bases for engineering glucocorticoids with precision bias for select downstream signaling pathways. Design of allosteric and/or cooperative control for the glucocorticoid receptor could help promote the beneficial and reduce the deleterious effects of cortisol on brain and behavior in disease conditions.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Onno C Meijer
- Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
29
|
Wieser M, Beckmann KM, Kutter APN, Mauri N, Richter H, Zölch N, Bektas RN. Ketamine administration in idiopathic epileptic and healthy control dogs: Can we detect differences in brain metabolite response with spectroscopy? Front Vet Sci 2023; 9:1093267. [PMID: 36686158 PMCID: PMC9853535 DOI: 10.3389/fvets.2022.1093267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction In recent years ketamine has increasingly become the focus of multimodal emergency management for epileptic seizures. However, little is known about the effect of ketamine on brain metabolites in epileptic patients. Magnetic resonance spectroscopy (MRS) is a non-invasive technique to estimate brain metabolites in vivo. Our aim was to measure the effect of ketamine on thalamic metabolites in idiopathic epileptic (IE) dogs using 3 Tesla MRS. We hypothesized that ketamine would increase the glutamine-glutamate (GLX)/creatine ratio in epileptic dogs with and without antiseizure drug treatment, but not in control dogs. Furthermore, we hypothesized that no different responses after ketamine administration in other measured brain metabolite ratios between the different groups would be detected. Methods In this controlled prospective experimental trial IE dogs with or without antiseizure drug treatment and healthy client-owned relatives of the breeds Border Collie and Greater Swiss Mountain Dog, were included. After sedation with butorphanol, induction with propofol and maintenance with sevoflurane in oxygen and air, a single voxel MRS at the level of the thalamus was performed before and 2 min after intravenous administration of 1 mg/kg ketamine. An automated data processing spectral fitting linear combination model algorithm was used to estimate all commonly measured metabolite ratios. A mixed ANOVA with the independent variables ketamine administration and group allocation was performed for all measured metabolites. A p < 0.05 was considered statistically significant. Results Twelve healthy control dogs, 10 untreated IE and 12 treated IE dogs were included. No significant effects for GLX/creatine were found. However, increased glucose/creatine ratios were found (p < 0.001) with no effect of group allocation. Furthermore, increases in the GABA/creatine ratio were found in IEU dogs. Discussion MRS was able to detect changes in metabolite/creatine ratios after intravenous administration of 1 mg/kg ketamine in dogs and no evidence was found that excitatory effects are induced in the thalamus. Although it is beyond the scope of this study to investigate the antiseizure potential of ketamine in dogs, results of this research suggest that the effect of ketamine on the brain metabolites could be dependent on the concentrations of brain metabolites before administration.
Collapse
Affiliation(s)
- Manuela Wieser
- Section of Anesthesiology, Department of Clinical Diagnostics and Services, University of Zurich, Zurich, Switzerland,*Correspondence: Manuela Wieser ✉
| | | | - Annette P. N. Kutter
- Section of Anesthesiology, Department of Clinical Diagnostics and Services, University of Zurich, Zurich, Switzerland
| | - Nico Mauri
- Department of Clinical Diagnostics and Services, Clinic for Diagnostic Imaging, University of Zurich, Zurich, Switzerland,Vetimage Diagnostik AG, Oberentfelden, Switzerland
| | - Henning Richter
- Department of Clinical Diagnostics and Services, Clinic for Diagnostic Imaging, University of Zurich, Zurich, Switzerland
| | - Niklaus Zölch
- Department of Forensic Medicine and Imaging, Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Rima Nadine Bektas
- Section of Anesthesiology, Department of Clinical Diagnostics and Services, University of Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Forro T, Volitaki E, Malagon-Vina H, Klausberger T, Nevian T, Ciocchi S. Anxiety-related activity of ventral hippocampal interneurons. Prog Neurobiol 2022; 219:102368. [PMID: 36273721 DOI: 10.1016/j.pneurobio.2022.102368] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 12/04/2022]
Abstract
Anxiety is an aversive mood reflecting the anticipation of potential threats. The ventral hippocampus (vH) is a key brain region involved in the genesis of anxiety responses. Recent studies have shown that anxiety is mediated by the activation of vH pyramidal neurons targeting various limbic structures. Throughout the cortex, the activity of pyramidal neurons is controlled by GABA-releasing inhibitory interneurons and the GABAergic system represents an important target of anxiolytic drugs. However, how the activity of vH inhibitory interneurons is related to different anxiety behaviours has not been investigated so far. Here, we integrated in vivo electrophysiology with behavioural phenotyping of distinct anxiety exploration behaviours in rats. We showed that pyramidal neurons and interneurons of the vH are selectively active when animals explore specific compartments of the elevated-plus-maze (EPM), an anxiety task for rodents. Moreover, rats with prior goal-related experience exhibited low-anxiety exploratory behaviour and showed a larger trajectory-related activity of vH interneurons during EPM exploration compared to high anxiety rats. Finally, in low anxiety rats, trajectory-related vH interneurons exhibited opposite activity to pyramidal neurons specifically in the open arms (i.e. more anxiogenic) of the EPM. Our results suggest that vH inhibitory micro-circuits could act as critical elements underlying different anxiety states.
Collapse
|
31
|
Kong Y, Kuss M, Shi Y, Fang F, Xue W, Shi W, Liu Y, Zhang C, Zhong P, Duan B. Exercise facilitates regeneration after severe nerve transection and further modulates neural plasticity. Brain Behav Immun Health 2022; 26:100556. [PMID: 36405423 PMCID: PMC9673108 DOI: 10.1016/j.bbih.2022.100556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with severe traumatic peripheral nerve injury (PNI) always suffer from incomplete recovery and poor functional outcome. Physical exercise-based rehabilitation, as a non-invasive interventional strategy, has been widely acknowledged to improve PNI recovery by promoting nerve regeneration and relieving pain. However, effects of exercise on chronic plastic changes following severe traumatic PNIs have been limitedly discussed. In this study, we created a long-gap sciatic nerve transection followed by autograft bridging in rats and tested the therapeutic functions of treadmill running with low intensity and late initiation. We demonstrated that treadmill running effectively facilitated nerve regeneration and prevented muscle atrophy and thus improved sensorimotor functions and walking performance. Furthermore, exercise could reduce inflammation at the injured nerve as well as prevent the overexpression of TRPV1, a pain sensor, in primary afferent sensory neurons. In the central nervous system, we found that PNI induced transcriptive changes at the ipsilateral lumber spinal dorsal horn, and exercise could reverse the differential expression for genes involved in the Notch signaling pathway. In addition, through neural imaging techniques, we found volumetric, microstructural, metabolite, and neuronal activity changes in supraspinal regions of interest (i.e., somatosensory cortex, motor cortex, hippocampus, etc.) after the PNI, some of which could be reversed through treadmill running. In summary, treadmill running with late initiation could promote recovery from long-gap nerve transection, and while it could reverse maladaptive plasticity after the PNI, exercise may also ameliorate comorbidities, such as chronic pain, mental depression, and anxiety in the long term.
Collapse
Affiliation(s)
- Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yu Shi
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| | - Fang Fang
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wen Xue
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yutong Liu
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
- Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Peng Zhong
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
32
|
Li X, Sun H, Zhu Y, Wang F, Wang X, Han L, Cui D, Luo D, Zhai Y, Zhuo L, Xu X, Yang J, Li Y. Dysregulation of prefrontal parvalbumin interneurons leads to adult aggression induced by social isolation stress during adolescence. Front Mol Neurosci 2022; 15:1010152. [PMID: 36267698 PMCID: PMC9577330 DOI: 10.3389/fnmol.2022.1010152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Social isolation during the juvenile stage results in structural and functional impairment of the brain and deviant adult aggression. However, the specific subregions and cell types that underpin this deviant behavior are still largely unknown. Here, we found that adolescent social isolation led to a shortened latency to attack onset and extended the average attack time, accompanied by anxiety-like behavior and deficits in social preference in adult mice. However, when exposed to social isolation during adulthood, the mice did not show these phenotypes. We also found that the structural plasticity of prefrontal pyramidal neurons, including the dendritic complexity and spine ratio, was impaired in mice exposed to adolescent social isolation. The parvalbumin (PV) interneurons in the prefrontal infralimbic cortex (IL) are highly vulnerable to juvenile social isolation and exhibit decreased cell numbers and reduced activation in adulthood. Moreover, chemogenetic inactivation of IL-PV interneurons can mimic juvenile social isolation-induced deviant aggression and social preference. Conversely, artificial activation of IL-PV interneurons significantly attenuated deviant aggression and rescued social preference during adulthood in mice exposed to adolescent social isolation. These findings implicate juvenile social isolation-induced damage to IL-PV interneurons in long-term aggressive behavior in adulthood.
Collapse
Affiliation(s)
- Xinyang Li
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Huan Sun
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuanyuan Zhu
- Department of Neurobiology, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Feidi Wang
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaodan Wang
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lin Han
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Dongqi Cui
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Danlei Luo
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yifang Zhai
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lixia Zhuo
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiangzhao Xu
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jian Yang
- Department of Diagnostic Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Jian Yang,
| | - Yan Li
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Yan Li,
| |
Collapse
|
33
|
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
|
34
|
Perić I, Lješević M, Beškoski V, Nikolić M, Filipović D. Metabolomic profiling relates tianeptine effectiveness with hippocampal GABA, myo-inositol, cholesterol, and fatty acid metabolism restoration in socially isolated rats. Psychopharmacology (Berl) 2022; 239:2955-2974. [PMID: 35776189 DOI: 10.1007/s00213-022-06180-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 06/16/2022] [Indexed: 12/28/2022]
Abstract
RATIONALE Discovering biomarkers of major depressive disorder (MDD) can give a deeper understanding of this mood disorder and improve the ability to screen for, diagnose, and treat MDD. OBJECTIVES In this study, metabolomics was used in unraveling metabolite fluctuations of MDD and drug outcome by creating specific metabolomic fingerprints. We report metabolomic patterns of change of the hippocampus of adult male Wistar rats following chronic social isolation (CSIS) (6 weeks), an animal model of depression, and/or chronic tianeptine (Tian) treatment (10 mg kg-1 per day) (lasting 3 weeks of 6-week CSIS), monitored by using comprehensive GC × GC-MS. RESULTS The comparative metabolomic analysis highlighted the role of gamma aminobutyric acid (GABA), iso-allocholate, and unsaturated fatty acid metabolism alterations following the CSIS, which was corroborated with moderate to strong negative Pearson's correlation of GABA, docosahexaenoic, 9-hexadecenoic acid, 5,8,11,14-eicosatetraynoic, and arachidonic acids with immobility behavior in the forced swim test. The antidepressant effect of Tian restored GABA levels, which was absent in Tian resilient rats. Tian decreased myo-inositol and increased TCA cycle intermediates, amino acids, and cholesterol and its metabolite. As key molecules of divergence between Tian effectiveness and resilience, metabolomics revealed myo-inositol, GABA, cholesterol, and its metabolite. A significant moderate positive correlation between myo-inositol and immobility was revealed. Tian probably acted by upregulating NMDAR's and α2 adrenergic receptors (AR) or norepinephrine transporter in both control and stressed animals. CONCLUSION Metabolomics revealed several dysregulations underlying CSIS-induced depressive-like behavior and responsiveness to Tian, predominantly converging into NMDAR-mediated glutamate and myo-inositol signalization and GABA inhibitory pathways.
Collapse
Affiliation(s)
- Ivana Perić
- Department of Molecular Biology and Endocrinology, "VINČA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11351, Vinča, Belgrade, Serbia
| | - Marija Lješević
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000, Belgrade, Serbia
| | - Vladimir Beškoski
- Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, 11000, Belgrade, Serbia
| | - Milan Nikolić
- Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, 11000, Belgrade, Serbia
| | - Dragana Filipović
- Department of Molecular Biology and Endocrinology, "VINČA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11351, Vinča, Belgrade, Serbia.
| |
Collapse
|
35
|
Cole AB, Montgomery K, Bale TL, Thompson SM. What the hippocampus tells the HPA axis: Hippocampal output attenuates acute stress responses via disynaptic inhibition of CRF+ PVN neurons. Neurobiol Stress 2022; 20:100473. [PMID: 35982732 PMCID: PMC9379952 DOI: 10.1016/j.ynstr.2022.100473] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
The hippocampus exerts inhibitory feedback on the release of glucocorticoids. Because the major hippocampal efferent projections are excitatory, it has been hypothesized that this inhibition is mediated by populations of inhibitory neurons in the hypothalamus or elsewhere. These regions would be excited by hippocampal efferents and project to corticotropin-releasing factor (CRF) cells in the paraventricular nucleus of the hypothalamus (PVN). A direct demonstration of the synaptic responses elicited by hippocampal outputs in PVN cells or upstream GABAergic interneurons has not been provided previously. Here, we used viral vectors to express channelrhodopsin (ChR) and enhanced yellow fluorescent protein (EYFP) in pyramidal cells in the ventral hippocampus (vHip) in mice expressing tdTomato in GABA- or CRF-expressing neurons. We observed dense innervation of the bed nucleus of the stria terminalis (BNST) by labeled vHip axons and sparse labeling within the PVN. Using whole-cell voltage-clamp recording in parasagittal brain slices containing the BNST and PVN, photostimulation of vHip terminals elicited rapid excitatory postsynaptic currents (EPSCs) and longer-latency inhibitory postsynaptic currents (IPSCs) in both CRF+ and GAD + cells. The ratio of synaptic excitation and inhibition was maintained in CRF + cells during 20 Hz stimulus trains. Photostimulation of hippocampal afferents to the BNST and PVN in vivo inhibited the rise in blood glucocorticoid levels produced by acute restraint stress. We thus provide functional evidence suggesting that hippocampal output to the BNST contributes to a net inhibition of the hypothalamic-pituitary axis, providing further mechanistic insights into this process using methods with enhanced spatial and temporal resolution.
Collapse
Affiliation(s)
- Anthony B. Cole
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Medical Scientist Training Program, Departments of University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kristen Montgomery
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tracy L. Bale
- Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Scott M. Thompson
- Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, USA
| |
Collapse
|
36
|
Usende IL, Olopade JO, Azeez IA, Andrioli A, Bankole MO, Olopade FE, Nafady AA, Bentivoglio M. Neuroecotoxicology: Effects of environmental heavy metal exposure on the brain of African giant rats and the contribution of vanadium to the neuropathology. IBRO Neurosci Rep 2022; 13:215-234. [PMID: 36590095 PMCID: PMC9795313 DOI: 10.1016/j.ibneur.2022.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023] Open
Abstract
Increased exploitation of minerals has led to pollution of confined environments as documented in Nigeria Niger Delta. Information on the effects on brain of such exposure is limited. Due to its exploratory activities, the African giant rat (Cricetomys gambianus) (AGR) provides a unique model for neuroecotoxicological research to determine levels of animal and human exposure to different pollutants. This study aims to unravel neuropathological features of AGR sampled from three agro-ecological zones of Nigeria. Fifteen AGR were sampled according to previously determined data on heavy metal exposure: high vanadium, high lead, and low metals. Eighteen AGR were collected from low metal zone and divided into two groups. Control group received vehicle while SMV exposed group received 3 mg/kg sodium metavanadate (SMV) intraperitoneally for 14days. Brain immunohistochemical analyses were conducted, and ultrastructural changes were studied in experimentally exposed group. Results showed significant loss of tyrosin hydroxylase, parvalbumin, orexin-A and melanin concentration hormone containing neuronal populations in brains obtained from high vanadium and high lead zones and in experimentally intoxicated SMV groups. Similarly, significant decrease numbers of dendritic arborations; extracellular matrix density, perineuronal nets; astrocytes and microglia activations are documented in same groups. Ultrastructural studies revealed mass denudation, cilia loss, disintegration of ependymal layer and intense destructions of myelin sheaths in SMV exposed group. These are the first "neuroecotoxicological" findings in distinct neuronal cells. The implications of these findings are highly relevant for human population living in these areas, not only in Nigeria but also in similarly polluted areas elsewhere in the world.
Collapse
Affiliation(s)
- Ifukibot Levi Usende
- Department of Veterinary Anatomy, University of Abuja, Nigeria,Department of Veterinary Anatomy, University of Ibadan, Nigeria,Corresponding author at: Department of Veterinary Anatomy, University of Abuja, Nigeria.
| | | | | | - Anna Andrioli
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - Molakun O. Bankole
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | | | - Allam A. Nafady
- Department of Veterinary Pathology, Electron Microscope Unit, Assuit University, Egypt
| | - Marina Bentivoglio
- Department of Neurological and Movement Sciences, University of Verona, Italy
| |
Collapse
|
37
|
Sinha P, Verma B, Ganesh S. Age-Dependent Reduction in the Expression Levels of Genes Involved in Progressive Myoclonus Epilepsy Correlates with Increased Neuroinflammation and Seizure Susceptibility in Mouse Models. Mol Neurobiol 2022; 59:5532-5548. [PMID: 35732865 DOI: 10.1007/s12035-022-02928-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022]
Abstract
Brain aging is characterized by a gradual decline in cellular homeostatic processes, thereby losing the ability to respond to physiological stress. At the anatomical level, the aged brain is characterized by degenerating neurons, proteinaceous plaques and tangles, intracellular deposition of glycogen, and elevated neuroinflammation. Intriguingly, such age-associated changes are also seen in neurodegenerative disorders suggesting that an accelerated aging process could be one of the contributory factors for the disease phenotype. Amongst these, the genetic forms of progressive myoclonus epilepsy (PME), resulting from loss-of-function mutations in genes, manifest symptoms that are common to age-associated disorders, and genes mutated in PME are involved in the cellular homeostatic processes. Intriguingly, the incidence and/or onset of epileptic seizures are known to increase with age, suggesting that physiological changes in the aged brain might contribute to increased susceptibility to seizures. We, therefore, hypothesized that the expression level of genes implicated in PME might decrease with age, thereby leading to a compromised neuronal response towards physiological stress and hence neuroinflammation in the aging brain. Using mice models, we demonstrate here that the expression level of PME genes shows an inverse correlation with age, neuroinflammation, and compromised heat shock response. We further show that the pharmacological suppression of neuroinflammation ameliorates seizure susceptibility in aged animals as well as in animal models for a PME. Taken together, our results indicate a functional role for the PME genes in normal brain aging and that neuroinflammation could be a major contributory player in susceptibility to seizures.
Collapse
Affiliation(s)
- Priyanka Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Bhupender Verma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India. .,Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
| |
Collapse
|
38
|
Komoltsev IG, Gulyaeva NV. Brain Trauma, Glucocorticoids and Neuroinflammation: Dangerous Liaisons for the Hippocampus. Biomedicines 2022; 10:biomedicines10051139. [PMID: 35625876 PMCID: PMC9138485 DOI: 10.3390/biomedicines10051139] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/30/2022] [Accepted: 05/13/2022] [Indexed: 12/02/2022] Open
Abstract
Glucocorticoid-dependent mechanisms of inflammation-mediated distant hippocampal damage are discussed with a focus on the consequences of traumatic brain injury. The effects of glucocorticoids on specific neuronal populations in the hippocampus depend on their concentration, duration of exposure and cell type. Previous stress and elevated level of glucocorticoids prior to pro-inflammatory impact, as well as long-term though moderate elevation of glucocorticoids, may inflate pro-inflammatory effects. Glucocorticoid-mediated long-lasting neuronal circuit changes in the hippocampus after brain trauma are involved in late post-traumatic pathology development, such as epilepsy, depression and cognitive impairment. Complex and diverse actions of the hypothalamic–pituitary–adrenal axis on neuroinflammation may be essential for late post-traumatic pathology. These mechanisms are applicable to remote hippocampal damage occurring after other types of focal brain damage (stroke, epilepsy) or central nervous system diseases without obvious focal injury. Thus, the liaisons of excessive glucocorticoids/dysfunctional hypothalamic–pituitary–adrenal axis with neuroinflammation, dangerous to the hippocampus, may be crucial to distant hippocampal damage in many brain diseases. Taking into account that the hippocampus controls both the cognitive functions and the emotional state, further research on potential links between glucocorticoid signaling and inflammatory processes in the brain and respective mechanisms is vital.
Collapse
Affiliation(s)
- Ilia G. Komoltsev
- Department of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117465 Moscow, Russia;
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia
| | - Natalia V. Gulyaeva
- Department of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117465 Moscow, Russia;
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia
- Correspondence: ; Tel.: +7-495-9524007 or +7-495-3347020
| |
Collapse
|
39
|
Impact of stress on inhibitory neuronal circuits, our tribute to Bruce McEwen. Neurobiol Stress 2022; 19:100460. [PMID: 35734023 PMCID: PMC9207718 DOI: 10.1016/j.ynstr.2022.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 05/10/2022] [Indexed: 12/03/2022] Open
Abstract
This manuscript is dedicated to the memory of Bruce S. McEwen, to commemorate the impact he had on how we understand stress and neuronal plasticity, and the profound influence he exerted on our scientific careers. The focus of this review is the impact of stressors on inhibitory circuits, particularly those of the limbic system, but we also consider other regions affected by these adverse experiences. We revise the effects of acute and chronic stress during different stages of development and lifespan, taking into account the influence of the sex of the animals. We review first the influence of stress on the physiology of inhibitory neurons and on the expression of molecules related directly to GABAergic neurotransmission, and then focus on specific interneuron subpopulations, particularly on parvalbumin and somatostatin expressing cells. Then we analyze the effects of stress on molecules and structures related to the plasticity of inhibitory neurons: the polysialylated form of the neural cell adhesion molecule and perineuronal nets. Finally, we review the potential of antidepressants or environmental manipulations to revert the effects of stress on inhibitory circuits.
Collapse
|
40
|
Kraus KL, Chordia AP, Drake AW, Herman JP, Danzer SC. Hippocampal interneurons are direct targets for circulating glucocorticoids. J Comp Neurol 2022; 530:2100-2112. [PMID: 35397117 PMCID: PMC9232959 DOI: 10.1002/cne.25322] [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: 11/23/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/08/2022]
Abstract
The hippocampus has become a significant target of stress research in recent years because of its role in cognitive functioning, neuropathology, and regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Despite the pervasive impact of stress on psychiatric and neurological disease, many of the circuit- and cell-dependent mechanisms giving rise to the limbic regulation of the stress response remain unknown. Hippocampal excitatory neurons generally express high levels of glucocorticoid receptors (GRs) and are therefore positioned to respond directly to serum glucocorticoids. These neurons are, in turn, regulated by neighboring interneurons, subtypes of which have been shown to respond to stress exposure. However, GR expression among hippocampal interneurons is not well characterized. To determine whether key interneuron populations are direct targets for glucocorticoid action, we used two transgenic mouse lines to label parvalbumin-positive (PV+) and somatostatin-positive (SST+) interneurons. GR immunostaining of labeled interneurons was characterized within the dorsal and ventral dentate hilus, dentate cell body layer, and CA1 and CA3 stratum oriens and stratum pyramidale. While nearly all hippocampal SST+ interneurons expressed GR across all regions, GR labeling of PV+ interneurons showed considerable subregion variability. The percentage of PV+, GR+ cells was highest in the CA3 stratum pyramidale and lowest in the CA1 stratum oriens, with other regions showing intermediate levels of expression. Together, these findings indicate that, under baseline conditions, hippocampal SST+ interneurons are a ubiquitous glucocorticoid target, while only distinct populations of PV+ interneurons are direct targets. This anatomical diversity suggests functional differences in the regulation of stress-dependent hippocampal responses.
Collapse
Affiliation(s)
- Kimberly L Kraus
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Arihant P Chordia
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Austin W Drake
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
41
|
Tanti A, Belliveau C, Nagy C, Maitra M, Denux F, Perlman K, Chen F, Mpai R, Canonne C, Théberge S, McFarquhar A, Davoli MA, Belzung C, Turecki G, Mechawar N. Child abuse associates with increased recruitment of perineuronal nets in the ventromedial prefrontal cortex: a possible implication of oligodendrocyte progenitor cells. Mol Psychiatry 2022; 27:1552-1561. [PMID: 34799691 PMCID: PMC9095471 DOI: 10.1038/s41380-021-01372-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022]
Abstract
Child abuse (CA) is a strong predictor of psychopathologies and suicide, altering normal trajectories of brain development in areas closely linked to emotional responses such as the prefrontal cortex (PFC). Yet, the cellular underpinnings of these enduring effects are unclear. Childhood and adolescence are marked by the protracted formation of perineuronal nets (PNNs), which orchestrate the closure of developmental windows of cortical plasticity by regulating the functional integration of parvalbumin interneurons into neuronal circuits. Using well-characterized post-mortem brain samples, we show that a history of CA is specifically associated with increased densities and morphological complexity of WFL-labeled PNNs in the ventromedial PFC (BA11/12), possibly suggesting increased recruitment and maturation of PNNs. Through single-nucleus sequencing and fluorescent in situ hybridization, we found that the expression of canonical components of PNNs is enriched in oligodendrocyte progenitor cells (OPCs), and that they are upregulated in CA victims. These correlational findings suggest that early-life adversity may lead to persistent patterns of maladaptive behaviors by reducing the neuroplasticity of cortical circuits through the enhancement of developmental OPC-mediated PNN formation.
Collapse
Affiliation(s)
- Arnaud Tanti
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France.
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Malosree Maitra
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Fanny Denux
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Kelly Perlman
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Frank Chen
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Refilwe Mpai
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Candice Canonne
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Stéphanie Théberge
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Ashley McFarquhar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | | | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, Canada.
| |
Collapse
|
42
|
Godoy LD, Prizon T, Rossignoli MT, Leite JP, Liberato JL. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Front Integr Neurosci 2022; 16:765324. [PMID: 35250498 PMCID: PMC8891758 DOI: 10.3389/fnint.2022.765324] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Parvalbumin is a calcium-binding protein present in inhibitory interneurons that play an essential role in regulating many physiological processes, such as intracellular signaling and synaptic transmission. Changes in parvalbumin expression are deeply related to epilepsy, which is considered one of the most disabling neuropathologies. Epilepsy is a complex multi-factor group of disorders characterized by periods of hypersynchronous activity and hyperexcitability within brain networks. In this scenario, inhibitory neurotransmission dysfunction in modulating excitatory transmission related to the loss of subsets of parvalbumin-expressing inhibitory interneuron may have a prominent role in disrupted excitability. Some studies also reported that parvalbumin-positive interneurons altered function might contribute to psychiatric comorbidities associated with epilepsy, such as depression, anxiety, and psychosis. Understanding the epileptogenic process and comorbidities associated with epilepsy have significantly advanced through preclinical and clinical investigation. In this review, evidence from parvalbumin altered function in epilepsy and associated psychiatric comorbidities were explored with a translational perspective. Some advances in potential therapeutic interventions are highlighted, from current antiepileptic and neuroprotective drugs to cutting edge modulation of parvalbumin subpopulations using optogenetics, designer receptors exclusively activated by designer drugs (DREADD) techniques, transcranial magnetic stimulation, genome engineering, and cell grafting. Creating new perspectives on mechanisms and therapeutic strategies is valuable for understanding the pathophysiology of epilepsy and its psychiatric comorbidities and improving efficiency in clinical intervention.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- João Pereira Leite,
| | - José Luiz Liberato
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: José Luiz Liberato,
| |
Collapse
|
43
|
Zhang X, Yang X, Chen B, Shen K, Liu G, Wang Z, Huang K, Zhu G, Wang T, Lv S, Zhang C, Yang H, Hou Z, Liu S. Glucocorticoid receptors participate in epilepsy in FCDII patients and MP model rats: A potential therapeutic target for epilepsy in patients with focal cortical dysplasia II (FCDII). Expert Opin Ther Targets 2022; 26:171-186. [PMID: 35132930 DOI: 10.1080/14728222.2022.2032650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs) are involved in neuronal excitability, neurogenesis, and neuroinflammation. However, the roles of GRs and MRs in epilepsy in focal cortical dysplasia II (FCDII) have not been reported. RESEARCH DESIGN AND METHODS We evaluated GRs and MRs expression and distribution in FCDII patients and methylazoxymethanol-pilocarpine-induced epilepsy model rats (MP rats), and the effects of a GR agonist on neurons in human FCDII and investigated the electrophysiological properties of cultured neurons and neurons of MP rats after lentivirus-mediated GR knockdown or overexpression and GR agonist or antagonist administration. RESULTS GR expression (not MR) was decreased in specimens from FCDII patients and model rats. GR agonist dexamethasone reduced neuronal excitatory transmission and increased neuronal inhibitory transmission in FCDII. GR knockdown increased the excitability of cultured neurons, and GR overexpression rescued the hyperexcitability of MP-treated neurons. Moreover, dexamethasone decreased neuronal excitability and excitatory transmission in MP rats, while GR antagonist exerted the opposite effects. Dexamethasone reduced the seizure number and duration by approximately 85% and 60% in MP rats within one to two hours. CONCLUSIONS These results suggested that GRs play an important role in epilepsy in FCDII and GR activation may have protective and antiepileptic effects in FCDII.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiaolin Yang
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Bing Chen
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Kaifeng Shen
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guolong Liu
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zhongke Wang
- Department of Neurosurgery, Armed police Hospital, Chongqing, China
| | - Kaixuan Huang
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Gang Zhu
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Tingting Wang
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Shengqing Lv
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chunqing Zhang
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Hui Yang
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zhi Hou
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Shiyong Liu
- National Comprehensive Epilepsy Center, Department of Neurosurgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
44
|
Chen S, Chen F, Amin N, Ren Q, Ye S, Hu Z, Tan X, Jiang M, Fang M. Defects of parvalbumin-positive interneurons in the ventral dentate gyrus region are implicated depression-like behavior in mice. Brain Behav Immun 2022; 99:27-42. [PMID: 34562597 DOI: 10.1016/j.bbi.2021.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/01/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022] Open
Abstract
Depression is an increasingly common but extremely serve mood disorder that remains poorly understood and inadequately treated. Fast-spiking parvalbumin-positive interneurons (PVIs), a subpopulation of GABAergic interneurons (GABA, g-aminobutyric acid), exhibit a widespread distribution throughout the hippocampus, and has been reported to play an important role in a variety of mental disorders. However, the relationship between depression and hippocampal PVIs remains unclear. Here in this present study, a series of experiments were conducted to clarify the potential relationship. Here, chronic unpredicted mild stress (CUMS) and Lipopolysaccharide (LPS) injection were introduced to induce depression-like behavior in mice, and led to a clear decline in PVIs numbers in the ventral hippocampal (vHPC), particularly in the ventral dentate gyrus (vDG) subfield. After a selectively removal of the PVIs in PV-ires-Cre::Ai14 mice, we confirmed that ablation of PVIs from the vDG induced depression-like behavior. Furthermore, we found that the removal of vDG-PVIs induced depression likely to be accounted for upregulation of neuroinflammation. These findings facilitate us better understand the role of hippocampal PVIs in depression.
Collapse
Affiliation(s)
- Shijia Chen
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; Institute of Neuroscience, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fengpei Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Nashwa Amin
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; Institute of Neuroscience, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Department of Zoology, Faculty of Science, Aswan University, Aswan 81521, Egypt
| | - Qiannan Ren
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; Institute of Neuroscience, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shan Ye
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhiying Hu
- Obstetrics & Gynecology Department, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou 310003, China
| | - Xiaoning Tan
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; Institute of Neuroscience, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Mizu Jiang
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Marong Fang
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China.
| |
Collapse
|
45
|
Sex differences in the rodent hippocampal opioid system following stress and oxycodone associated learning processes. Pharmacol Biochem Behav 2022; 212:173294. [PMID: 34752798 PMCID: PMC8748406 DOI: 10.1016/j.pbb.2021.173294] [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: 08/14/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 01/03/2023]
Abstract
Over the past two decades, opioid abuse has risen especially among women. In both sexes hippocampal neural circuits involved in associative memory formation and encoding of motivational incentives are critically important in the transition from initial drug use to drug abuse/dependence. Opioid circuits, particularly the mossy fiber pathway, are crucial for associative memory processes important for addiction. Our anatomical studies, especially those utilizing electron microscopic immunocytochemistry, have provided unique insight into sex differences in the distribution of opioid peptides and receptors in specific hippocampal circuits and how these distributions are altered following stress and oxycodone-associative learning processes. Here we review the hippocampal opioid system in rodents with respect to ovarian hormones effects and baseline sex differences then sex differences following acute and chronic stress. Next, we review sex differences in the hippocampal opioid system in unstressed and chronically stressed rats following oxycodone conditioned place preference. We show that opioid peptides and receptors are distributed within hippocampal circuits in females with elevated estrogen states in a manner that would enhance sensitivity to endogenous and exogenous opioids. Moreover, chronic stress primes the opioid system in females in a manner that would promote opioid-associative learning processes. In contrast, chronic stress has limited effects on the opioid system in males and reduces its capacity to support opioid-mediated learning processes. Interestingly, acute stress appears to prime males for opioid associative learning. On a broader scale the findings highlighted in this review have important implications in understanding sex differences in opioid drug use and abuse.
Collapse
|
46
|
Juarez P, Martínez Cerdeño V. Parvalbumin and parvalbumin chandelier interneurons in autism and other psychiatric disorders. Front Psychiatry 2022; 13:913550. [PMID: 36311505 PMCID: PMC9597886 DOI: 10.3389/fpsyt.2022.913550] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Parvalbumin (PV) is a calcium binding protein expressed by inhibitory fast-spiking interneurons in the cerebral cortex. By generating a fast stream of action potentials, PV+ interneurons provide a quick and stable inhibitory input to pyramidal neurons and contribute to the generation of gamma oscillations in the cortex. Their fast-firing rates, while advantageous for regulating cortical signaling, also leave them vulnerable to metabolic stress. Chandelier (Ch) cells are a type of PV+ interneuron that modulate the output of pyramidal neurons and synchronize spikes within neuron populations by directly innervating the pyramidal axon initial segment. Changes in the morphology and/or function of PV+ interneurons, mostly of Ch cells, are linked to neurological disorders. In ASD, the number of PV+ Ch cells is decreased across several cortical areas. Changes in the morphology and/or function of PV+ interneurons have also been linked to schizophrenia, epilepsy, and bipolar disorder. Herein, we review the role of PV and PV+ Ch cell alterations in ASD and other psychiatric disorders.
Collapse
Affiliation(s)
- Pablo Juarez
- Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospital for Children and UC Davis School of Medicine, Sacramento, CA, United States.,Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, United States
| | - Verónica Martínez Cerdeño
- Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospital for Children and UC Davis School of Medicine, Sacramento, CA, United States.,Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, United States.,MIND Institute, UC Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
47
|
Ho TC, Gifuni AJ, Gotlib IH. Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of the role of puberty. Mol Psychiatry 2022; 27:606-623. [PMID: 34117365 PMCID: PMC8960417 DOI: 10.1038/s41380-021-01171-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023]
Abstract
Suicide is the second leading cause of death among adolescents. While clinicians and researchers have begun to recognize the importance of considering multidimensional factors in understanding risk for suicidal thoughts and behaviors (STBs) during this developmental period, the role of puberty has been largely ignored. In this review, we contend that the hormonal events that occur during puberty have significant effects on the organization and development of brain systems implicated in the regulation of social stressors, including amygdala, hippocampus, striatum, medial prefrontal cortex, orbitofrontal cortex, and anterior cingulate cortex. Guided by previous experimental work in adults, we also propose that the influence of pubertal hormones and social stressors on neural systems related to risk for STBs is especially critical to consider in adolescents with a neurobiological sensitivity to hormonal changes. Furthermore, facets of the pubertal transition, such as pubertal timing, warrant deeper investigation and may help us gain a more comprehensive understanding of sex differences in the neurobiological and psychosocial mechanisms underlying adolescent STBs. Ultimately, advancing our understanding of the pubertal processes that contribute to suicide risk will improve early detection and facilitate the development of more effective, sex-specific, psychiatric interventions for adolescents.
Collapse
Affiliation(s)
- Tiffany C. Ho
- grid.168010.e0000000419368956Department of Psychology, Stanford University, Stanford, CA USA ,grid.266102.10000 0001 2297 6811Department of Psychiatry and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA USA
| | - Anthony J. Gifuni
- grid.168010.e0000000419368956Department of Psychology, Stanford University, Stanford, CA USA ,grid.14709.3b0000 0004 1936 8649Psychiatry Department and Douglas Mental Health University Institute, McGill University, Montréal, QC Canada
| | - Ian H. Gotlib
- grid.168010.e0000000419368956Department of Psychology, Stanford University, Stanford, CA USA
| |
Collapse
|
48
|
Abebe EC, Mengstie MA, Seid MA, Dejenie TA. Regulatory effects and potential therapeutic implications of alarin in depression, and arguments on its receptor. Front Psychiatry 2022; 13:1051235. [PMID: 36506414 PMCID: PMC9732279 DOI: 10.3389/fpsyt.2022.1051235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Alarin is a pleiotropic peptide involved in a multitude of putative biological activities, notably, it has a regulatory effect on depression-like behaviors. Although further elucidating research is needed, animal-based cumulative evidence has shown the antidepressant-like effects of alarin. In light of its regulatory role in depression, alarin could be used as a promising antidepressant in future treatment for depression. Nevertheless, the available information is still insufficient and the therapeutic relevance of alarin in depression is still of concern. Moreover, a plethora of studies have reported that the actions of alarin, including antidepressant activities, are mediated by a separate yet unidentified receptor, highlighting the need for more extensive research. This review focuses on the current understanding of the regulatory effects and future therapeutic relevance of alarin on depression, and the arguments on its receptors.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Misganaw Asmamaw Mengstie
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Mohammed Abdu Seid
- Department of Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Medical Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
49
|
Edem EE, Ihaza BE, Fafure AA, Ishola AO, Nebo KE, Enye LA, Akinluyi ET. Virgin coconut oil abrogates depression-associated cognitive deficits by modulating hippocampal antioxidant balance, GABAergic and glutamatergic receptors in mice. Drug Metab Pers Ther 2021; 37:177-190. [PMID: 34881837 DOI: 10.1515/dmpt-2021-0126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES GABA and glutamate neurotransmission play critical roles in both the neurobiology of depression and cognition; and Virgin coconut oil (VCO) is reported to support brain health. The present study investigated the effect of VCO on depression-associated cognitive deficits in mice. METHODS Thirty male mice divided into five groups were either exposed to chronic unpredicted mild stress (CUMS) protocol for 28 days or pre-treated with 3 mL/kg b. wt. of VCO for 21 days or post-treated with 3 mL/kg b. wt. of VCO for 21 days following 28 days of CUMS exposure. Mice were subjected to behavioural assessments for depressive-like behaviours and short-term memory, and thereafter euthanised. Hippocampal tissue was dissected from the harvested whole brain for biochemical and immunohistochemical evaluations. RESULTS Our results showed that CUMS exposure produced depressive-like behaviours, cognitive deficits and altered hippocampal redox balance. However, treatment with VCO abrogated depression-associated cognitive impairment, and enhanced hippocampal antioxidant concentration. Furthermore, immunohistochemical evaluation revealed significant improvement in GABAA and mGluR1a immunoreactivity following treatment with VCO in the depressed mice. CONCLUSIONS Therefore, findings from this study support the dietary application of VCO to enhance neural resilience in patients with depression and related disorders.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Blessing Eghosa Ihaza
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Adedamola Adediran Fafure
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Azeez Olakunle Ishola
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Kate Eberechukwu Nebo
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Linus Anderson Enye
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Elizabeth Toyin Akinluyi
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
50
|
Woodward EM, Coutellier L. Age- and sex-specific effects of stress on parvalbumin interneurons in preclinical models: Relevance to sex differences in clinical neuropsychiatric and neurodevelopmental disorders. Neurosci Biobehav Rev 2021; 131:1228-1242. [PMID: 34718048 PMCID: PMC8642301 DOI: 10.1016/j.neubiorev.2021.10.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/23/2021] [Indexed: 01/06/2023]
Abstract
Stress is a major risk factor for neurodevelopmental and neuropsychiatric disorders, with the capacity to impact susceptibility to disease as well as long-term neurobiological and behavioral outcomes. Parvalbumin (PV) interneurons, the most prominent subtype of GABAergic interneurons in the cortex, are uniquely responsive to stress due to their protracted development throughout the highly plastic neonatal period and into puberty and adolescence. Additionally, PV + interneurons appear to respond to stress in a sex-specific manner. This review aims to discuss existing preclinical studies that support our overall hypothesis that the sex-and age-specific impacts of stress on PV + interneurons contribute to differences in individual vulnerability to stress across the lifespan, particularly in regard to sex differences in the diagnostic rate of neurodevelopmental and neuropsychiatric diseases in clinical populations. We also emphasize the importance of studying sex as a biological variable to fully understand the mechanistic and behavioral differences between males and females in models of neuropsychiatric disease.
Collapse
Affiliation(s)
- Emma M Woodward
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States; Department of Psychology, Ohio State University, 53 Psychology Building, 1835 Neil Avenue, Columbus, OH, 43210, United States.
| |
Collapse
|