1
|
Moradi Khankani A, Hossein Meftahi G. Pretreatment with 4-methylumbilliferon improves anxiety-like behaviors and memory impairment in stressed rats via modulation of neuronal cell death and oxidative stress. Brain Res 2024; 1844:149196. [PMID: 39181223 DOI: 10.1016/j.brainres.2024.149196] [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: 05/18/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.
Collapse
Affiliation(s)
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Grizzell JA, Clarity TT, Rodriguez RM, Marshall ZQ, Cooper MA. Effects of social dominance and acute social stress on morphology of microglia and structural integrity of the medial prefrontal cortex. Brain Behav Immun 2024; 122:353-367. [PMID: 39187049 PMCID: PMC11402560 DOI: 10.1016/j.bbi.2024.08.043] [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: 11/28/2023] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic stress increases activity of the brain's innate immune system and impairs function of the medial prefrontal cortex (mPFC). However, whether acute stress triggers similar neuroimmune mechanisms is poorly understood. Across four studies, we used a Syrian hamster model to investigate whether acute stress drives changes in mPFC microglia in a time-, subregion-, and social status-dependent manner. We found that acute social defeat increased expression of ionized calcium binding adapter molecule 1 (Iba1) in the infralimbic (IL) and prelimbic (PL) and altered the morphology Iba1+ cells 1, 2, and 7 days after social defeat. We also investigated whether acute defeat induced tissue degeneration and reductions of synaptic plasticity 2 days post-defeat. We found that while social defeat increased deposition of cellular debris and reduced synaptophysin immunoreactivity in the PL and IL, treatment with minocycline protected against these cellular changes. Finally, we tested whether a reduced conditioned defeat response in dominant compared to subordinate hamsters was associated with changes in microglia reactivity in the IL and PL. We found that while subordinate hamsters and those without an established dominance relationships showed defeat-induced changes in morphology of Iba1+ cells and cellular degeneration, dominant hamsters showed resistance to these effects of social defeat. Taken together, these findings indicate that acute social defeat alters microglial morphology, increases markers of tissue degradation, and impairs structural integrity in the IL and PL, and that experience winning competitive interactions can specifically protect the IL and reduce stress vulnerability.
Collapse
Affiliation(s)
- J Alex Grizzell
- Neuroscience and Behavioral Biology Program, Emory University, United States; Department of Psychology, University of Tennessee Knoxville, United States; Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Thomas T Clarity
- Department of Psychology, University of Tennessee Knoxville, United States
| | - R Mason Rodriguez
- Department of Psychology, University of Tennessee Knoxville, United States
| | - Zachary Q Marshall
- Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Matthew A Cooper
- Department of Psychology, University of Tennessee Knoxville, United States.
| |
Collapse
|
3
|
Kovačević S, Pavković Ž, Brkljačić J, Elaković I, Vojnović Milutinović D, Djordjevic A, Pešić V. High-Fructose Diet and Chronic Unpredictable Stress Modify Each Other's Neurobehavioral Effects in Female Rats. Int J Mol Sci 2024; 25:11721. [PMID: 39519293 PMCID: PMC11546065 DOI: 10.3390/ijms252111721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
A pervasive exposure to stressors and the consumption of fructose-containing beverages usually go hand-in-hand in everyday life. In contrast to their metabolic outcomes, their impact on the brain and behavior is still understudied. We examined the behavioral response to a novelty (open field test), the expression of biochemical indicators of neuronal activity (Egr1 and FosB/ΔFosB), the synaptic potentiation (CaMKIIα and pCaMKIIThr286), the synaptic plasticity (synaptophysin, PSD95, gephyrin, and drebrin), and the GABAergic system (parvalbumin and GAD67), along with the glucocorticoid receptor (GR) and AMPK, in the medial prefrontal cortex of female Wistar rats subjected to liquid fructose supplementation (F), chronic unpredictable stress (S), or both (SF) over 9 weeks. The only hallmark of the F group was an increased expression of pCaMKIIThr286, which was also observed in the S group, but not in the SF group. The SF group did not show hyperactivity, a decreased expression of FosB, or an increased expression of parvalbumin, as the S group did. The SF group, as with the S group, showed a decreased expression of the GR, although the basal level of corticosterone was unchanged. The SF group showed, as de novo marks, thigmotactic behavior, increased drebrin, and decreased gephyrin expression. These findings suggest that the long-term consumption of fructose, which itself has subtle neurobehavioral consequences, in combination with stress prevents some of its effects, but also contributes to novel outcomes not seen in single treatments.
Collapse
Affiliation(s)
- Sanja Kovačević
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11060 Belgrade, Serbia
| | - Željko Pavković
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Jelena Brkljačić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11060 Belgrade, Serbia
| | - Ivana Elaković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11060 Belgrade, Serbia
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11060 Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd., 11060 Belgrade, Serbia
| | - Vesna Pešić
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| |
Collapse
|
4
|
Al-Shargie F, Tariq U, Al-Ameri S, Al-Hammadi A, Vladimirovna SD, Al-Nashash H. Assessment of Brain Function After 240 Days Confinement Using Functional Near Infrared Spectroscopy. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2024; 6:54-60. [PMID: 39564559 PMCID: PMC11573361 DOI: 10.1109/ojemb.2024.3457240] [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: 01/03/2024] [Revised: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 09/23/2024] Open
Abstract
Future space exploration missions will expose astronauts to various stressors, making the early detection of mental stress crucial for prolonged missions. Our study proposes using functional near infrared spectroscopy (fNIRS) combined with multiple machine learning models to assess the level of mental stress. Objective: The objective is to identify and quantify stress levels during 240 days confinement scenario. In this study, we utilize a diverse set of stress indicators including salivary alpha amylase (sAA) levels, reaction time (RT) to stimuli, accuracy of target detection, and power spectral density (PSD), in conjunction with functional connectivity networks (FCN). We estimate the PSD using Fast Fourier Transform (FFT) and the FCN using partial directed coherence. Results: Our findings reveal several intriguing insights. The sAA levels increased from the first 30 days in confinement to the culmination of the lengthy 240-day mission, suggesting a cumulative impact of stress. Conversely, RT and the accuracy of target detection exhibit significant fluctuations over the course of the mission. The power spectral density shows a significant increase with time-in-mission across all participants in most of the frontal area. The FCN shows a significant decrease in most of the right frontal areas. Five different machine learning classifiers are employed to differentiate between two levels of stress resulting in impressive classification accuracy rates: 96.44% with-nearest neighbor (KNN), 95.52% with linear discriminant analysis (LDA), 88.71% with Naïve Bayes (NB), 87.41 with decision trees (DT) and 96.48% with Support Vector Machine (SVM). In conclusion, this study demonstrates the effectiveness of combining functional near infrared spectroscopy (fNIRS) with multiple machine learning models to accurately assess and quantify mental stress levels during prolonged space missions, providing a promising approach for early stress detection in astronauts.
Collapse
|
5
|
Rodrigues B, Leitão RA, Santos M, Trofimov A, Silva M, Inácio ÂS, Abreu M, Nobre RJ, Costa J, Cardoso AL, Milosevic I, Peça J, Oliveiros B, Pereira de Almeida L, Pinheiro PS, Carvalho AL. MiR-186-5p inhibition restores synaptic transmission and neuronal network activity in a model of chronic stress. Mol Psychiatry 2024:10.1038/s41380-024-02715-1. [PMID: 39237722 DOI: 10.1038/s41380-024-02715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Chronic stress exerts profound negative effects on cognitive and emotional behaviours and is a major risk factor for the development of neuropsychiatric disorders. However, the molecular links between chronic stress and its deleterious effects on neuronal and synaptic function remain elusive. Here, using a combination of in vitro and in vivo approaches, we demonstrate that the upregulation of miR-186-5p triggered by chronic stress may be a key mediator of such changes, leading to synaptic dysfunction. Our results show that the expression levels of miR-186-5p are increased both in the prefrontal cortex (PFC) of mice exposed to chronic stress and in cortical neurons chronically exposed to dexamethasone. Additionally, viral overexpression of miR-186-5p in the PFC of naïve mice induces anxiety- and depressive-like behaviours. The upregulation of miR-186-5p through prolonged glucocorticoid receptor activation in vitro, or in a mouse model of chronic stress, differentially affects glutamatergic and GABAergic synaptic transmission, causing an imbalance in excitation/inhibition that leads to altered neuronal network activity. At glutamatergic synapses, we observed both a reduction in synaptic AMPARs and synaptic transmission, whereas GABAergic synaptic transmission was strengthened. These changes could be rescued in vitro by a miR-186-5p inhibitor. Overall, our results establish a novel molecular link between chronic glucocorticoid receptor activation, the upregulation of miR-186-5p and the synaptic changes induced by chronic stress, that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Beatriz Rodrigues
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ricardo A Leitão
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Santos
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Alexander Trofimov
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Integrative Brain Function Neurobiology Lab, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, 197022, St. Petersburg, Russia
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Mariline Silva
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Department of Applied Physics and Science for Life Laboratory (SciLifeLab), KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Ângela S Inácio
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Abreu
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rui J Nobre
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Jéssica Costa
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ana Luísa Cardoso
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ira Milosevic
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - João Peça
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Bárbara Oliveiros
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- iCRB-Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, 3000-548, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Paulo S Pinheiro
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| |
Collapse
|
6
|
Datta D, Yang S, Joyce MKP, Woo E, McCarroll SA, Gonzalez-Burgos G, Perone I, Uchendu S, Ling E, Goldman M, Berretta S, Murray J, Morozov Y, Arellano J, Duque A, Rakic P, O'Dell R, van Dyck CH, Lewis DA, Wang M, Krienen FM, Arnsten AFT. Key Roles of CACNA1C/Cav1.2 and CALB1/Calbindin in Prefrontal Neurons Altered in Cognitive Disorders. JAMA Psychiatry 2024; 81:870-881. [PMID: 38776078 PMCID: PMC11112502 DOI: 10.1001/jamapsychiatry.2024.1112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/14/2024] [Indexed: 05/25/2024]
Abstract
Importance The risk of mental disorders is consistently associated with variants in CACNA1C (L-type calcium channel Cav1.2) but it is not known why these channels are critical to cognition, and whether they affect the layer III pyramidal cells in the dorsolateral prefrontal cortex that are especially vulnerable in cognitive disorders. Objective To examine the molecular mechanisms expressed in layer III pyramidal cells in primate dorsolateral prefrontal cortices. Design, Setting, and Participants The design included transcriptomic analyses from human and macaque dorsolateral prefrontal cortex, and connectivity, protein expression, physiology, and cognitive behavior in macaques. The research was performed in academic laboratories at Yale, Harvard, Princeton, and the University of Pittsburgh. As dorsolateral prefrontal cortex only exists in primates, the work evaluated humans and macaques. Main Outcomes and Measures Outcome measures included transcriptomic signatures of human and macaque pyramidal cells, protein expression and interactions in layer III macaque pyramidal cells using light and electron microscopy, changes in neuronal firing during spatial working memory, and working memory performance following pharmacological treatments. Results Layer III pyramidal cells in dorsolateral prefrontal cortex coexpress a constellation of calcium-related proteins, delineated by CALB1 (calbindin), and high levels of CACNA1C (Cav1.2), GRIN2B (NMDA receptor GluN2B), and KCNN3 (SK3 potassium channel), concentrated in dendritic spines near the calcium-storing smooth endoplasmic reticulum. L-type calcium channels influenced neuronal firing needed for working memory, where either blockade or increased drive by β1-adrenoceptors, reduced neuronal firing by a mean (SD) 37.3% (5.5%) or 40% (6.3%), respectively, the latter via SK potassium channel opening. An L-type calcium channel blocker or β1-adrenoceptor antagonist protected working memory from stress. Conclusions and Relevance The layer III pyramidal cells in the dorsolateral prefrontal cortex especially vulnerable in cognitive disorders differentially express calbindin and a constellation of calcium-related proteins including L-type calcium channels Cav1.2 (CACNA1C), GluN2B-NMDA receptors (GRIN2B), and SK3 potassium channels (KCNN3), which influence memory-related neuronal firing. The finding that either inadequate or excessive L-type calcium channel activation reduced neuronal firing explains why either loss- or gain-of-function variants in CACNA1C were associated with increased risk of cognitive disorders. The selective expression of calbindin in these pyramidal cells highlights the importance of regulatory mechanisms in neurons with high calcium signaling, consistent with Alzheimer tau pathology emerging when calbindin is lost with age and/or inflammation.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Shengtao Yang
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Mary Kate P Joyce
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Elizabeth Woo
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | | | - Isabella Perone
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Stacy Uchendu
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Emi Ling
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Melissa Goldman
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Sabina Berretta
- Basic Neuroscience Division, McLean Hospital, Belmont, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - John Murray
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Yury Morozov
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Jon Arellano
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Pasko Rakic
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Ryan O'Dell
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher H van Dyck
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - David A Lewis
- Departments of Psychiatry and Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Fenna M Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
7
|
Tripathi A, Bartosh A, Mata J, Jacks C, Madeshiya AK, Hussein U, Hong LE, Zhao Z, Pillai A. Microglial type I interferon signaling mediates chronic stress-induced synapse loss and social behavior deficits. Mol Psychiatry 2024:10.1038/s41380-024-02675-6. [PMID: 39095477 DOI: 10.1038/s41380-024-02675-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Inflammation and synapse loss have been associated with deficits in social behavior and are involved in pathophysiology of many neuropsychiatric disorders. Synapse loss, characterized by reduction in dendritic spines can significantly disrupt synaptic connectivity and neural circuitry underlying social behavior. Chronic stress is known to induce loss of spines and dendrites in the prefrontal cortex (PFC), a brain region implicated in social behavior. However, the underlying mechanisms are not well understood. In the present study, we investigated the role of type I Interferon (IFN-I) signaling in chronic unpredictable stress (CUS)-induced synapse loss and behavior deficits in mice. We found increased expression of type I IFN receptor (IFNAR) in microglia following CUS. Conditional knockout of microglial IFNAR in adult mice rescued CUS-induced social behavior deficits and synapse loss. Bulk RNA sequencing data show that microglial IFNAR deletion attenuated CUS-mediated changes in the expression of genes such as Keratin 20 (Krt20), Claudin-5 (Cldn5) and Nuclear Receptor Subfamily 4 Group A Member 1 (Nr4a1) in the PFC. Cldn5 and Nr4a1 are known for their roles in synaptic plasticity. Krt20 is an intermediate filament protein responsible for the structural integrity of epithelial cells. The reduction in Krt20 following CUS presents a novel insight into the potential contribution of cytokeratin in stress-induced alterations in neuroplasticity. Overall, these results suggest that microglial IFNAR plays a critical role in regulating synaptic plasticity and social behavior deficits associated with chronic stress conditions.
Collapse
Affiliation(s)
- Ashutosh Tripathi
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alona Bartosh
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jocelyn Mata
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chale Jacks
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Amit Kumar Madeshiya
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Usama Hussein
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Lymphoma & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Elliot Hong
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anilkumar Pillai
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
8
|
Mancone S, Corrado S, Tosti B, Spica G, Di Siena F, Diotaiuti P. Exploring the Interplay between Sleep Quality, Stress, and Somatization among Teachers in the Post-COVID-19 Era. Healthcare (Basel) 2024; 12:1472. [PMID: 39120175 PMCID: PMC11311578 DOI: 10.3390/healthcare12151472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
(1) Background. The post-COVID-19 era has imposed unique challenges on educators, significantly impacting their psychological and physical well-being. This study examines the interrelationships among psychological stress, sleep quality, and somatization in a sample of teachers, elucidating the impact of these factors during the ongoing recovery from the pandemic. (2) Methods. Using validated instruments such as the Pittsburgh Sleep Quality Index (PSQI) and the Mesure du Stress Psychologique (MSP), this research investigates how stress and sleep disturbances correlate with somatization among teachers. The study also considers the influence of demographic factors such as age, gender, and years of experience. (3) Results. The results indicated that sleep quality significantly correlates with both psychological stress and somatic pain, emphasizing the crucial role of sleep in managing stress-induced physical symptoms. Additionally, the fear of COVID-19 significantly exacerbates these effects, illustrating the complex interplay of psychological and physical health factors during the pandemic. Contrary to initial hypotheses, demographic factors such as gender, age, and years of experience did not significantly influence these primary relationships. (4) Conclusions. The findings emphasize the necessity of addressing both psychological stress and sleep quality to mitigate their combined effects on somatization. Educational institutions and policymakers are urged to develop targeted interventions that address these issues to support teachers' health and well-being in a post-pandemic landscape.
Collapse
Affiliation(s)
| | | | | | | | | | - Pierluigi Diotaiuti
- Department of Human Sciences, Society and Health, University of Cassino and Southern Lazio, 03043 Cassino, Italy; (S.M.); (S.C.); (B.T.); (G.S.); (F.D.S.)
| |
Collapse
|
9
|
Joyce MKP, Uchendu S, Arnsten AFT. Stress and Inflammation Target Dorsolateral Prefrontal Cortex Function: Neural Mechanisms Underlying Weakened Cognitive Control. Biol Psychiatry 2024:S0006-3223(24)01420-3. [PMID: 38944141 DOI: 10.1016/j.biopsych.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Most mental disorders involve dysfunction of the dorsolateral prefrontal cortex (dlPFC), a recently evolved brain region that subserves working memory, abstraction, and the thoughtful regulation of attention, action, and emotion. For example, schizophrenia, depression, long COVID, and Alzheimer's disease are all associated with dlPFC dysfunction, with neuropathology often being focused in layer III. The dlPFC has extensive top-down projections, e.g., to the posterior association cortices to regulate attention and to the subgenual cingulate cortex via the rostral and medial PFC to regulate emotional responses. However, the dlPFC is particularly dependent on arousal state and is very vulnerable to stress and inflammation, which are etiological and/or exacerbating factors for most mental disorders. The cellular mechanisms by which stress and inflammation impact the dlPFC are a topic of current research and are summarized in this review. For example, the layer III dlPFC circuits that generate working memory-related neuronal firing have unusual neurotransmission, depending on NMDA receptor and nicotinic α7 receptor actions that are blocked under inflammatory conditions by kynurenic acid. These circuits also have unusual neuromodulation, with the molecular machinery to magnify calcium signaling in spines needed to support persistent firing, which must be tightly regulated to prevent toxic calcium actions. Stress rapidly weakens layer III connectivity by driving feedforward calcium-cAMP (cyclic adenosine monophosphate) opening of potassium channels on spines. This is regulated by postsynaptic noradrenergic α2A adrenergic receptor and mGluR3 (metabotropic glutamate receptor 3) signaling but dysregulated by inflammation and/or chronic stress exposure, which contribute to spine loss. Treatments that strengthen the dlPFC via pharmacological (the α2A adrenergic receptor agonist, guanfacine) or repetitive transcranial magnetic stimulation manipulation provide a rational basis for therapy.
Collapse
Affiliation(s)
- Mary Kate P Joyce
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Stacy Uchendu
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut.
| |
Collapse
|
10
|
Liu H, Guo D, Wang J, Zhang W, Zhu Z, Zhu K, Bi S, Pan P, Liang G. Aloe-emodin from Sanhua Decoction inhibits neuroinflammation by regulating microglia polarization after subarachnoid hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117583. [PMID: 38122912 DOI: 10.1016/j.jep.2023.117583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 11/20/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Subarachnoid hemorrhage (SAH) triggers a cascade of events that lead to early brain injury (EBI), which contributes to poor outcomes and appears within 3 days after SAH initiation. EBI involves multiple process including neuronal death, blood-brain barrier (BBB) injury and inflammation response. Microglia are cluster of immune cells originating in the brain which respond to SAH by changing their states and releasing inflammatory molecules through various signaling pathways. M0, M1, M2 are three states of microglia represent resting state, promoting inflammation state, and anti-inflammation state respectively, which can be modulated by pharmacological strategies. AIM OF THE STUDY After identified potential active ingredients and targets of Sanhua Decoction (SHD) for SAH, we selected aloe-emodin (AE) as a potential ingredient modulating microglia activation states. MATERIALS AND METHODS Molecular mechanisms, targets and pathways of SHD were reveal by network pharmacology technique. The effects of AE on SAH were evaluated in vivo by assessing neurological deficits, neuronal apoptosis and BBB integrity in a mouse SAH model. Furthermore, BV-2 cells were used to examine the effects of AE on microglial polarization. The influence of AE on microglia transformation was measured by Iba-1, TNF-α, CD68, Arg-1 and CD206 staining. The signal pathways of neuronal apoptosis and microglia polarization was measured by Western blot. RESULTS Network pharmacology identified potential active ingredients and targets of SHD for SAH. And AE is one of the active ingredients. We also confirmed that AE via NF-κB and PKA/CREB pathway inhibited the microglia activation and promoted transformation from M1 phenotype to M2 at EBI stage after SAH. CONCLUSIONS AE, as one ingredient of SHD, can alleviate the inflammatory response and protecting neurons from SAH-induced injury. AE has potential value for treating SAH-induced nerve injury and is expected to be applied in clinical practice.
Collapse
Affiliation(s)
- Hui Liu
- Department of Clinical Medicine, College of Medicine, Lishui University, Lishui, China
| | - Dan Guo
- Department of First Outpatients, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiao Wang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Lishui University, Lishui, China
| | - Wenxu Zhang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zechao Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Kunyuan Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Shijun Bi
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Pengyu Pan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
11
|
McElwee C, Lopez Hernandez DW. The influence of early life socio-environmental factors on executive performance in a healthy adult sample. APPLIED NEUROPSYCHOLOGY. ADULT 2024:1-12. [PMID: 38447195 DOI: 10.1080/23279095.2024.2323630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
OBJECTIVE Attempts have been made (with research efforts encouraged) to deconstruct the "race" concept into language, cultural, and life experience variables that can help explain performance differences found between ethnic groups (Romero et al., 2009). The extant empirical literature reveals that early environmental factors and life experiences (e.g., socioeconomic status) are related to cognitive test performance in adulthood (Byrd et al., 2006). This study examined the explanatory value of early life childhood resources in the relationship between ethnicity and neuropsychological test performance in adulthood. PARTICIPANTS/ METHODS Neurologically and psychologically healthy African American (n = 40), Caucasian (n = 14), and Hispanic (n = 107) college students ranging from 19-38 years of age. On average, participants had completed around 13 years of education, indicating that the majority were in the early stages of their undergraduate studies and mostly consisted of females (72%). Each participant completed a comprehensive neuropsychological battery that included tests of executive function and an extensive background questionnaire. RESULTS A one-way analysis of variance (ANOVA) revealed that the CA group was significantly older (F (2, 160) = 18.38, p = .045) compared to the AA and H groups, but the groups did not differ in terms of number of years of educations or gender. Also, an ANOVA revealed significant group test performance differences on the Stroop-C [F (2, 160) = 1.53, p = .047], but not on the TMT-B and COWAT. Furthermore, a Tukey post hoc revealed that there were no significant differences in test performance on Stroop-C between the groups. Hierarchical multiple regression analyses revealed that group performance differences on executive function tests were medium or non-existent and only partially explained by years of education and early life financial resources. CONCLUSION The results are discussed in light of the existing literature, study strengths and limitations, as well as directions for future research. This research can aid in pinpointing variables crucial for interpreting differences in neuropsychological assessments among diverse populations, holding potential implications for intervention research and policy settings. It is particularly relevant in the context of the continuously evolving social, political, and economic landscapes of societies.
Collapse
Affiliation(s)
- C McElwee
- Department of Psychology, University of California, Riverside, CA, USA
| | - D W Lopez Hernandez
- Department of Psychology, California State University, Dominguez Hills, CA, USA
| |
Collapse
|
12
|
Yi L, Lin X, She X, Gao W, Wu M. Chronic stress as an emerging risk factor for the development and progression of glioma. Chin Med J (Engl) 2024; 137:394-407. [PMID: 38238191 PMCID: PMC10876262 DOI: 10.1097/cm9.0000000000002976] [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: 05/13/2023] [Indexed: 02/21/2024] Open
Abstract
ABSTRACT Gliomas tend to have a poor prognosis and are the most common primary malignant tumors of the central nervous system. Compared with patients with other cancers, glioma patients often suffer from increased levels of psychological stress, such as anxiety and fear. Chronic stress (CS) is thought to impact glioma profoundly. However, because of the complex mechanisms underlying CS and variability in individual tolerance, the role of CS in glioma remains unclear. This review suggests a new proposal to redivide the stress system into two parts. Neuronal activity is dominant upstream. Stress-signaling molecules produced by the neuroendocrine system are dominant downstream. We discuss the underlying molecular mechanisms by which CS impacts glioma. Potential pharmacological treatments are also summarized from the therapeutic perspective of CS.
Collapse
Affiliation(s)
- Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
| | - Xiaoling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wei Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Minghua Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
13
|
Abdelhamid M, Jung CG, Zhou C, Inoue R, Chen Y, Sento Y, Hida H, Michikawa M. Potential Therapeutic Effects of Bifidobacterium breve MCC1274 on Alzheimer's Disease Pathologies in AppNL-G-F Mice. Nutrients 2024; 16:538. [PMID: 38398861 PMCID: PMC10893354 DOI: 10.3390/nu16040538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
We previously demonstrated that orally supplemented Bifidobacterium breve MCC1274 (B. breve MCC1274) mitigated Alzheimer's disease (AD) pathologies in both 7-month-old AppNL-G-F mice and wild-type mice; thus, B. breve MCC1274 supplementation might potentially prevent the progression of AD. However, the possibility of using this probiotic as a treatment for AD remains unclear. Thus, we investigated the potential therapeutic effects of this probiotic on AD using 17-month-old AppNL-G-F mice with memory deficits and amyloid beta saturation in the brain. B. breve MCC1274 supplementation ameliorated memory impairment via an amyloid-cascade-independent pathway. It reduced hippocampal and cortical levels of phosphorylated extracellular signal-regulated kinase and c-Jun N-terminal kinase as well as heat shock protein 90, which might have suppressed tau hyperphosphorylation and chronic stress. Moreover, B. breve MCC1274 supplementation increased hippocampal synaptic protein levels and upregulated neuronal activity. Thus, B. breve MCC1274 supplementation may alleviate cognitive dysfunction by reducing chronic stress and tau hyperphosphorylation, thereby enhancing both synaptic density and neuronal activity in 17-month-old AppNL-G-F mice. Overall, this study suggests that B. breve MCC1274 has anti-AD effects and can be used as a potential treatment for AD.
Collapse
Affiliation(s)
- Mona Abdelhamid
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Cha-Gyun Jung
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
- Department of Neurophysiology and Brain Science, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Chunyu Zhou
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Rieko Inoue
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Yuxin Chen
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
| | - Yoshiki Sento
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan;
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (M.A.); (C.Z.); (R.I.); (Y.C.)
- Department of Geriatric Medicine School of Life, Dentistry at Niigata, Nippon Dental University, 1-8 Hamaura-cho, Chuo-ku, Niigata 951-8580, Japan
| |
Collapse
|
14
|
Reyes-Lizaola S, Luna-Zarate U, Tendilla-Beltrán H, Morales-Medina JC, Flores G. Structural and biochemical alterations in dendritic spines as key mechanisms for severe mental illnesses. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110876. [PMID: 37863171 DOI: 10.1016/j.pnpbp.2023.110876] [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: 08/01/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023]
Abstract
Severe mental illnesses (SMI) collectively affect approximately 20% of the global population, as estimated by the World Health Organization (WHO). Despite having diverse etiologies, clinical symptoms, and pharmacotherapies, these diseases share a common pathophysiological characteristic: the misconnection of brain areas involved in reality perception, executive control, and cognition, including the corticolimbic system. Dendritic spines play a crucial role in excitatory neurotransmission within the central nervous system. These small structures exhibit remarkable plasticity, regulated by factors such as neurotransmitter tone, neurotrophic factors, and innate immunity-related molecules, and other mechanisms - all of which are associated with the pathophysiology of SMI. However, studying dendritic spine mechanisms in both healthy and pathological conditions in patients is fraught with technical limitations. This is where animal models related to these diseases become indispensable. They have played a pivotal role in elucidating the significance of dendritic spines in SMI. In this review, the information regarding the potential role of dendritic spines in SMI was summarized, drawing from clinical and animal model reports. Also, the implications of targeting dendritic spine-related molecules for SMI treatment were explored. Specifically, our focus is on major depressive disorder and the neurodevelopmental disorders schizophrenia and autism spectrum disorder. Abundant clinical and basic research has studied the functional and structural plasticity of dendritic spines in these diseases, along with potential pharmacological targets that modulate the dynamics of these structures. These targets may be associated with the clinical efficacy of the pharmacotherapy.
Collapse
Affiliation(s)
- Sebastian Reyes-Lizaola
- Departamento de Ciencias de la Salud, Licenciatura en Medicina, Universidad Popular del Estado de Puebla (UPAEP), Puebla, Mexico
| | - Ulises Luna-Zarate
- Departamento de Ciencias de la Salud, Licenciatura en Medicina, Universidad de las Américas Puebla (UDLAP), Puebla, Mexico
| | - Hiram Tendilla-Beltrán
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Julio César Morales-Medina
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
15
|
Wang R, Li L, Chen M, Li X, Liu Y, Xue Z, Ma Q, Chen J. Gene expression insights: Chronic stress and bipolar disorder: A bioinformatics investigation. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:392-414. [PMID: 38303428 DOI: 10.3934/mbe.2024018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Bipolar disorder (BD) is a psychiatric disorder that affects an increasing number of people worldwide. The mechanisms of BD are unclear, but some studies have suggested that it may be related to genetic factors with high heritability. Moreover, research has shown that chronic stress can contribute to the development of major illnesses. In this paper, we used bioinformatics methods to analyze the possible mechanisms of chronic stress affecting BD through various aspects. We obtained gene expression data from postmortem brains of BD patients and healthy controls in datasets GSE12649 and GSE53987, and we identified 11 chronic stress-related genes (CSRGs) that were differentially expressed in BD. Then, we screened five biomarkers (IGFBP6, ALOX5AP, MAOA, AIF1 and TRPM3) using machine learning models. We further validated the expression and diagnostic value of the biomarkers in other datasets (GSE5388 and GSE78936) and performed functional enrichment analysis, regulatory network analysis and drug prediction based on the biomarkers. Our bioinformatics analysis revealed that chronic stress can affect the occurrence and development of BD through many aspects, including monoamine oxidase production and decomposition, neuroinflammation, ion permeability, pain perception and others. In this paper, we confirm the importance of studying the genetic influences of chronic stress on BD and other psychiatric disorders and suggested that biomarkers related to chronic stress may be potential diagnostic tools and therapeutic targets for BD.
Collapse
Affiliation(s)
- Rongyanqi Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lan Li
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Man Chen
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiaojuan Li
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yueyun Liu
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhe Xue
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jiaxu Chen
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
16
|
Michael C, Taxali A, Angstadt M, Kardan O, Weigard A, Molloy MF, McCurry KL, Hyde LW, Heitzeg MM, Sripada C. Socioeconomic resources in youth are linked to divergent patterns of network integration and segregation across the brain's transmodal axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.565517. [PMID: 38014302 PMCID: PMC10680554 DOI: 10.1101/2023.11.08.565517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Socioeconomic resources (SER) calibrate the developing brain to the current context, which can confer or attenuate risk for psychopathology across the lifespan. Recent multivariate work indicates that SER levels powerfully influence intrinsic functional connectivity patterns across the entire brain. Nevertheless, the neurobiological meaning of these widespread alterations remains poorly understood, despite its translational promise for early risk identification, targeted intervention, and policy reform. In the present study, we leverage the resources of graph theory to precisely characterize multivariate and univariate associations between household SER and the functional integration and segregation (i.e., participation coefficient, within-module degree) of brain regions across major cognitive, affective, and sensorimotor systems during the resting state in 5,821 youth (ages 9-10 years) from the Adolescent Brain Cognitive Development (ABCD) Study. First, we establish that decomposing the brain into profiles of integration and segregation captures more than half of the multivariate association between SER and functional connectivity with greater parsimony (100-fold reduction in number of features) and interpretability. Second, we show that the topological effects of SER are not uniform across the brain; rather, higher SER levels are related to greater integration of somatomotor and subcortical systems, but greater segregation of default mode, orbitofrontal, and cerebellar systems. Finally, we demonstrate that the effects of SER are spatially patterned along the unimodal-transmodal gradient of brain organization. These findings provide critical interpretive context for the established and widespread effects of SER on brain organization, indicating that SER levels differentially configure the intrinsic functional architecture of developing unimodal and transmodal systems. This study highlights both sensorimotor and higher-order networks that may serve as neural markers of environmental stress and opportunity, and which may guide efforts to scaffold healthy neurobehavioral development among disadvantaged communities of youth.
Collapse
Affiliation(s)
- Cleanthis Michael
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Aman Taxali
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Mike Angstadt
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Omid Kardan
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Weigard
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - M. Fiona Molloy
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | | | - Luke W. Hyde
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center at the Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Mary M. Heitzeg
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Chandra Sripada
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
17
|
Brieler JA, Salas J, Amick ME, Sheth P, Keegan-Garrett EA, Morley JE, Scherrer JF. Anxiety disorders, benzodiazepine prescription, and incident dementia. J Am Geriatr Soc 2023; 71:3376-3389. [PMID: 37503956 DOI: 10.1111/jgs.18515] [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: 11/18/2022] [Revised: 05/26/2023] [Accepted: 06/24/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Prescribing benzodiazepines to older patients is controversial. Anxiety disorders and benzodiazepines have been associated with dementia, but literature is inconsistent. It is unknown if anxiety treated with a benzodiazepine, compared to anxiety disorder alone is associated with dementia risk. METHODS A retrospective cohort study (n = 72,496) was conducted using electronic health data from 2014 to 2021. Entropy balancing controlled for bias by indication and other confounding factors. PARTICIPANTS Eligible patients were ≥65 years old, had clinic encounters before and after index date and were free of dementia for 2 years prior to index date. Of the 72,496 eligible patients, 85.6% were White and 59.9% were female. Mean age was 74.1 (SD ± 7.1) years. EXPOSURE Anxiety disorder was a composite of generalized anxiety disorder, anxiety not otherwise specified, panic disorder, and social phobia. Sustained benzodiazepine use was defined as at least two separate prescription orders in any 6-month period. MAIN OUTCOME AND MEASURES ICD-9 or ICD-10 dementia diagnoses. RESULTS Six percent of eligible patients had an anxiety diagnosis and 3.6% received sustained benzodiazepine prescriptions. There were 6640 (9.2%) incident dementia events. After controlling for confounders, both sustained benzodiazepine use (HR 1.28, 95% CI: 1.11-1.47) and a diagnosis of anxiety (HR 1.19, 95% CI: 1.06-1.33) were associated with incident dementia in patients aged 65-75. Anxiety disorder with sustained benzodiazepine, compared to anxiety disorder alone, was not associated with incident dementia (HR 1.18, 95% CI: 0.92-1.51) after controlling for confounding. Results were not significant when limiting the sample to those ≥75 years of age. CONCLUSIONS Benzodiazepines and anxiety disorders are associated with increased risk for dementia. In patients with anxiety disorders, benzodiazepines were not associated with additional dementia risk. Further research is warranted to determine if benzodiazepines are associated with a reduced or increased risk for dementia compared to other anxiolytic medications in patients with anxiety disorders.
Collapse
Affiliation(s)
- Jay A Brieler
- Department of Family and Community Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Joanne Salas
- Department of Family and Community Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Harry S. Truman Veterans Administration Medical Center, Columbia, Missouri, USA
- The Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Matthew E Amick
- Department of Family and Community Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Poorva Sheth
- Department of Family and Community Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth A Keegan-Garrett
- Department of Family and Community Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - John E Morley
- School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Jeffrey F Scherrer
- Department of Family and Community Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Harry S. Truman Veterans Administration Medical Center, Columbia, Missouri, USA
- The Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry and Behavioral Neuroscience, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
18
|
Arnsten AFT, Ishizawa Y, Xie Z. Scientific rationale for the use of α2A-adrenoceptor agonists in treating neuroinflammatory cognitive disorders. Mol Psychiatry 2023; 28:4540-4552. [PMID: 37029295 PMCID: PMC10080530 DOI: 10.1038/s41380-023-02057-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Neuroinflammatory disorders preferentially impair the higher cognitive and executive functions of the prefrontal cortex (PFC). This includes such challenging disorders as delirium, perioperative neurocognitive disorder, and the sustained cognitive deficits from "long-COVID" or traumatic brain injury. There are no FDA-approved treatments for these symptoms; thus, understanding their etiology is important for generating therapeutic strategies. The current review describes the molecular rationale for why PFC circuits are especially vulnerable to inflammation, and how α2A-adrenoceptor (α2A-AR) actions throughout the nervous and immune systems can benefit the circuits in PFC needed for higher cognition. The layer III circuits in the dorsolateral PFC (dlPFC) that generate and sustain the mental representations needed for higher cognition have unusual neurotransmission and neuromodulation. They are wholly dependent on NMDAR neurotransmission, with little AMPAR contribution, and thus are especially vulnerable to kynurenic acid inflammatory signaling which blocks NMDAR. Layer III dlPFC spines also have unusual neuromodulation, with cAMP magnification of calcium signaling in spines, which opens nearby potassium channels to rapidly weaken connectivity and reduce neuronal firing. This process must be tightly regulated, e.g. by mGluR3 or α2A-AR on spines, to prevent loss of firing. However, the production of GCPII inflammatory signaling reduces mGluR3 actions and markedly diminishes dlPFC network firing. Both basic and clinical studies show that α2A-AR agonists such as guanfacine can restore dlPFC network firing and cognitive function, through direct actions in the dlPFC, but also by reducing the activity of stress-related circuits, e.g. in the locus coeruleus and amygdala, and by having anti-inflammatory actions in the immune system. This information is particularly timely, as guanfacine is currently the focus of large clinical trials for the treatment of delirium, and in open label studies for the treatment of cognitive deficits from long-COVID.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department Neuroscience, Yale University School of Medicine, New Haven, CT, 056510, USA.
| | - Yumiko Ishizawa
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhongcong Xie
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
19
|
Guo L, Wang S, Tian H, Shang M, Xu J, Wang C. Novel synergistic treatment for depression: involvement of GSK3β-regulated AMPA receptors in the prefrontal cortex of mice. Cereb Cortex 2023; 33:10504-10513. [PMID: 37566915 DOI: 10.1093/cercor/bhad299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Previous evidence has suggested a vital role of glycogen synthase kinase 3β-mediated α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors trafficking in depression. Considering the antidepressant effect of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors activation in the prefrontal cortex, we hypothesized that glycogen synthase kinase 3β-induced alterations in α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors function in the prefrontal cortex participate in depression. Herein, we confirmed that the levels of phosphorylated glycogen synthase kinase 3β and GluA1, the latter being a subunit of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors, were decreased in the prefrontal cortex of the chronic social defeat stress model mice presenting with depressive-like behaviors. We then found that a glycogen synthase kinase 3β (p.S9A) point mutation downregulated GluA1 and induced depressive-like behaviors in mice, whereas an agonist of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors, PF-4778574 (2 mg/kg) did not reversed the molecular changes. On the other hand, the antidepressant effect of PF-4778574 was dose dependent, and the single administration of PF-4778574 at a lower dose (0.5 mg/kg) or of the glycogen synthase kinase 3β inhibitor SB216763 (5 and 10 mg/kg) did not evoke an antidepressant effect. In contrast, co-treatment with PF-4778574 (0.5 mg/kg) and SB216763 (10 mg/kg) led to antidepressant effects similar to those of PF-4778574 (2 mg/kg). Our results suggest that glycogen synthase kinase 3β-induced α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors dysfunction in the prefrontal cortex is one of the key mechanisms of depression, and the combination of a lower dose of PF-4778574 with SB216763 shows potential as a novel synergistic treatment for depression.
Collapse
Affiliation(s)
- Lei Guo
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Shuzhuo Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Haihua Tian
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, PR China
| | - Mengyuan Shang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Jia Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| |
Collapse
|
20
|
Rakesh D, Whittle S, Sheridan MA, McLaughlin KA. Childhood socioeconomic status and the pace of structural neurodevelopment: accelerated, delayed, or simply different? Trends Cogn Sci 2023; 27:833-851. [PMID: 37179140 PMCID: PMC10524122 DOI: 10.1016/j.tics.2023.03.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 05/15/2023]
Abstract
Socioeconomic status (SES) is associated with children's brain and behavioral development. Several theories propose that early experiences of adversity or low SES can alter the pace of neurodevelopment during childhood and adolescence. These theories make contrasting predictions about whether adverse experiences and low SES are associated with accelerated or delayed neurodevelopment. We contextualize these predictions within the context of normative development of cortical and subcortical structure and review existing evidence on SES and structural brain development to adjudicate between competing hypotheses. Although none of these theories are fully consistent with observed SES-related differences in brain development, existing evidence suggests that low SES is associated with brain structure trajectories more consistent with a delayed or simply different developmental pattern than an acceleration in neurodevelopment.
Collapse
Affiliation(s)
| | - Sarah Whittle
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Victoria, Australia
| | - Margaret A Sheridan
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
21
|
Tripathi A, Bartosh A, Whitehead C, Pillai A. Activation of cell-free mtDNA-TLR9 signaling mediates chronic stress-induced social behavior deficits. Mol Psychiatry 2023; 28:3806-3815. [PMID: 37528226 PMCID: PMC10730412 DOI: 10.1038/s41380-023-02189-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Inflammation and social behavior deficits are associated with a number of neuropsychiatric disorders. Chronic stress, a major risk factor for depression and other mental health conditions is known to increase inflammatory responses and social behavior impairments. Disturbances in mitochondria function have been found in chronic stress conditions, however the mechanisms that link mitochondrial dysfunction to stress-induced social behavior deficits are not well understood. In this study, we found that chronic restraint stress (RS) induces significant increases in serum cell-free mitochondrial DNA (cf-mtDNA) levels in mice, and systemic Deoxyribonuclease I (DNase I) treatment attenuated RS-induced social behavioral deficits. Our findings revealed potential roles of mitophagy and Mitochondrial antiviral-signaling protein (MAVS) in mediating chronic stress-induced changes in cf-mtDNA levels and social behavior. Furthermore, we showed that inhibition of Toll-like receptor 9 (TLR9) attenuates mtDNA-induced social behavior deficits. Together, these findings show that cf-mtDNA-TLR9 signaling is critical in mediating stress-induced social behavior deficits.
Collapse
Affiliation(s)
- Ashutosh Tripathi
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Alona Bartosh
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Carl Whitehead
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Department of Psychiatry and Health Behavior, Augusta University, Augusta, GA, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
22
|
Walton NL, Antonoudiou P, Maguire JL. Neurosteroid influence on affective tone. Neurosci Biobehav Rev 2023; 152:105327. [PMID: 37499891 PMCID: PMC10528596 DOI: 10.1016/j.neubiorev.2023.105327] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/07/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
Affective disorders such as depression and anxiety are among the most prevalent psychiatric illnesses and causes of disability worldwide. The recent FDA-approval of a novel antidepressant treatment, ZULRESSO® (Brexanolone), a synthetic neurosteroid has fueled interest into the role of neurosteroids in the pathophysiology of depression as well as the mechanisms mediating the antidepressant effects of these compounds. The majority of studies examining the impact of neurosteroids on affective states have relied on the administration of exogenous neurosteroids; however, neurosteroids can also be synthesized endogenously from cholesterol or steroid hormone precursors. Despite the well-established influence of exogenous neurosteroids on affective states, we still lack an understanding of the role of endogenous neurosteroids in modulating affective tone. This review aims to summarize the current literature supporting the influence of neurosteroids on affective states in clinical and preclinical studies, as well as recent evidence suggesting that endogenous neurosteroids may set a baseline affective tone.
Collapse
Affiliation(s)
- Najah L Walton
- Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Pantelis Antonoudiou
- Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jamie L Maguire
- Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
23
|
Pickering G, Noah L, Pereira B, Goubayon J, Leray V, Touron A, Macian N, Bernard L, Dualé C, Roux V, Chassain C. Assessing brain function in stressed healthy individuals following the use of a combination of green tea, Rhodiola, magnesium, and B vitamins: an fMRI study. Front Nutr 2023; 10:1211321. [PMID: 37662591 PMCID: PMC10469327 DOI: 10.3389/fnut.2023.1211321] [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: 04/24/2023] [Accepted: 07/13/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction This randomized, controlled, single-blinded trial assessed the effect of magnesium (Mg)-Teadiola (Mg, vitamins B6, B9, B12, Rhodiola, and green tea/L-theanine) versus placebo on the brain response to stressful thermal stimulus in chronically stressed, but otherwise healthy subjects. Impacts on stress-related quality-of-life parameters (depression, anxiety, sleep, and perception of pain) were also explored. Methods The study recruited a total of 40 adults (20 per group), suffering from stress for more than 1 month and scaling ≥14 points on the Depression Anxiety Stress Scale (DASS)-42 questionnaire at the time of inclusion. Individuals received oral Mg-Teadiola or placebo for 28 days (D). fMRI analysis was used to visualize the interplay between stress and pain cerebral matrices, using thermal stress model, at baseline (D0) and after D28. Results Based on blood-oxygen-level-dependent (BOLD) signal variations during the stress stimulation (before pain perception), a significantly increased activation between D0 and D28 was observed for left and right frontal area (p = 0.001 and p = 0.002, respectively), left and right anterior cingulate cortex (ACC) (p = 0.035 and p = 0.04, respectively), and left and right insula (p = 0.034 and p = 0.0402, respectively) in Mg-Teadiola versus placebo group. During thermal pain stimulation, a significantly diminished activation of the pain matrix was observed between D0 and D28, for left and right prefrontal area (both p = 0.001), left and right insula (p = 0.008 and p = 0.019, respectively), and left and right ventral striatum (both p = 0.001) was observed in Mg-Teadiola versus placebo group. These results reinforce the clinical observations, showing a perceived benefit of Mg-Teadiola on several parameters. After 1 month of treatment, DASS-42 stress score significantly decreased in Mg-Teadiola group [effect size (ES) -0.46 (-0.91; -0.01), p = 0.048]. Similar reductions were observed on D14 (p = 0.011) and D56 (p = 0.008). Sensitivity to cold also improved from D0 to D28 for Mg-Teadiola versus placebo [ES 0.47 (0.02; 0.92) p = 0.042]. Conclusion Supplementation with Mg-Teadiola reduced stress on D28 in chronically stressed but otherwise healthy individuals and modulated the stress and pain cerebral matrices during stressful thermal stimulus.
Collapse
Affiliation(s)
- Gisèle Pickering
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
- Department of Pharmacology, University Clermont Auvergne, Inserm, Clermont-Ferrand, France
| | | | - Bruno Pereira
- Clinical Research and Innovation Department, University Hospital Clermont-Ferrand, Clermont-Ferrand, France
| | - Jonathan Goubayon
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
- Sanofi, Gentilly, France
| | - Vincent Leray
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
| | - Ambre Touron
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
| | - Nicolas Macian
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
| | - Lise Bernard
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, CHU Clermont Ferrand, ICCF, Clermont-Ferrand, France
| | - Christian Dualé
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
- Department of Pharmacology, University Clermont Auvergne, Inserm, Clermont-Ferrand, France
| | - Veronique Roux
- Platform of Clinical Investigation Department, University Hospital Clermont-Ferrand, INSERM CIC, Clermont-Ferrand, France
| | - Carine Chassain
- Université Clermont Auvergne, CHU, CNRS, Clermont Auvergne INP, Institut Pascal, Clermont-Ferrand, France
| |
Collapse
|
24
|
Meftahi GH, Hatef B, Pirzad Jahromi G. Creatine Activity as a Neuromodulator in the Central Nervous System. ARCHIVES OF RAZI INSTITUTE 2023; 78:1169-1175. [PMID: 38226371 PMCID: PMC10787915 DOI: 10.32592/ari.2023.78.4.1169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/05/2023] [Indexed: 01/17/2024]
Abstract
Creatine is a nutritional compound that potentially influences cognitive processing and neuroprotection. Recent evidence has demonstrated that similar to neurotransmitters, creatine is released in an excitotoxic and action potential-dependent manner and acts as a neuromodulator. Creatine deficiency syndromes are characterized by severe mental and developmental disorders. Studies have reported that brain creatine content could be enhanced with creatine supplementation. Nevertheless, there is still limited knowledge about the effects of creatine on the central nervous system. However, ample evidence has proved the neuroprotective effects of creatine on various mental aspects, such as cognition, memory skills, and spatial memory. The present review aimed to review available experimental data and clinical observations confirming creatine roles in the central transmission process. A systematic search in the literature was performed in PubMed, Scopus, Embase, Cochrane Library, Web of Science, and Google Scholar database using all available MeSH terms for Creatine, Phosphocreatine, Bioenergetics, Nervous system, Brain, Cognition, and Neuroprotection. Electronic database searches were combined and duplicates were removed. Here, first, creatine and its potential influence on cognitive health and performance were briefly reviewed. Next, the existing experimental and clinical evidence was specifically explored to understand how creatine could interact as a neurotransmitter in the nervous system. Studies have revealed that exogenous creatine supplementation decreases neuronal cell loss in experimental paradigms of neurological diseases. It was observed that creatine could interact with the N-methyl-D-aspartate receptor, Na+-K+-ATPase enzyme, GABAA receptor, serotonin 1A receptors, and presumably α1-adrenoceptor and play critical roles in the central transmission process which implies that creatine can be considered a neuromodulator.
Collapse
Affiliation(s)
- G H Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - B Hatef
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - G Pirzad Jahromi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Rai AR, Joy T, Poojari M, Pai MM, Massand A, Murlimanju BV. Role of Acorus calamus in preventing depression, anxiety, and oxidative stress in long-term socially isolated rats. Vet World 2023; 16:1755-1764. [PMID: 37766700 PMCID: PMC10521175 DOI: 10.14202/vetworld.2023.1755-1764] [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: 04/27/2023] [Accepted: 07/22/2023] [Indexed: 09/29/2023] Open
Abstract
Background and Aim Social isolation stress (SIS) and individual housing have been shown to cause abnormal cognitive insufficiencies, altered anxiety levels, and signs of psychiatric diseases. Acorus calamus (AC), commonly known as Sweet Flag, has been widely used in India to treat neurological, metabolic, and respiratory disorders, indicating its potential therapeutic value. This study aimed to determine the antidepressant and antioxidative effects of AC on rats subjected to long-term, social isolation-induced stress. Materials and Methods This study involved 2-month-old male rats (24) weighing approximately 180-200 g bred in-house. The rats were divided into four groups (n = 6): Group 1 received saline, Group 2 received SIS, Group 3 received only 50 mg/kg AC, and Group 4 received 50 mg/kg AC and SIS for 6 weeks. After this, behavioral, biochemical, and neuronal assays were conducted. Results Behavioral experiments showed significantly higher activity levels (p < 0.001) in AC-treated rats than in the SIS group. In addition, rats subjected to SIS with AC treatment exhibited enhanced total antioxidants, superoxide dismutase, and neuronal assays compared to rats subjected to SIS alone. Conclusion Acorus calamus treatment improved the antidepressant and antioxidant potential against SIS in rat brain tissue. Moreover, we proved that AC can effectively reverse the neurotoxicity induced by SIS in animal models. As we battle against the coronavirus disease 2019 pandemic and social isolation, AC could be considered a supplementary treatment to alleviate depressive-like symptoms in our present-day lifestyle.
Collapse
Affiliation(s)
- Ashwin Rohan Rai
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Teresa Joy
- Department of Anatomy, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Coolidge, St. John’s, Antigua, West Indies
| | - Meghana Poojari
- Department of Anatomy, Basaveshwara Medical College and Hospital, Chitradurga, India
| | - Mangala M. Pai
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Amit Massand
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - B. V. Murlimanju
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
26
|
Graves CL, Norloff E, Thompson D, Kosyk O, Sang Y, Chen A, Zannas AS, Wallet SM. Chronic early life stress alters the neuroimmune profile and functioning of the developing zebrafish gut. Brain Behav Immun Health 2023; 31:100655. [PMID: 37449287 PMCID: PMC10336164 DOI: 10.1016/j.bbih.2023.100655] [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: 02/25/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Chronic early life stress (ELS) potently impacts the developing central nervous and immune systems and is associated with the onset of gastrointestinal disease in humans. Though the gut-brain axis is appreciated to be a major target of the stress response, the underlying mechanisms linking ELS to gut dysfunction later in life is incompletely understood. Zebrafish are a powerful model validated for stress research and have emerged as an important tool in delineating neuroimmune mechanisms in the developing gut. Here, we developed a novel model of ELS and utilized a comparative transcriptomics approach to assess how chronic ELS modulated expression of neuroimmune genes in the developing gut and brain. Zebrafish exposed to ELS throughout larval development exhibited anxiety-like behavior and altered expression of neuroimmune genes in a time- and tissue-dependent manner. Further, the altered gut neuroimmune profile, which included increased expression of genes associated with neuronal modulation, correlated with a reduction in enteric neuronal density and delayed gut transit. Together, these findings provide insights into the mechanisms linking ELS with gastrointestinal dysfunction and highlight the zebrafish model organism as a valuable tool in uncovering how "the body keeps the score."
Collapse
Affiliation(s)
- Christina L. Graves
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Carolina Stress Initiative, University of North Carolina School of Medicine, Chapel Hill, NC, 27514, USA
| | - Erik Norloff
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Darius Thompson
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Oksana Kosyk
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yingning Sang
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Angela Chen
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Anthony S. Zannas
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Carolina Stress Initiative, University of North Carolina School of Medicine, Chapel Hill, NC, 27514, USA
| | - Shannon M. Wallet
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
27
|
Coelho A, Lima-Bastos S, Gobira P, Lisboa S. Endocannabinoid signaling and epigenetics modifications in the neurobiology of stress-related disorders. Neuronal Signal 2023; 7:NS20220034. [PMID: 37520658 PMCID: PMC10372471 DOI: 10.1042/ns20220034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
Stress exposure is associated with psychiatric conditions, such as depression, anxiety, and post-traumatic stress disorder (PTSD). It is also a vulnerability factor to developing or reinstating substance use disorder. Stress causes several changes in the neuro-immune-endocrine axis, potentially resulting in prolonged dysfunction and diseases. Changes in several transmitters, including serotonin, dopamine, glutamate, gamma-aminobutyric acid (GABA), glucocorticoids, and cytokines, are associated with psychiatric disorders or behavioral alterations in preclinical studies. Complex and interacting mechanisms make it very difficult to understand the physiopathology of psychiatry conditions; therefore, studying regulatory mechanisms that impact these alterations is a good approach. In the last decades, the impact of stress on biology through epigenetic markers, which directly impact gene expression, is under intense investigation; these mechanisms are associated with behavioral alterations in animal models after stress or drug exposure, for example. The endocannabinoid (eCB) system modulates stress response, reward circuits, and other physiological functions, including hypothalamus-pituitary-adrenal axis activation and immune response. eCBs, for example, act retrogradely at presynaptic neurons, limiting the release of neurotransmitters, a mechanism implicated in the antidepressant and anxiolytic effects after stress. Epigenetic mechanisms can impact the expression of eCB system molecules, which in turn can regulate epigenetic mechanisms. This review will present evidence of how the eCB system and epigenetic mechanisms interact and the consequences of this interaction in modulating behavioral changes after stress exposure in preclinical studies or psychiatric conditions. Moreover, evidence that correlates the involvement of the eCB system and epigenetic mechanisms in drug abuse contexts will be discussed.
Collapse
Affiliation(s)
- Arthur A. Coelho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | - Sávio Lima-Bastos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | - Pedro H. Gobira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | - Sabrina F. Lisboa
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| |
Collapse
|
28
|
Fesharaki Zadeh A, Arnsten AFT, Wang M. Scientific Rationale for the Treatment of Cognitive Deficits from Long COVID. Neurol Int 2023; 15:725-742. [PMID: 37368329 PMCID: PMC10303664 DOI: 10.3390/neurolint15020045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/26/2023] [Accepted: 05/11/2023] [Indexed: 06/28/2023] Open
Abstract
Sustained cognitive deficits are a common and debilitating feature of "long COVID", but currently there are no FDA-approved treatments. The cognitive functions of the dorsolateral prefrontal cortex (dlPFC) are the most consistently afflicted by long COVID, including deficits in working memory, motivation, and executive functioning. COVID-19 infection greatly increases kynurenic acid (KYNA) and glutamate carboxypeptidase II (GCPII) in brain, both of which can be particularly deleterious to PFC function. KYNA blocks both NMDA and nicotinic-alpha-7 receptors, the two receptors required for dlPFC neurotransmission, and GCPII reduces mGluR3 regulation of cAMP-calcium-potassium channel signaling, which weakens dlPFC network connectivity and reduces dlPFC neuronal firing. Two agents approved for other indications may be helpful in restoring dlPFC physiology: the antioxidant N-acetyl cysteine inhibits the production of KYNA, and the α2A-adrenoceptor agonist guanfacine regulates cAMP-calcium-potassium channel signaling in dlPFC and is also anti-inflammatory. Thus, these agents may be helpful in treating the cognitive symptoms of long COVID.
Collapse
Affiliation(s)
- Arman Fesharaki Zadeh
- Departments of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Amy F. T. Arnsten
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Min Wang
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA;
| |
Collapse
|
29
|
Mahmud A, Avramescu RG, Niu Z, Flores C. Awakening the dormant: Role of axonal guidance cues in stress-induced reorganization of the adult prefrontal cortex leading to depression-like behavior. Front Neural Circuits 2023; 17:1113023. [PMID: 37035502 PMCID: PMC10079902 DOI: 10.3389/fncir.2023.1113023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic and disabling disorder affecting roughly 280 million people worldwide. While multiple brain areas have been implicated, dysfunction of prefrontal cortex (PFC) circuitry has been consistently documented in MDD, as well as in animal models for stress-induced depression-like behavioral states. During brain development, axonal guidance cues organize neuronal wiring by directing axonal pathfinding and arborization, dendritic growth, and synapse formation. Guidance cue systems continue to be expressed in the adult brain and are emerging as important mediators of synaptic plasticity and fine-tuning of mature neural networks. Dysregulation or interference of guidance cues has been linked to depression-like behavioral abnormalities in rodents and MDD in humans. In this review, we focus on the emerging role of guidance cues in stress-induced changes in adult prefrontal cortex circuitry and in precipitating depression-like behaviors. We discuss how modulating axonal guidance cue systems could be a novel approach for precision medicine and the treatment of depression.
Collapse
Affiliation(s)
- Ashraf Mahmud
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | | | - Zhipeng Niu
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Department of Psychiatry, Neurology, and Neurosurgery, McGill University, Montréal, QC, Canada
| |
Collapse
|
30
|
Singh D, Preetam Ambati A, Aich P. Sex and Time: Important Variables for Understanding the Impact of Constant Darkness on Behavior, Brain, and Physiology. Neuroscience 2023; 519:73-89. [PMID: 36966879 DOI: 10.1016/j.neuroscience.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
The circadian clock can coordinate, regulate and predict physiology and behavior in response to the standard light-dark (LD: 12 h light and 12 h dark) cycle. If we alter the LD cycle by exposing mice to constant darkness (DD: 00 h light and 24 h dark), it can perturb behavior, the brain, and associated physiological parameters. The length of DD exposure and the sex of experimental animals are crucial variables that could alter the impact of DD on the brain, behavior, and physiology, which have not yet been explored. We exposed mice to DD for three and five weeks and studied their impact on (1) behavior, (2) hormones, (3) the prefrontal cortex, and (4) metabolites in male and female mice. We also studied the effect of three weeks of standard light-dark cycle restoration after five weeks of DD on the parameters mentioned above. We found that DD exposure was associated with anxiety-like behavior, increased corticosterone and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), downregulated neurotrophins (BDNF and NGF), and altered metabolites profile in a duration of DD exposure and sex-dependent manner. Females showed a more robust adaptation than males under DD exposure. Three weeks of restoration was adequate to establish homeostasis in both sexes. To the best of our knowledge, this study is the first of its kind to look at how DD exposure impacts physiology and behavior as a function of sex- and time. These findings would have translational value and may help in establishing sex-specific interventions for addressing DD-related psychological issues.
Collapse
Affiliation(s)
- Dhyanendra Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Abhilash Preetam Ambati
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
31
|
Woodburn SC, Asrat HS, Flurer JK, Schwierling HC, Bollinger JL, Vollmer LL, Wohleb ES. Depletion of microglial BDNF increases susceptibility to the behavioral and synaptic effects of chronic unpredictable stress. Brain Behav Immun 2023; 109:127-138. [PMID: 36681359 PMCID: PMC10023455 DOI: 10.1016/j.bbi.2023.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/22/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
In the medial prefrontal cortex (PFC), chronic stress reduces synaptic expression of glutamate receptors, leading to decreased excitatory signaling from layer V pyramidal neurons and working memory deficits. One key element driving these changes is a reduction in brain-derived neurotrophic factor (BDNF) signaling. BDNF is a potent mediator of synaptic growth and deficient BDNF signaling has been linked to stress susceptibility. Prior studies indicated that neurons are the primary source of BDNF, but more recent work suggests that microglia are also an important source of BDNF. Adding to this, our work showed that 14 days of chronic unpredictable stress (CUS) reduced Bdnf transcript in PFC microglia, evincing its relevance in the effects of stress. To explore this further, we utilized transgenic mice with microglia-specific depletion of BDNF (Cx3cr1Cre/+:Bdnffl/fl) and genotype controls (Cx3cr1Cre/+:Bdnf+/+). In the following experiments, mice were exposed to a shortened CUS paradigm (7 days) to determine if microglial Bdnf depletion promotes stress susceptibility. Analyses of PFC microglia revealed that Cx3cr1Cre/+:Bdnffl/fl mice had shifts in phenotypic markers and gene expression. In a separate cohort, synaptoneurosomes were collected from the PFC and western blotting was performed for synaptic markers. These experiments showed that Cx3cr1Cre/+:Bdnffl/fl mice had baseline deficits in GluN2B, and that 7 days of CUS additionally reduced GluN2A levels in Cx3cr1Cre/+:Bdnffl/fl mice, but not genotype controls. Behavioral and cognitive testing showed that this coincided with exacerbated stress effects on temporal object recognition in Cx3cr1Cre/+:Bdnffl/fl mice. These results indicate that microglial BDNF promotes glutamate receptor expression in the PFC. As such, mice with deficient microglial BDNF had increased susceptibility to the behavioral and cognitive consequences of stress.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Helina S Asrat
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James K Flurer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hana C Schwierling
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lauren L Vollmer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
32
|
Rossetti AC, Paladini MS, Brüning CA, Spero V, Cattaneo MG, Racagni G, Papp M, Riva MA, Molteni R. Involvement of the IL-6 Signaling Pathway in the Anti-Anhedonic Effect of the Antidepressant Agomelatine in the Chronic Mild Stress Model of Depression. Int J Mol Sci 2022; 23:ijms232012453. [PMID: 36293308 PMCID: PMC9604470 DOI: 10.3390/ijms232012453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 12/03/2022] Open
Abstract
Neuroinflammation has emerged as an important factor in the molecular underpinnings of major depressive disorder (MDD) pathophysiology and in the mechanism of action of antidepressants. Among the inflammatory mediators dysregulated in depressed patients, interleukin (IL)-6 has recently been proposed to play a crucial role. IL-6 activates a signaling pathway comprising the JAK/STAT proteins and characterized by a specific negative feedback loop exerted by the cytoplasmic protein suppressor of cytokine signalling-3 (SOCS3). On these bases, here, we explored the potential involvement of IL-6 signaling in the ability of the antidepressant drug agomelatine to normalize the anhedonic-like phenotype induced in the rat by chronic stress exposure. To this aim, adult male Wistar rats were subjected to the chronic mild stress (CMS) paradigm and chronically treated with vehicle or agomelatine. The behavioral evaluation was assessed by the sucrose consumption test, whereas molecular analyses were performed in the prefrontal cortex. We found that CMS was able to stimulate IL-6 production and signaling, including SOCS3 gene and protein expression, but the SOCS3-mediated feedback-loop inhibition failed to suppress the IL-6 cascade in stressed animals. Conversely, agomelatine treatment normalized the stress-induced decrease in sucrose consumption and restored the negative modulation of the IL-6 signaling via SOCS3 expression and activity. Our results provide additional information about the pleiotropic mechanisms that contribute to agomelatine’s therapeutic effects.
Collapse
Affiliation(s)
- Andrea C. Rossetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Cesar Augusto Brüning
- Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas, Pelotas 96010-900, RS, Brazil
| | - Vittoria Spero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Giorgio Racagni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Marco A. Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
- Correspondence:
| |
Collapse
|
33
|
Musazzi L, Tornese P, Sala N, Lee FS, Popoli M, Ieraci A. Acute stress induces an aberrant increase of presynaptic release of glutamate and cellular activation in the hippocampus of BDNF Val/Met mice. J Cell Physiol 2022; 237:3834-3844. [PMID: 35908196 PMCID: PMC9796250 DOI: 10.1002/jcp.30833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/15/2022] [Accepted: 07/11/2022] [Indexed: 01/01/2023]
Abstract
Stressful life events are considered major risk factors for the development of several psychiatric disorders, though people differentially cope with stress. The reasons for this are still largely unknown but could be accounted for by individual genetic variants, previous life events, or the kind of stressors. The human brain-derived neurotrophic factor (BDNF) Val66Met variant, which was found to impair intracellular trafficking and activity-dependent secretion of BDNF, has been associated with increased susceptibility to develop several neuropsychiatric disorders, although there is still some controversial evidence. On the other hand, acute stress has been consistently demonstrated to promote the release of glutamate in cortico-limbic regions and altered glutamatergic transmission has been reported in psychiatric disorders. However, it is not known if the BDNF Val66Met single-nucleotide polymorphism (SNP) affects the stress-induced presynaptic glutamate release. In this study, we exposed adult male BDNFVal/Val and BDNFVal/Met knock-in mice to 30 min of acute restraint stress. Plasma corticosterone levels, glutamate release, protein, and gene expression in the hippocampus were analyzed immediately after the end of the stress session. Acute restraint stress similarly increased plasma corticosterone levels and nuclear glucocorticoid receptor levels and phosphorylation in both BDNFVal/Val and BDNFVal/Met mice. However, acute restraint stress induced higher increases in hippocampal presynaptic release of glutamate, phosphorylation of cAMP-response element binding protein (CREB), and levels of the immediate early gene c-fos of BDNFVal/Met compared to BFNFVal/Val mice. Moreover, acute restraint stress selectively increased phosphorylation levels of synapsin I at Ser9 and at Ser603 in BDNFVal/Val and BDNFVal/Met mice, respectively. In conclusion, we report here that the BDNF Val66Met SNP knock-in mice display an altered response to acute restraint stress in terms of hippocampal glutamate release, CREB phosphorylation, and neuronal activation, compared to wild-type animals. Taken together, these results could partially explain the enhanced vulnerability to stressful events of Met carriers reported in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Laura Musazzi
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Paolo Tornese
- Dipartimento di Scienze FarmaceuticheUniversity of MilanMilanItaly
| | - Nathalie Sala
- Dipartimento di Scienze FarmaceuticheUniversity of MilanMilanItaly
| | - Francis S. Lee
- Department of PsychiatryWeill Cornell Medical CollegeNew YorkNew YorkUSA
| | - Maurizio Popoli
- Dipartimento di Scienze FarmaceuticheUniversity of MilanMilanItaly
| | | |
Collapse
|
34
|
Góralczyk-Bińkowska A, Szmajda-Krygier D, Kozłowska E. The Microbiota-Gut-Brain Axis in Psychiatric Disorders. Int J Mol Sci 2022; 23:11245. [PMID: 36232548 PMCID: PMC9570195 DOI: 10.3390/ijms231911245] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Modulating the gut microbiome and its influence on human health is the subject of intense research. The gut microbiota could be associated not only with gastroenterological diseases but also with psychiatric disorders. The importance of factors such as stress, mode of delivery, the role of probiotics, circadian clock system, diet, and occupational and environmental exposure in the relationship between the gut microbiota and brain function through bidirectional communication, described as "the microbiome-gut-brain axis", is especially underlined. In this review, we discuss the link between the intestinal microbiome and the brain and host response involving different pathways between the intestinal microbiota and the nervous system (e.g., neurotransmitters, endocrine system, immunological mechanisms, or bacterial metabolites). We review the microbiota alterations and their results in the development of psychiatric disorders, including major depressive disorder (MDD), schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), and attention-deficit hyperactivity disorder (ADHD).
Collapse
Affiliation(s)
- Aleksandra Góralczyk-Bińkowska
- Department of Microbiology and Experimental Immunology, MOLecoLAB: Lodz Centre of Molecular Studies on Civilisation Diseases, Medical University of Lodz, Mazowiecka 5 Street, 92-215 Lodz, Poland
| | - Dagmara Szmajda-Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Elżbieta Kozłowska
- Department of Microbiology and Experimental Immunology, MOLecoLAB: Lodz Centre of Molecular Studies on Civilisation Diseases, Medical University of Lodz, Mazowiecka 5 Street, 92-215 Lodz, Poland
| |
Collapse
|
35
|
Arnsten AFT, Woo E, Yang S, Wang M, Datta D. Unusual Molecular Regulation of Dorsolateral Prefrontal Cortex Layer III Synapses Increases Vulnerability to Genetic and Environmental Insults in Schizophrenia. Biol Psychiatry 2022; 92:480-490. [PMID: 35305820 PMCID: PMC9372235 DOI: 10.1016/j.biopsych.2022.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 02/06/2023]
Abstract
Schizophrenia is associated with reduced numbers of spines and dendrites from layer III of the dorsolateral prefrontal cortex (dlPFC), the layer that houses the recurrent excitatory microcircuits that subserve working memory and abstract thought. Why are these synapses so vulnerable, while synapses in deeper or more superficial layers are little affected? This review describes the special molecular properties that govern layer III neurotransmission and neuromodulation in the primate dlPFC and how they may render these circuits particularly vulnerable to genetic and environmental insults. These properties include a reliance on NMDA receptor rather than AMPA receptor neurotransmission; cAMP (cyclic adenosine monophosphate) magnification of calcium signaling near the glutamatergic synapse of dendritic spines; and potassium channels opened by cAMP/PKA (protein kinase A) signaling that dynamically alter network strength, with built-in mechanisms to take dlPFC "offline" during stress. A variety of genetic and/or environmental insults can lead to the same phenotype of weakened layer III connectivity, in which mechanisms that normally strengthen connectivity are impaired and those that normally weaken connectivity are intensified. Inflammatory mechanisms, such as increased kynurenic acid and glutamate carboxypeptidase II expression, are especially detrimental to layer III dlPFC neurotransmission and modulation, mimicking genetic insults. The combination of genetic and inflammatory insults may cross the threshold into pathology.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut.
| | - Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Shengtao Yang
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Min Wang
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| |
Collapse
|
36
|
Rosen JB, Schulkin J. Hyperexcitability: From Normal Fear to Pathological Anxiety and Trauma. Front Syst Neurosci 2022; 16:727054. [PMID: 35993088 PMCID: PMC9387392 DOI: 10.3389/fnsys.2022.727054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Hyperexcitability in fear circuits is suggested to be important for development of pathological anxiety and trauma from adaptive mechanisms of fear. Hyperexcitability is proposed to be due to acquired sensitization in fear circuits that progressively becomes more severe over time causing changing symptoms in early and late pathology. We use the metaphor and mechanisms of kindling to examine gains and losses in function of one excitatory and one inhibitory neuropeptide, corticotrophin releasing factor and somatostatin, respectively, to explore this sensitization hypothesis. We suggest amygdala kindling induced hyperexcitability, hyper-inhibition and loss of inhibition provide clues to mechanisms for hyperexcitability and progressive changes in function initiated by stress and trauma.
Collapse
Affiliation(s)
- Jeffrey B. Rosen
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
- *Correspondence: Jeffrey B. Rosen,
| | - Jay Schulkin
- School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
37
|
Saulskaya NB, Burmakina MA, Trofimova NA. Effect of Activation and Blockade of Nitrergic Neurotransmission on Serotonin System Activity of the Rat Medial Prefrontal Cortex. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
38
|
Csabai D, Sebők-Tornai A, Wiborg O, Czéh B. A Preliminary Quantitative Electron Microscopic Analysis Reveals Reduced Number of Mitochondria in the Infralimbic Cortex of Rats Exposed to Chronic Mild Stress. Front Behav Neurosci 2022; 16:885849. [PMID: 35600987 PMCID: PMC9115382 DOI: 10.3389/fnbeh.2022.885849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
Exposure to severe, uncontrollable and long-lasting stress is a strong risk factor for the development of numerous mental and somatic disorders. Animal studies document that chronic stress can alter neuronal morphology and functioning in limbic brain structures such as the prefrontal cortex. Mitochondria are intracellular powerhouses generating chemical energy for biochemical reactions of the cell. Recent findings document that chronic stress can lead to changes in mitochondrial function and metabolism. Here, we studied putative mitochondrial damage in response to chronic stress in neurons of the medial prefrontal cortex. We performed a systematic quantitative ultrastructural analysis to examine the consequences of 9-weeks of chronic mild stress on mitochondria number and morphology in the infralimbic cortex of adult male rats. In this preliminary study, we analyzed 4,250 electron microscopic images and 67000 mitochondria were counted and examined in the brains of 4 control and 4 stressed rats. We found significantly reduced number of mitochondria in the infralimbic cortex of the stressed animals, but we could not detect any significant alteration in mitochondrial morphology. These data support the concept that prolonged stress can lead to mitochondrial loss. This in turn may result in impaired energy production. Reduced cellular energy may sensitize the neurons to additional injuries and may eventually trigger the development of psychopathologies.
Collapse
Affiliation(s)
- Dávid Csabai
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Abigél Sebők-Tornai
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
39
|
Lee MT, Peng WH, Kan HW, Wu CC, Wang DW, Ho YC. Neurobiology of Depression: Chronic Stress Alters the Glutamatergic System in the Brain-Focusing on AMPA Receptor. Biomedicines 2022; 10:biomedicines10051005. [PMID: 35625742 PMCID: PMC9138646 DOI: 10.3390/biomedicines10051005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/25/2022] Open
Abstract
Major depressive disorder (MDD) is a common neuropsychiatric disorder affecting the mood and mental well-being. Its pathophysiology remains elusive due to the complexity and heterogeneity of this disorder that affects millions of individuals worldwide. Chronic stress is frequently cited as the one of the risk factors for MDD. To date, the conventional monoaminergic theory (serotonin, norepinephrine, and/or dopamine dysregulation) has received the most attention in the treatment of MDD, and all available classes of antidepressants target these monoaminergic systems. However, the contributions of other neurotransmitter systems in MDD have been widely reported. Emerging preclinical and clinical findings reveal that maladaptive glutamatergic neurotransmission might underlie the pathophysiology of MDD, thus revealing its critical role in the neurobiology of MDD and as the therapeutic target. Aiming beyond the monoaminergic hypothesis, studies of the neurobiological mechanisms underlying the stress-induced impairment of AMPA (a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-glutamatergic neurotransmission in the brain could provide novel insights for the development of a new generation of antidepressants without the detrimental side effects. Here, the authors reviewed the recent literature focusing on the role of AMPA-glutamatergic neurotransmission in stress-induced maladaptive responses in emotional and mood-associated brain regions, including the hippocampus, amygdala, prefrontal cortex, nucleus accumbens and periaqueductal gray.
Collapse
Affiliation(s)
- Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Wei-Hao Peng
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (W.-H.P.); (H.-W.K.)
| | - Hung-Wei Kan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (W.-H.P.); (H.-W.K.)
| | - Cheng-Chun Wu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-C.W.); (D.-W.W.)
| | - Deng-Wu Wang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-C.W.); (D.-W.W.)
- Department of Psychiatry, E-Da Hospital, Kaohsiung City 82445, Taiwan
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-C.W.); (D.-W.W.)
- Correspondence:
| |
Collapse
|
40
|
Tubbs AS, Fernandez FX, Grandner MA, Perlis ML, Klerman EB. The Mind After Midnight: Nocturnal Wakefulness, Behavioral Dysregulation, and Psychopathology. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 1:830338. [PMID: 35538929 PMCID: PMC9083440 DOI: 10.3389/fnetp.2021.830338] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
Sufficient sleep with minimal interruption during the circadian/biological night supports daytime cognition and emotional regulation. Conversely, disrupted sleep involving significant nocturnal wakefulness leads to cognitive and behavioral dysregulation. Most studies to-date have examined how fragmented or insufficient sleep affects next-day functioning, but recent work highlights changes in cognition and behavior that occur when someone is awake during the night. This review summarizes the evidence for day-night alterations in maladaptive behaviors, including suicide, violent crime, and substance use, and examines how mood, reward processing, and executive function differ during nocturnal wakefulness. Based on this evidence, we propose the Mind after Midnight hypothesis in which attentional biases, negative affect, altered reward processing, and prefrontal disinhibition interact to promote behavioral dysregulation and psychiatric disorders.
Collapse
Affiliation(s)
- Andrew S. Tubbs
- Sleep and Health Research Program, Department of Psychiatry, University of Arizona College of Medicine—Tucson, Tucson, AZ, United States
| | - Fabian-Xosé Fernandez
- Department of Psychology, Evelyn F Mcknight Brain Institute, University of Arizona, Tucson, AZ, United States
| | - Michael A. Grandner
- Sleep and Health Research Program, Department of Psychiatry, University of Arizona College of Medicine—Tucson, Tucson, AZ, United States
| | - Michael L. Perlis
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA, United States
| | - Elizabeth B. Klerman
- Department of Neurology, Division of Sleep Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
41
|
Gururajan A. The impact of chronic stress on the PFC transcriptome: a bioinformatic meta-analysis of publicly available RNA-sequencing datasets. Stress 2022; 25:305-312. [PMID: 35983587 DOI: 10.1080/10253890.2022.2111211] [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] [Indexed: 10/15/2022] Open
Abstract
The prefrontal cortex (PFC) is one of several brain structures that are sensitive to chronic stress exposure. There have been several studies which have examined the effects on chronic stress, using various protocols such as chronic unpredictable stress and chronic social defeat stress, on the PFC transcriptome. In this report, a bioinformatic meta-analysis of publicly available RNA sequencing datasets (101 samples) from seven chronic stress studies was carried out to identify core PFC transcriptional signatures that underpin behavioral phenotypes including resilience and susceptibility. The results showed 160 differentially expressed genes in chronic stress mice compared to controls with significant enrichment in mechanisms associated with translation and localization of membrane-bound proteins with a putative effect on synaptic plasticity in glutamatergic neurons. Moreover, the meta-analysis revealed no differentially expressed genes in resilient mice but 144 in susceptible mice compared to controls, of which 44 were not identified in the individual studies. Enrichment analysis revealed that susceptibility genes were most affected in oligodendrocytes and linked to mechanisms which mediate biochemical, bidirectional communication between this cell-type and myelinated axons. These results provide new avenues for further research into the neurobiology and treatment of chronic stress-induced disorders.
Collapse
Affiliation(s)
- Anand Gururajan
- Brain & Mind Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
42
|
Kira IA, Shuwiekh HA, Alhuwailah A, Balaghi D. Does COVID-19 Type III Continuous Existential Trauma Deplete the Traditional Coping, Diminish Health and Mental Health, and Kindle Spirituality?: An Exploratory Study on Arab Countries. JOURNAL OF LOSS & TRAUMA 2021. [DOI: 10.1080/15325024.2021.2006501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Ibrahim A. Kira
- Center for Cumulative Trauma Studies, Stone Mountain, GA, USA
- Affiliate of Center for Stress, Trauma and Resiliency, Georgia State University, Atlanta, GA, USA
| | | | | | | |
Collapse
|