1
|
Guan L, Qiu M, Li N, Zhou Z, Ye R, Zhong L, Xu Y, Ren J, Liang Y, Shao X, Fang J, Fang J, Du J. Inhibitory gamma-aminobutyric acidergic neurons in the anterior cingulate cortex participate in the comorbidity of pain and emotion. Neural Regen Res 2025; 20:2838-2854. [PMID: 39314159 PMCID: PMC11826466 DOI: 10.4103/nrr.nrr-d-24-00429] [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: 04/17/2024] [Revised: 06/19/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Pain is often comorbid with emotional disorders such as anxiety and depression. Hyperexcitability of the anterior cingulate cortex has been implicated in pain and pain-related negative emotions that arise from impairments in inhibitory gamma-aminobutyric acid neurotransmission. This review primarily aims to outline the main circuitry (including the input and output connectivity) of the anterior cingulate cortex and classification and functions of different gamma-aminobutyric acidergic neurons; it also describes the neurotransmitters/neuromodulators affecting these neurons, their intercommunication with other neurons, and their importance in mental comorbidities associated with chronic pain disorders. Improving understanding on their role in pain-related mental comorbidities may facilitate the development of more effective treatments for these conditions. However, the mechanisms that regulate gamma-aminobutyric acidergic systems remain elusive. It is also unclear as to whether the mechanisms are presynaptic or postsynaptic. Further exploration of the complexities of this system may reveal new pathways for research and drug development.
Collapse
Affiliation(s)
- Lu Guan
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Mengting Qiu
- Fuchun Community Health Service Center of Fuyang District, Hangzhou, Zhejiang Province, China
| | - Na Li
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhengxiang Zhou
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Ru Ye
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Liyan Zhong
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yashuang Xu
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Junhui Ren
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi Liang
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaomei Shao
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jianqiao Fang
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Junfan Fang
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Junying Du
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Yabuki Y, Hori K, Zhang Z, Matsuo K, Kudo K, Usuki S, Gadotti VM, Chen L, Ueno S, Chiba S, Fukunaga K, Zamponi GW, Shioda N. Cav3.1 T-Type Calcium Channel Acts as a Gateway for GABAergic Excitation in the Medial Prefrontal Cortex That Leads to Chronic Psychological Stress Responses in Mice. Acta Physiol (Oxf) 2025; 241:e70043. [PMID: 40197682 DOI: 10.1111/apha.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
AIM The molecular mechanisms of chronic stress-induced psychiatric disorders, including depression, remain unknown. The current study aimed to assess the role of Cav3.1 T-type calcium channels as a gateway for the chronic stress-induced activation of parvalbumin (PV)-positive gamma-aminobutyric acidergic (GABAergic) neurons in the medial prefrontal cortex (mPFC) of mice. METHODS The function of the Cav3.1 T-type calcium channel in the mouse mPFC following chronic stress was investigated using behavioral tests, electrophysiological analyses, transcriptome analyses, and optogenetic approaches. RESULTS Cav3.1-knockout (Cav3.1-/-) mice were resistant to chronic stress-induced depressive-like behaviors induced by repeated forced-swimming test or tail-suspension test. Immunohistochemical analysis revealed that Cav3.1 was predominantly localized in PV-positive GABAergic neurons in the mPFC. Based on transcriptomic and electrophysiological analyses, the excitatory-inhibitory (E-I) balance was disrupted by the chronic stress-induced activation of PV-positive GABAergic neurons in the mPFC of wild-type (WT) mice, but not in that of Cav3.1-/- mice. Optogenetic control of PV-positive GABAergic neurons in the mPFC revealed that they played a pivotal role in depressive-like behaviors. The administration of TTA-A2, a selective T-type calcium channel antagonist, reduced chronic stress-induced depressive-like behaviors. CONCLUSION The Cav3.1 T-type calcium channel acts as a gateway for the activation of GABAergic neurons in the mPFC of mice, thereby eliciting chronic psychobiological stress responses.
Collapse
Affiliation(s)
- Yasushi Yabuki
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Chuo-ku, Kumamoto, Japan
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Karin Hori
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Zizhen Zhang
- Department of Clinical Neurosciences, Cuming School of Medicine, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kazuya Matsuo
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Chuo-ku, Kumamoto, Japan
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Kenta Kudo
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Chuo-ku, Kumamoto, Japan
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Shingo Usuki
- Liaison Laboratory Research Promotion Center, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, Cuming School of Medicine, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, University of Vale do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Lina Chen
- Department of Clinical Neurosciences, Cuming School of Medicine, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Shinya Ueno
- Department of Neurophysiology, Graduate School of Medicine, Hirosaki University, Hirosaki, Japan
| | - Shuji Chiba
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Cuming School of Medicine, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Norifumi Shioda
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Chuo-ku, Kumamoto, Japan
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan
| |
Collapse
|
3
|
Yoshioka T, Yamada D, Hagiwara A, Kajino K, Iio K, Saitoh T, Nagase H, Saitoh A. Delta opioid receptor agonists activate PI3K-mTORC1 signaling in parvalbumin-positive interneurons in mouse infralimbic prefrontal cortex to exert acute antidepressant-lie effects. Mol Psychiatry 2025; 30:2038-2048. [PMID: 39643691 PMCID: PMC12015109 DOI: 10.1038/s41380-024-02814-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 12/09/2024]
Abstract
The delta opioid receptor (DOP) is a promising target for novel antidepressants due to its potential for rapid action with minimal adverse effects; however, the functional mechanism underlying acute antidepressant actions remains elusive. We report that subcutaneous injection of the selective DOP agonist KNT-127 reduced immobility in the forced swimming test, and that this antidepressant-like response was reversed by intracerebroventricular injection of the selective mechanistic (or mammalian) target of rapamycin (mTOR) inhibitor rapamycin or the phosphatidylinositol-3 kinase (PI3K) inhibitor LY294002. KNT-127 also alleviated social avoidance and reduced sucrose consumption (anhedonia) among chronic vicarious social defeat stress model mice, which were similarly reversed by PI3K and mTOR inhibitors. In addition, KNT-127 increased phosphorylation levels of the mTOR signaling-related proteins Akt and p70S6 kinase in medial prefrontal cortex as revealed by immunoblotting. In the forced swimming test, a microinfusion of KNT-127 and another DOP agonist SNC80 in the infralimbic prefrontal cortex (IL-PFC) attenuated the immobility, which were blocked by rapamycin and LY294002. Perfusion of KNT-127 onto IL-PFC slices increased miniature excitatory postsynaptic current frequency and reduced miniature inhibitory postsynaptic current frequency in pyramidal neurons as measured by whole-cell patch-clamping, and both responses were reversed by rapamycin. Imaging of brain slices from transgenic mice with DOP-promoter-driven green fluorescent protein revealed that most DOPs were expressed in parvalbumin-positive interneurons in the IL-PFC. These findings suggest that DOP agonists exert antidepressant-like actions by suppressing GABA release from parvalbumin-positive interneurons via the PI3K-Akt-mTORC1-p70S6 kinase pathway, thereby enhancing IL-PFC pyramidal neuron excitation.
Collapse
Affiliation(s)
- Toshinori Yoshioka
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Daisuke Yamada
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Akari Hagiwara
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Keita Kajino
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Keita Iio
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Akiyoshi Saitoh
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan.
| |
Collapse
|
4
|
Lu G, Huang R, Zeng S, Xing Y, Xie H, Du L. [ 1H- 13C]-NMR-Based Metabolic Kinetics Reveals Brain Neurochemical Alterations in Mice After Retinal Ischemia-Reperfusion Injury. Mol Neurobiol 2025; 62:5758-5773. [PMID: 39621232 DOI: 10.1007/s12035-024-04641-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/22/2024] [Indexed: 03/29/2025]
Abstract
Retinal ischemia-reperfusion injury (RIRI) is a pathological process that occurs in various blinding eye diseases and is often accompanied by anxiety and depression. However, the underlying metabolic mechanism of mood disorders remains unclear. This study aimed to investigate the metabolic dynamics of the brain after RIRI. C57BL/6 J mice were used to establish the RIRI model and assessed after 1 and 7 days. Mood-related behaviors were examined using open-field, elevated plus-maze, and forced swimming tests. Retinal injury histology was assessed using retinal hematoxylin and eosin staining. Retinal apoptosis was measured via the TdT-mediated dUTP nick-end labeling staining. The 13C-labeled metabolite information for six brain regions of interest was obtained using the [1H-13C]-NMR technique. Retinal tissue damage and cell apoptosis in the retina were observed 1 and 7 days after RIRI. One day after RIRI, mice displayed anxiety- and depression-like behaviors, and multiple metabolites involved in the glutamine (Gln)/glutamate (Glu)-γ-aminobutyric acid (GABA) and tricarboxylic acid (TCA) cycles exhibited reductions in all studied brain regions, with frontal cortex (FC) and temporal cortex (TC) being the most markedly altered. Metabolites and behavioral indicators nearly returned to normal after 7 days. Significant positive correlations between Gln/Glu-GABA and TCA cycle metabolites were observed in the RIRI brain. The results revealed that within a short period after RIRI, there was a reduction in brain metabolites and a disruption of the Gln/Glu-GABA and TCA cycles, which may contribute to mood disorders in mice.
Collapse
Affiliation(s)
- Guojing Lu
- Eye Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Rong Huang
- Eye Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Siyu Zeng
- Eye Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Yiqiao Xing
- Eye Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Hang Xie
- Eye Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Lei Du
- Eye Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
5
|
Lesch KP, Gorbunov N. Antisocial personality disorder:Failure to balance excitation/inhibition? Neuropharmacology 2025; 268:110321. [PMID: 39855295 DOI: 10.1016/j.neuropharm.2025.110321] [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: 08/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While healthy brain function relies on a dynamic but tightly regulated interaction between excitation (E) and inhibition (I), a spectrum of social cognition disorders, including antisocial behavior and antisocial personality disorder (ASPD), frequently ensuing from irregular neurodevelopment, may be associated with E/I imbalance and concomitant alterations in neural connectivity. Technological advances in the evaluation of structural and functional E/I balance proxies in clinical settings and in human cell culture models provide a general basis for identification of biomarkers providing a powerful concept for prevention and intervention across different dimensions of mental health and disease. In this perspective we outline a framework for research to characterize neurodevelopmental pathways to antisocial behavior and ASPD driven by (epi)genetic factors across life, and to identify molecular targets for preventing the detrimental effects of cognitive dysfunction and maladaptive social behavior, considering psychosocial experience; to validate signatures of E/I imbalance and altered myelination proxies as biomarkers of pathogenic neural circuitry mechanisms to determine etiological processes in the transition from mental health to antisocial behavior and ASPD and in the switch from prevention to treatment; to develop a neurobiologically-grounded integrative model of antisocial behavior and ASPD resultant of disrupted E/I balance, allowing to establish objective diagnoses and monitoring tools, to personalize prevention and therapeutic decisions, to predict treatment response, and thus counteract relapse; and finally, to promote transformation of dimensional disorder taxonomy and to enhance societal awareness and reception of the neurobiological basis of antisocial behavior and ASPD.
Collapse
Affiliation(s)
- Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Child- and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
6
|
Reneaux M, Mayberg H, Friston K, Pinotsis DA. A computational account of joint SSRI and anti-inflammatory treatment. Front Immunol 2025; 16:1472732. [PMID: 40352929 PMCID: PMC12061865 DOI: 10.3389/fimmu.2025.1472732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Depression is a chronic disorder that impacts millions worldwide. Traditional treatments may not always work. Inflammation seems to be an underlying cause for chronicity and treatment non-response. Methods We present a computational model that elucidates the interplay between inflammation, serotonin levels, and brain activity. Results The model delineates how inflammation impacts extracellular serotonin, while cerebral activity reciprocally influences serotonin concentration. Understanding the reciprocal interplay between the immune system and brain dynamics is important, as unabated inflammation can lead to relapsing depression. The model predicts dynamics within the prefrontal cortex (PFC) and subcallosal cingulate cortex (SCC), mirroring patterns observed in depressive conditions. It also accommodates pharmaceutical interventions that encompass anti-inflammatory and antidepressant agents, concurrently evaluating their efficacy with regard to the severity of depressive symptoms Our model shows that for mild and moderate levels of depression anti-depressant agents or anti-inflammatory agents acting in isolation can bring serotonergic levels and brain activity to control levels. However, for severe depression only joint treatment of anti-depressant and anti-inflammatory agents can bring the serotonergic levels and activity to control levels. Discussion This study is a first step to mechanistically understand the intricate link between the immune system and depression, the role of inflammation and potential treatments. It explores the impact of anti-depressant and anti-inflammatory drug treatments and assesses their relevance with regard to depression severity.
Collapse
Affiliation(s)
- Melissa Reneaux
- Centre for Mathematical Neuroscience and Psychology and Department of Psychology, City St. George’s —University of London, London, United Kingdom
- Psychology and Behavior Program, School of Liberal Studies and Media, UPES, Dehradun, India
| | - Helen Mayberg
- Department of Neurology and Neurosurgery, Icahn School of Medicine at Mt. Sinai, New York, NY, United States
| | - Karl Friston
- Wellcome Centre for Human Neuroimaging, University College London (UCL), London, United Kingdom
| | - Dimitris A. Pinotsis
- Centre for Mathematical Neuroscience and Psychology and Department of Psychology, City St. George’s —University of London, London, United Kingdom
- The Picower Institute for Learning & Memory and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
7
|
Zhang J, Duan J, Li W, Wang X, Ren S, Ye L, Liu F, Tian X, Xie Y, Huang Y, Sun Y, Song N, Li T, Cai X, Liu Z, Zhou H, Huang C, Li Y, Zhu S, Guo F. An antidepressant mechanism underlying the allosteric inhibition of GluN2D-incorporated NMDA receptors at GABAergic interneurons. SCIENCE ADVANCES 2025; 11:eadq0444. [PMID: 40043126 PMCID: PMC11881904 DOI: 10.1126/sciadv.adq0444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025]
Abstract
N-methyl-d-aspartate receptors (NMDARs), key excitatory ion channels, have gained attention as anti-depression targets. NMDARs consist of two GluN1 and two GluN2 subunits (2A-2D), which determine their pharmacological properties. Few compounds selectively targeting GluN2 subunits with antidepressant effects have been identified. Here, we present YY-23, a compound that selectively inhibits GluN2C- or GluN2D-containing NMDARs. Cryo-EM analysis revealed that YY-23 binds to the transmembrane domain of the GluN2D subunit. YY-23 primarily affects GluN2D-containing NMDARs on GABAergic interneurons in the prefrontal cortex, suppressing GABAergic neurotransmission and enhancing excitatory transmission. Behavioral assays demonstrate YY-23's rapid antidepressant effects in both stress-naïve and stress-exposed models, which are lost in mice with global or selective knockout of the grin2d gene in parvalbumin-positive interneurons. These findings highlight GluN2D-containing NMDARs on GABAergic interneurons as potential depression treatment targets.
Collapse
Affiliation(s)
- Jilin Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jinjin Duan
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Wei Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Xian Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shimin Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Luyu Ye
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fang Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoting Tian
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Xie
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yiming Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Song
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianyu Li
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiang Cai
- Oujiang Laboratory, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhiqiang Liu
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| | - Hu Zhou
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| | - Chenggang Huang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Li
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Fei Guo
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| |
Collapse
|
8
|
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 2025; 30:1034-1046. [PMID: 39237722 PMCID: PMC11835755 DOI: 10.1038/s41380-024-02715-1] [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: 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
|
9
|
Santander O, Arredondo SB, García-Rojas F, Estay SF, Belforte JE, Chávez AE, Varela-Nallar L, Fuenzalida M. Ketamine administration during adolescence impairs synaptic integration and inhibitory synaptic transmission in the adult dentate gyrus. Prog Neurobiol 2025; 246:102718. [PMID: 39855537 DOI: 10.1016/j.pneurobio.2025.102718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/10/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Ketamine administration during adolescence affects cognitive performance; however, its long-term impact on synaptic function and neuronal integration in the hippocampus a brain region critical for cognition remains unclear. Using functional and molecular analyses, we found that chronic ketamine administration during adolescence exerts long-term effects on synaptic integration, expanding the temporal window in an input-specific manner affecting the inner molecular layer but not the medial perforant path inputs in the adult mouse dorsal hippocampal dentate gyrus. Ketamine also alters the excitatory/inhibitory balance by reducing the efficacy of inhibitory inputs likely due to a reduction in parvalbumin-positive interneurons number and function. These findings indicate that during adolescence, ketamine exerts a strong effect on inhibitory synaptic function mediated by parvalbumin-positive neurons that ultimately impact synaptic integration in the dorsal adult dentate gyrus, which could help to understand the neurobiological and functional bases that confer greater vulnerability to the adolescent brain.
Collapse
Affiliation(s)
- Odra Santander
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile; Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Sebastián B Arredondo
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile
| | - Francisca García-Rojas
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Sebastián F Estay
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile; Instituto de Neurociencias y Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Juan E Belforte
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Andrés E Chávez
- Instituto de Neurociencias y Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Lorena Varela-Nallar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
| |
Collapse
|
10
|
Wang B, He T, Qiu G, Li C, Xue S, Zheng Y, Wang T, Xia Y, Yao L, Yan J, Chen Y. Altered synaptic homeostasis: a key factor in the pathophysiology of depression. Cell Biosci 2025; 15:29. [PMID: 40001206 PMCID: PMC11863845 DOI: 10.1186/s13578-025-01369-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Depression, a widespread psychiatric disorder, is characterized by a diverse array of symptoms such as melancholic mood and anhedonia, imposing a significant burden on both society and individuals. Despite extensive research into the neurobiological foundations of depression, a complete understanding of its complex mechanisms is yet to be attained, and targeted therapeutic interventions remain under development. Synaptic homeostasis, a compensatory feedback mechanism, involves neurons adjusting synaptic strength by regulating pre- or postsynaptic processes. Recent advancements in depression research reveal a crucial association between the disorder and disruptions in synaptic homeostasis within neural regions and circuits pivotal for emotional and cognitive functions. This paper explores the mechanisms governing synaptic homeostasis in depression, focusing on the role of ion channels, the regulation of presynaptic neurotransmitter release, synaptic scaling processes, and essential signaling molecules. By mapping new pathways in the study of synaptic homeostasis as it pertains to depression, this research aims to provide valuable insights for identifying novel therapeutic targets for more effective antidepressant treatments.
Collapse
Affiliation(s)
- Bokai Wang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Teng He
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Guofan Qiu
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chong Li
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Song Xue
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanjia Zheng
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Taiyi Wang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yucen Xia
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lin Yao
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
11
|
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 2025; 30:423-434. [PMID: 39095477 DOI: 10.1038/s41380-024-02675-6] [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: 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
|
12
|
Bisharat G, Kaganovski E, Sapir H, Temnogorod A, Levy T, Resnik J. Repeated stress gradually impairs auditory processing and perception. PLoS Biol 2025; 23:e3003012. [PMID: 39932893 PMCID: PMC11813133 DOI: 10.1371/journal.pbio.3003012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
Repetitive stress, a common feature of modern life, is a major risk factor for psychiatric and sensory disorders. Despite the prevalence of perceptual abnormalities in these disorders, little is known about how repetitive stress affects sensory processing and perception. Here, we combine repetitive stress in mice, longitudinal measurement of cortical activity, and auditory-guided behaviors to test if sound processing and perception of neutral sounds in adults are modulated by repetitive stress. We found that repetitive stress alters sound processing, increasing spontaneous cortical activity while dampening sound-evoked responses in pyramidal and PV cells and heightening sound-evoked responses in SST cells. These alterations in auditory processing culminated in perceptual shifts, particularly a reduction in loudness perception. Additionally, our work reveals that the impact of stress on perception evolves gradually as the stressor persists over time, emphasizing the dynamic and evolving nature of this mechanism. Our findings provide insight into a possible mechanism by which repetitive stress alters sensory processing and behavior, challenging the idea that stress primarily modulates emotionally charged stimuli.
Collapse
Affiliation(s)
- Ghattas Bisharat
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ekaterina Kaganovski
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Hila Sapir
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anita Temnogorod
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tal Levy
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jennifer Resnik
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
13
|
Hepsomali P, Costabile A, Schoemaker M, Imakulata F, Allen P. Adherence to unhealthy diets is associated with altered frontal gamma-aminobutyric acid and glutamate concentrations and grey matter volume: preliminary findings. Nutr Neurosci 2025; 28:125-137. [PMID: 38794782 DOI: 10.1080/1028415x.2024.2355603] [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] [Indexed: 05/26/2024]
Abstract
OBJECTIVES Common mental disorders (CMD) are associated with impaired frontal excitatory/inhibitory (E/I) balance and reduced grey matter volume (GMV). Larger GMV (in the areas that are implicated in CMD-pathology) and improved CMD-symptomatology have been observed in individuals who adhere to high quality diets. Moreover, preclinical studies have shown altered neurometabolites (primarily gamma-aminobutyric acid: GABA and glutamate: GLU) in relation to diet quality. However, neurochemical correlates of diet quality and how these neurobiological changes are associated with CMD and with its transdiagnostic factor, rumination, is unknown in humans. Therefore, in this study, we examined the associations between diet quality and frontal cortex neuro-chemistry and structure, as well as CMD and rumination in humans. METHODS Thirty adults were classified into high and low diet quality groups and underwent 1H-MRS to measure medial prefrontal cortex (mPFC) metabolite concentrations and volumetric imaging to measure GMV. RESULTS Low (vs High) diet quality group had reduced mPFC-GABA and elevated mPFC-GLU concentrations, as well as reduced right precentral gyrus (rPCG) GMV. However, CMD and rumination were not associated with diet quality. Notably, we observed a significant negative correlation between rumination and rPCG-GMV and a marginally significant association between rumination and mPFC-GLU concentrations. There was also a marginally significant association between mPFC-GLU concentrations and rPCG-GMV. DISCUSSION Adhering to unhealthy dietary patterns may be associated with compromised E/I balance, and this could affect GMV, and subsequently, rumination.
Collapse
Affiliation(s)
- Piril Hepsomali
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London, UK
| | | | | | - Paul Allen
- Department of Neuroimaging, Kings College London, Institute of Psychology and Neuroscience, London, UK
| |
Collapse
|
14
|
Orso R, Creutzberg KC, Begni V, Petrillo G, Cattaneo A, Riva MA. Emotional dysregulation following prenatal stress is associated with altered prefrontal cortex responsiveness to an acute challenge in adolescence. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111162. [PMID: 39383932 DOI: 10.1016/j.pnpbp.2024.111162] [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: 07/04/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Exposure to prenatal stress (PNS) has the potential to elicit multiple neurobiological alterations and increase the susceptibility to psychiatric disorders. Moreover, gestational stress may sensitize the brain toward an altered response to subsequent challenges. Here, we investigated the effects of PNS in rats and assessed whether these animals exhibit an altered brain responsiveness to an acute stress (AS) during adolescence. From gestational day 14 until delivery, Sprague Dawley dams were exposed to PNS or left undisturbed. During adolescence (PND38 to PND41), offspring were tested in the social interaction and splash test. At PND44 half of the animals were exposed to 5 min of forced swim stress. Males and Females exposed to PNS showed reduced sociability and increased anhedonic-like behavior. At the molecular level, exposure of adolescent rats to AS produced increased activation of the amygdala and ventral and dorsal hippocampus. Regarding the prefrontal cortex (PFC), we observed a pronounced activation in PNS males exposed to AS. Cell-type specific transcriptional analyses revealed a significant imbalance in the activation of PFC excitatory and inhibitory neurons in PNS males and females exposed to AS. Furthermore, stressed males exhibited disrupted HPA-axis function, while females showed impairments in the modulation of antioxidant genes. Our study shows that PNS induces emotional dysregulation and alters the responsiveness of the PFC to an acute stressor. Moreover, the disruption of excitatory and inhibitory balance during adolescence could influence the ability to respond to challenging events that may contribute to precipitate a full-blown pathologic condition.
Collapse
Affiliation(s)
- Rodrigo Orso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | | | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Giulia Petrillo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
15
|
Horie I, Muroi Y, Ishii T. Noradrenergic Regulation of the Medial Prefrontal Cortex Mediates Stress Coping in Postpartum Female Mice. Mol Neurobiol 2025; 62:137-155. [PMID: 38829510 DOI: 10.1007/s12035-024-04240-2] [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: 08/29/2023] [Accepted: 05/10/2024] [Indexed: 06/05/2024]
Abstract
The prevalence of depression in women increases during the postpartum period. We previously reported that subchronic exposure to social stress decreased passive coping in postpartum female mice. This study aimed to investigate whether noradrenaline regulation might regulate coping styles in mice. We first determined whether a different type of stress, subchronic physical stress, decreases passive coping in postpartum females. Postpartum female, virgin female, and male mice were exposed to subchronic restraint stress (restraint stress for 4 h for 5 consecutive days). Subchronic restraint stress decreased passive coping in postpartum females but not in virgin females and males in the forced swim and tail suspension tests. We next examined the neuronal mechanism by which subchronic stress decreases passive coping in postpartum female mice. Neuronal activity and expression of noradrenergic receptors in the medial prefrontal cortex (mPFC) were analyzed using immunohistochemistry and reverse transcription-quantitative polymerase chain reaction, respectively. The mPFC was manipulated using chemogenetics, knockdown, or an α2A adrenergic receptor (AR) antagonist. Immunohistochemistry revealed that subchronic restraint stress increased glutamatergic neuron activation in the mPFC via forced swim stress and decreased α2A AR expression in postpartum females. Chemogenetic activation of glutamatergic neurons in the mPFC, knockdown of α2AAR in the mPFC, and the α2A AR receptor antagonist atipamezole treatment decreased passive coping in postpartum females. Subchronic restraint stress decreased passive coping in postpartum females by increasing glutamatergic neuron activity in the mPFC through α2A AR attenuation. The noradrenergic regulation of the mPFC may be a new target for treating postpartum depression.
Collapse
Affiliation(s)
- Ikuko Horie
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, National University Cooperation Hokkaido Higher Education and Research, Hokkaido, 080-8555, Japan
| | - Yoshikage Muroi
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, National University Cooperation Hokkaido Higher Education and Research, Hokkaido, 080-8555, Japan.
| | - Toshiaki Ishii
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, National University Cooperation Hokkaido Higher Education and Research, Hokkaido, 080-8555, Japan
| |
Collapse
|
16
|
Sacks DD, Levin AR, Nelson CA, Enlow MB. Associations Among EEG Aperiodic Slope, Infant Temperament, and Maternal Anxiety/Depression Symptoms in Infancy. Psychophysiology 2025; 62:e14757. [PMID: 39760248 PMCID: PMC11789922 DOI: 10.1111/psyp.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
The aperiodic "slope" of the EEG power spectrum (i.e., aperiodic exponent, commonly represented as a slope in log-log space) is hypothesized to index the cortical excitatory-inhibitory balance. Slope has been associated with various neurodevelopmental outcomes in older children and adults, as well as with family history of ADHD in infants. Here, we investigate associations among EEG aperiodic slope, temperament, and maternal internalizing (anxiety and depression) symptoms in a large cohort of typically developing infants. A steeper slope was associated with higher scores on the temperament domains of orienting/regulation and surgency but was not associated with negative affectivity. Maternal symptoms did not appear to be directly associated with the slope, but the slope moderated the association between maternal symptoms and temperament. Specifically, a steeper slope was associated with a stronger negative association between maternal internalizing symptoms and infant orienting/regulation. These results demonstrate associations between slope and behavior as early as infancy, which may reflect early differences in the development of global inhibitory networks. Longitudinal research in early childhood is necessary to better understand the nature of these relations during development and their potential impact on later socioemotional outcomes.
Collapse
Affiliation(s)
- Dashiell D. Sacks
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - April R. Levin
- Department of Neurology, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Charles A. Nelson
- Division of Developmental Medicine, Boston Children’s Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Harvard Graduate School of Education, Cambridge, MA
| | - Michelle Bosquet Enlow
- Department of Psychiatry and Behavioral Sciences, Boston Children’s Hospital, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| |
Collapse
|
17
|
Nawreen N, Oshima K, Chambers J, Smail M, Herman JP. Inhibition of prefrontal cortex parvalbumin interneurons mitigates behavioral and physiological sequelae of chronic stress in male mice. Stress 2024; 27:2361238. [PMID: 38962839 PMCID: PMC11725266 DOI: 10.1080/10253890.2024.2361238] [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: 05/26/2023] [Accepted: 05/22/2024] [Indexed: 07/05/2024] Open
Abstract
Chronic stress leads to hypofunction of the medial prefrontal cortex (mPFC), mechanisms of which remain to be determined. Enhanced activation of GABAergic of parvalbumin (PV) expressing interneurons (INs) is thought to play a role in stress-induced prefrontal inhibition. In this study, we tested whether chemogenetic inhibition of mPFC PV INs after chronic stress can rescue chronic stress-related behavioral and physiological phenotypes. Mice underwent 2 weeks of chronic variable stress (CVS) followed by a battery of behavioral tests known to be affected by chronic stress exposure, e.g. an open field (OF), novel object recognition (NOR), tail suspension test (TST), sucrose preference test (SPT), and light dark (LD) box. Inhibitory DREADDs were actuated by 3 mg/kg CNO administered 30 min prior to each behavioral test. CVS caused hyperactivity in the OF, reduced sucrose preference in the SPT (indicative of enhanced anhedonia), and increased anxiety-like behavior in the LD box. Inhibition of PV IN after stress mitigated these effects. In addition, CVS also resulted in reduced thymus weight and body weight loss, which were also mitigated by PV IN inhibition. Our results indicate that chronic stress leads to plastic changes in PV INs that may be mitigated by chemogenetic inhibition. Our findings implicate cortical GABAergic INs as a therapeutic target in stress-related diseases.
Collapse
Affiliation(s)
- Nawshaba Nawreen
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- Veterans Affairs Medical Center, Cincinnati, Ohio 45221, United States
| | - Kristen Oshima
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- College of Allied Health Sciences, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
| | - James Chambers
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
| | - Marissa Smail
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- Veterans Affairs Medical Center, Cincinnati, Ohio 45221, United States
| | - James P. Herman
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio 45237-0506, United States
- Veterans Affairs Medical Center, Cincinnati, Ohio 45221, United States
- Dept. of Neurology, University of Cincinnati, Cincinnati, Ohio 45237, United States
| |
Collapse
|
18
|
Page CE, Epperson CN, Novick AM, Duffy KA, Thompson SM. Beyond the serotonin deficit hypothesis: communicating a neuroplasticity framework of major depressive disorder. Mol Psychiatry 2024; 29:3802-3813. [PMID: 38816586 PMCID: PMC11692567 DOI: 10.1038/s41380-024-02625-2] [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: 11/17/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The serotonin deficit hypothesis explanation for major depressive disorder (MDD) has persisted among clinicians and the general public alike despite insufficient supporting evidence. To combat rising mental health crises and eroding public trust in science and medicine, researchers and clinicians must be able to communicate to patients and the public an updated framework of MDD: one that is (1) accessible to a general audience, (2) accurately integrates current evidence about the efficacy of conventional serotonergic antidepressants with broader and deeper understandings of pathophysiology and treatment, and (3) capable of accommodating new evidence. In this article, we summarize a framework for the pathophysiology and treatment of MDD that is informed by clinical and preclinical research in psychiatry and neuroscience. First, we discuss how MDD can be understood as inflexibility in cognitive and emotional brain circuits that involves a persistent negativity bias. Second, we discuss how effective treatments for MDD enhance mechanisms of neuroplasticity-including via serotonergic interventions-to restore synaptic, network, and behavioral function in ways that facilitate adaptive cognitive and emotional processing. These treatments include typical monoaminergic antidepressants, novel antidepressants like ketamine and psychedelics, and psychotherapy and neuromodulation techniques. At the end of the article, we discuss this framework from the perspective of effective science communication and provide useful language and metaphors for researchers, clinicians, and other professionals discussing MDD with a general or patient audience.
Collapse
Affiliation(s)
- Chloe E Page
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Family Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Helen and Arthur E. Johnson Depression Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew M Novick
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Korrina A Duffy
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Scott M Thompson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
19
|
Faulkner P, Allen P, Costabile A, Schoemaker MH, Imakulata F, Hepsomali P. Greater resting state functional connectivity of the medial prefrontal cortex with the thalamus, caudate, and putamen in individuals who adhere to the Mediterranean style diets. Eur J Nutr 2024; 64:34. [PMID: 39607478 PMCID: PMC11604750 DOI: 10.1007/s00394-024-03548-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024]
Abstract
PURPOSE Healthy diets are believed to be associated with a reduced risk of experiencing common mental disorders (CMDs) and related symptomatology (such as ruminative thinking), and with healthier brain chemistry and structure, especially in the frontal regions implicated in CMDs, cognitive control, and food choice. Nevertheless, there is very limited research on the relationship between diet health/quality and brain function. In this study we assessed the associations between adherence to the Mediterranean diet and resting state functional connectivity (rs-FC) of the prefrontal cortex (PFC) with the whole brain and whether this connectivity would be associated with ruminative thinking as a transdiagnostic factor for CMDs. METHODS Thirty-seven adults (Mean Age = 25.57, SD = 7.18) completed the Mediterranean Diet Adherence Screener (MEDAS) and were classified into high- and low-quality diet groups and completed the Ruminative Response Scale. All participants underwent resting-state functional MRI (fMRI) to determine whole-brain rs-FC of the medial prefrontal cortex (mPFC). RESULTS Participants in the high MEDAS group (vs. low MEDAS group) exhibited significantly greater rs-FC of the mPFC seed with the thalamus, caudate and putamen. Additionally, the strength of rs-FC of the mPFC seed with these regions was positively associated with the MEDAS scores across groups in both crude and adjusted models. There were no significant associations between the strength of rs-FC of the mPFC seed with the cluster of voxels with the thalamus, caudate, and putamen and ruminative thinking. DISCUSSION This work shows that healthy dietary patterns are associated with rs-FC in the frontal-subcortical circuitry in healthy volunteers. Considering the implications of the dysregulation of this circuity, adhering to healthy dietary patterns may offer a promising alternative/complementary method to improve CMDs, cognitive control, and food choices.
Collapse
Affiliation(s)
- Paul Faulkner
- School of Psychology, University of Roehampton, London, SW155 4JD, UK
| | - Paul Allen
- Department of Neuroimaging, Institute of Psychology and Neuroscience, Kings College London, London, UK
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London, SW155 4JD, UK
| | | | | | - Piril Hepsomali
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, RG6 6ET, UK.
| |
Collapse
|
20
|
Sagi R, Chakraborty M, Bogdanovic M, Asraf H, Sekler I, Kofman O, Cohen H, Hershfinkel M. Loss of the zinc receptor ZnR/GPR39 in mice enhances anxiety-related behavior and motor deficits, and modulates KCC2 expression in the amygdala. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:31. [PMID: 39581978 PMCID: PMC11587656 DOI: 10.1186/s12993-024-00254-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Mood disorders, particularly depression and anxiety, are associated with zinc dyshomeostasis and aberrant GABAergic signaling. Activation of ZnR/GPR39 by synaptic zinc in the hippocampus triggers phosphorylation of extracellular regulated kinase (ERK1/2), which regulates the K+/Cl- cotransporter (KCC2) and thereby GABAergic inhibitory neurotransmission and seizure activity. Therefore, we studied whether impaired ZnR/GPR39 signaling is linked to anxiety-related behavior in male or female mice. RESULTS Using the acoustic startle response, elevated plus maze, and open field test, we found increased anxiety-related behavior in ZnR/GPR39 knockout (KO) mice. Despite a well-established sex difference, where females are typically more prone to anxiety, both male and female ZnR/GPR39 KO mice exhibited increased anxiety-related behavior compared to wildtype (WT) mice. Additionally, ZnR/GPR39 KO mice displayed impaired motor coordination in the pole and rotarod tests but did not show reduced muscle strength, as indicated by a grip test. Finally, we found intrinsic alterations in the expression level of KCC2, a major Cl- transporter regulating GABAergic signaling, in the amygdala of naïve ZnR/GPR39 KO mice compared to controls. CONCLUSIONS Our findings indicate that loss of ZnR/GPR39 enhances anxiety-related behavior in both male and female mice. Moreover, ZnR/GPR39 KO mice exhibit impaired motor coordination, which may be associated with increased anxiety. Finally, we demonstrate that loss of ZnR/GPR39 modulates the expression of KCC2 in the amygdala. Thus, we propose that ZnR/GPR39 can serve as a target for regulating GABAergic signaling in anxiety treatment.
Collapse
Affiliation(s)
- Romi Sagi
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel.
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.
| | - Moumita Chakraborty
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
- Translational Hepatology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Milos Bogdanovic
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Hila Asraf
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Ora Kofman
- Psychology Department, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Hagit Cohen
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, POB 653, Beer-Sheva, 84105, Israel
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel.
| |
Collapse
|
21
|
Ma YN, Yang CJ, Zhang CC, Sun YX, Yao XD, Liu X, Li XX, Wang HL, Wang H, Wang T, Wang XD, Zhang C, Su YA, Li JT, Si TM. Prefrontal parvalbumin interneurons mediate CRHR1-dependent early-life stress-induced cognitive deficits in adolescent male mice. Mol Psychiatry 2024:10.1038/s41380-024-02845-6. [PMID: 39578519 DOI: 10.1038/s41380-024-02845-6] [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: 11/07/2023] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
Cognitive impairment, a core symptom of psychiatric disorders, is frequently observed in adolescents exposed to early-life stress (ES). However, the underlying neural mechanisms are unclear, and therapeutic efficacy is limited. Targeting parvalbumin-expressing interneurons (PVIs) in the medial prefrontal cortex (mPFC), we report that ES reduces mPFC PVI activity, which causally mediated ES-induced cognitive deficits in adolescent male mice through chemogenetic and optogenetic experiments. To understand the possible causes of PVI activity reduction following ES, we then demonstrated that ES upregulated corticotropin-releasing hormone (CRH) receptor 1 [CRHR1, mainly expressed in pyramidal neurons (PNs)] and reduced activity of local pyramidal neurons (PNs) and their excitatory inputs to PVIs. The subsequent genetic manipulation experiments (CRHR1 knockout, CRH overexpression, and chemogenetics) highlight that ES-induced PVI activity reduction may result from CRHR1 upregulation and PN activity downregulation and that PVIs play indispensable roles in CRHR1- or PN-mediated cognitive deficits induced by ES. These results suggest that ES-induced cognitive deficits could be attributed to the prefrontal CRHR1-PN-PVI pathway. Finally, treatment with antalarmin (a CRHR1 antagonist) and environmental enrichment successfully restored the PVI activity and cognitive deficits induced by ES. These findings reveal the neurobiological mechanisms underlying ES-induced cognitive deficits in adolescent male mice and highlight the therapeutic potentials of PVIs in stress-related cognitive deficits in adolescent individuals.
Collapse
Affiliation(s)
- Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Chao-Juan Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xing-Duo Yao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Hong-Li Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| |
Collapse
|
22
|
Zhou L, Zhang C, Xie Z, Yu Q, Wang J, Gong Y, Zhao J, Bai S, Yang L, Deng D, Zhang R, Shi Y. Neural Circuit Mechanisms of Sinisan formula for the Treatment of adolescent Depression: prefrontal cortex to dorsal raphe nucleus. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118529. [PMID: 38972528 DOI: 10.1016/j.jep.2024.118529] [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: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sinisan formula (SNSF), documented in the classic books Shanghan Lun, is known for its ability to regulate liver-qi and treat depression. However, its underlying mechanism, particularly its effects on dynamic real-time neuron activity and circuits remains to be fully elucidated. AIM OF THE STUDY This study aimed to investigate the antidepressant effect of SNSF and its central nervous system mechanism on depression-like behaviors, focusing on the prefrontal cortex (PFC) to dorsal raphe nucleus (DRN) neural circuit in a stress-induced adolescent animal model. MATERIALS AND METHODS SNSF comprised four herbs, the root of Bupleurum chinense DC., the root of Paeonia lactiflora Pall., the fruit of Citrus aurantium L., the rhizome of Glycyrrhiza uralensis Fisch., in equal propotions. The adolescent depression animal model was induced by maternal separation (MS) and chronic restraint stress (CRS). In-vivo multichannel physiological electrodes were implanted into the PFC on PND 28 and animals were recorded 5 times during PND 35-46. From PND 47, the behavioral tests were performed to evaluate the antidepressant efficacy of SNSF. Subsequently, brain tissue was collected for Western blot and immunofluorescence staining analysis. Retro virus was injected into the DRN to explore sources of projections received by serotonergic (5-HTergic) neurons. And the PFC-to-DRN circuit was activated or inhibited through chemogenetic techniques to investigate the effects of SNSF on depression-like behaviors. RESULTS Administration of SNSF for 18 days effectively alleviated depression-like behaviors in MS&CRS adolescent mice. The PFC emerged as the primary glutamatergic projection source of the DRN5-HT neurons. Following SNSF administration for 13/15/18 days, there was an increase in the firing rate of excitatory neurons and excitatory/inhibitory (E/I) ratio in the PFC. MS&CRS stress let to a reduction in the density of 5-HT+ and CaMKII + neurons in the DRN, accompanied by an increase in the density of GAD + neurons in the DRN, while SNSF administration reversed the alterations. Chemogenetic activation of the PFC-to-DRN circuit rescued the depression-like behaviors induced by MS&CRS, whereas suppression of this circuit attenuated the antidepressant effect of SNSF. CONCLUSIONS SNSF significantly mitigated depression-like behaviors in MS&CRS mice. SNSF exerts its antidepressant effects by increasing the E/I ratio in the PFC and enhancing glutamatergic projections from the PFC to the DRN.
Collapse
Affiliation(s)
- Liuchang Zhou
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Caixia Zhang
- Outpatient Department, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zedan Xie
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qingying Yu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Junjie Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuwen Gong
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jinlan Zhao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shasha Bai
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lei Yang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Di Deng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Rong Zhang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
23
|
Shoji H, Maeda Y, Miyakawa T. Chronic corticosterone exposure causes anxiety- and depression-related behaviors with altered gut microbial and brain metabolomic profiles in adult male C57BL/6J mice. Mol Brain 2024; 17:79. [PMID: 39511657 PMCID: PMC11545877 DOI: 10.1186/s13041-024-01146-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
Chronic exposure to glucocorticoids in response to long-term stress is thought to be a risk factor for major depression. Depression is associated with disturbances in the gut microbiota composition and peripheral and central energy metabolism. However, the relationship between chronic glucocorticoid exposure, the gut microbiota, and brain metabolism remains largely unknown. In this study, we first investigated the effects of chronic corticosterone exposure on various domains of behavior in adult male C57BL/6J mice treated with the glucocorticoid corticosterone to evaluate them as an animal model of depression. We then examined the gut microbial composition and brain and plasma metabolome in corticosterone-treated mice. Chronic corticosterone treatment resulted in reduced locomotor activity, increased anxiety-like and depression-related behaviors, decreased rotarod latency, reduced acoustic startle response, decreased social behavior, working memory deficits, impaired contextual fear memory, and enhanced cued fear memory. Chronic corticosterone treatment also altered the composition of gut microbiota, which has been reported to be associated with depression, such as increased abundance of Bifidobacterium, Turicibacter, and Corynebacterium and decreased abundance of Barnesiella. Metabolomic data revealed that long-term exposure to corticosterone led to a decrease in brain neurotransmitter metabolites, such as serotonin, 5-hydroxyindoleacetic acid, acetylcholine, and gamma-aminobutyric acid, as well as changes in betaine and methionine metabolism, as indicated by decreased levels of adenosine, dimethylglycine, choline, and methionine in the brain. These results indicate that mice treated with corticosterone have good face and construct validity as an animal model for studying anxiety and depression with altered gut microbial composition and brain metabolism, offering new insights into the neurobiological basis of depression arising from gut-brain axis dysfunction caused by prolonged exposure to excessive glucocorticoids.
Collapse
Affiliation(s)
- Hirotaka Shoji
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Yasuhiro Maeda
- Open Facility Center, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
24
|
Chu LH, Chau CQ, Kamel N, Thanh HHT, Yahya N. Functional excitation-inhibition ratio for social anxiety analysis and severity assessment. Front Psychiatry 2024; 15:1461290. [PMID: 39502299 PMCID: PMC11536370 DOI: 10.3389/fpsyt.2024.1461290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Social anxiety disorder (SAD) is a prevalent psychiatric condition characterized by an intense fear of and avoidance of social situations. Traditional assessment methods for SAD primarily rely on subjective self-report questionnaires and clinical interviews, which can be prone to biases and inaccuracies. This study aims to explore the functional excitation-inhibition (fEI) ratio derived from EEG data as a potential objective biomarker for assessing SAD severity. Methods Resting-state EEG data were collected from 20 control subjects and 60 individuals with varying degrees of SAD severity (mild, moderate, and severe). The fEI ratio was estimated across different EEG bands and analyzed, focusing on differences between control subjects and SAD groups. Results Significantly higher fEI ratios were observed in the alpha and low beta EEG bands in individuals with SAD compared to controls, especially within the prefrontal cortex. Additionally, a positive correlation was found between the fEI ratio and the severity of social anxiety symptoms across SAD severity levels. Discussion The findings indicate that the fEI ratio in the alpha and low beta bands may serve as a promising biomarker for assessing SAD severity. These results contribute to a deeper understanding of the neural mechanisms underlying social anxiety, offering a potentially more objective approach to SAD assessment compared to traditional methods.
Collapse
Affiliation(s)
- Linh Ha Chu
- College of Engineering and Computer Science, Center of Environmental Intelligence (CEI), Vin University, Hanoi, Vietnam
| | - Chi Que Chau
- College of Engineering and Computer Science, Center of Environmental Intelligence (CEI), Vin University, Hanoi, Vietnam
| | - Nidal Kamel
- College of Engineering and Computer Science, Center of Environmental Intelligence (CEI), Vin University, Hanoi, Vietnam
| | - Huong Ha Thi Thanh
- Department of Tissue Engineering and Regenerative Medicine, School of Biomedical Engineering, International University, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Norashikin Yahya
- Department of Electrical and Electronics Engineering, Universiti Teknologi PETRONAS (UTP), Bandar Seri Iskandar, Malaysia
| |
Collapse
|
25
|
Núñez-Ríos DL, Nagamatsu ST, Martínez-Magaña JJ, Hurd Y, Rompala G, Krystal JH, Montalvo-Ortiz JL. Mapping the epigenomic landscape of post-traumatic stress disorder in human cortical neurons. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315258. [PMID: 39484232 PMCID: PMC11527063 DOI: 10.1101/2024.10.11.24315258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The study conducted a comprehensive genome-wide analysis of differential 5mC and 5hmC modifications at both CpG and non-CpG sites in postmortem orbitofrontal neurons from 25 PTSD cases and 13 healthy controls. It was observed that PTSD patients exhibit a greater number of differential 5hmC sites compared to 5mC sites. Specifically, individuals with PTSD tend to show hyper-5mC/5hmC at CpG sites, particularly within CpG islands and promoter regions, and hypo-5mC/5hmC at non-CpG sites, especially within intragenic regions. Functional enrichment analysis indicated distinct yet interconnected roles for 5mC and 5hmC in PTSD. The 5mC marks primarily regulate cell-cell adhesion processes, whereas 5hmC marks are involved in embryonic morphogenesis and cell fate commitment. By integrating published PTSD findings from central and peripheral tissues through multi-omics approaches, several biological mechanisms were prioritized, including developmental processes, HPA axis regulation, and immune responses. Based on the consistent enrichment in developmental processes, we hypothesize that if epigenetic changes occur during early developmental stages, they may increase the risk of developing PTSD following trauma exposure. Conversely, if these epigenetic changes occur in adulthood, they may influence neuronal apoptosis and survival mechanisms.
Collapse
Affiliation(s)
- Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Jose Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Yasmin Hurd
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | | | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| |
Collapse
|
26
|
Thörnblom E, Cunningham JL, Gingnell M, Landén M, Bergquist J, Bodén R. Allopregnanolone and progesterone in relation to a single electroconvulsive therapy seizure and subsequent clinical outcome: an observational cohort study. BMC Psychiatry 2024; 24:687. [PMID: 39407178 PMCID: PMC11476534 DOI: 10.1186/s12888-024-06167-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is an important treatment for several severe psychiatric conditions, yet its precise mechanism of action remains unknown. Increased inhibition in the brain after ECT seizures, mediated by γ-aminobutyric acid (GABA), has been linked to clinical effectiveness. Case series on epileptic patients report a postictal serum concentration increase of the GABAA receptor agonist allopregnanolone. Serum allopregnanolone remains unchanged after a full ECT series, but possible transient effects directly after a single ECT seizure remain unexplored. The primary aim was to measure serum concentrations of allopregnanolone and its substrate progesterone after one ECT seizure. Secondary aims were to examine whether concentrations at baseline, or postictal changes, either correlate with seizure generalization or predict clinical outcome ratings after ECT. METHODS A total of 130 participants (18-85 years) were included. Generalization parameters comprised peak ictal heart rate, electroencephalographic (EEG) seizure duration, and prolactin increase. Outcome measures were ratings of clinical global improvement, perceived health status and subjective memory impairment. Non-parametric tests were used for group comparisons and correlations. The prediction analyses were conducted with binary logistic and simple linear regression analyses. RESULTS Allopregnanolone and progesterone remained unchanged and correlated neither with seizure generalization nor with clinical outcome. In men (n = 50), progesterone increased and allopregnanolone change correlated negatively with EEG seizure duration. In a subgroup analysis (n = 62), higher baseline allopregnanolone and progesterone correlated with postictal EEG suppression. CONCLUSIONS ECT seizures have different physiologic effects than generalized seizures in epilepsy. Progesterone might have implications for psychiatric illness in men.
Collapse
Affiliation(s)
- Elin Thörnblom
- Department of Medical Sciences, Uppsala University, Entrance 10, Uppsala, 751 85, Sweden.
| | - Janet L Cunningham
- Department of Medical Sciences, Uppsala University, Entrance 10, Uppsala, 751 85, Sweden
| | - Malin Gingnell
- Department of Medical Sciences, Uppsala University, Entrance 10, Uppsala, 751 85, Sweden
| | - Mikael Landén
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Jonas Bergquist
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Robert Bodén
- Department of Medical Sciences, Uppsala University, Entrance 10, Uppsala, 751 85, Sweden
| |
Collapse
|
27
|
Lei L, Wang YF, Chen CY, Wang YT, Zhang Y. Novel insight into astrocyte-mediated gliotransmission modulates the synaptic plasticity in major depressive disorder. Life Sci 2024; 355:122988. [PMID: 39153595 DOI: 10.1016/j.lfs.2024.122988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Major depressive disorder (MDD) is a form of glial cell-based synaptic dysfunction disease in which glial cells interact closely with neuronal synapses and perform synaptic information processing. Glial cells, particularly astrocytes, are active components of the brain and are responsible for synaptic activity through the release gliotransmitters. A reduced density of astrocytes and astrocyte dysfunction have both been identified the brains of patients with MDD. Furthermore, gliotransmission, i.e., active information transfer mediated by gliotransmitters between astrocytes and neurons, is thought to be involved in the pathogenesis of MDD. However, the mechanism by which astrocyte-mediated gliotransmission contributes to depression remains unknown. This review therefore summarizes the alterations in astrocytes in MDD, including astrocyte marker, connexin 43 (Cx43) expression, Cx43 gap junctions, and Cx43 hemichannels, and describes the regulatory mechanisms of astrocytes involved in synaptic plasticity. Additionally, we investigate the mechanisms acting of the glutamatergic, gamma-aminobutyric acidergic, and purinergic systems that modulate synaptic function and the antidepressant mechanisms of the related receptor antagonists. Further, we summarize the roles of glutamate, gamma-aminobutyric acid, d-serine, and adenosine triphosphate in depression, providing a basis for the identification of diagnostic and therapeutic targets for MDD.
Collapse
Affiliation(s)
- Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
28
|
Gonda X, Tarazi FI, Dome P. The emergence of antidepressant drugs targeting GABA A receptors: A concise review. Biochem Pharmacol 2024; 228:116481. [PMID: 39147329 DOI: 10.1016/j.bcp.2024.116481] [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: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Depression is among the most common psychiatric illnesses, which imposes a major socioeconomic burden on patients, caregivers, and the public health system. Treatment with classical antidepressants (e.g. tricyclic antidepressants and selective serotonine reuptake inhibitors), which primarily affect monoaminergic systems has several limitations, such as delayed onset of action and moderate efficacy in a relatively large proportion of depressed patients. Furthermore, depression is highly heterogeneus, and its different subtypes, including post-partum depression, involve distinct neurobiology, warranting a differential approach to pharmacotherapy. Given these shortcomings, the need for novel antidepressants that are superior in efficacy and faster in onset of action is fully justified. The development and market introduction of rapid-acting antidepressants has accelerated in recent years. Some of these new antidepressants act through the GABAergic system. In this review, we discuss the discovery, efficacy, and limitations of treatment with classic antidepressants. We provide a detailed discussion of GABAergic neurotransmission, with a special focus on GABAA receptors, and possible explanations for the mood-enhancing effects of GABAergic medications (in particular neurosteroids acting at GABAA receptors), and, ultimately, we present the most promising molecules belonging to this family which are currently used in clinical practice or are in late phases of clinical development.
Collapse
Affiliation(s)
- Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.
| | - Frank I Tarazi
- Department of Psychiatry and Neurology, Harvard Medical School and McLean Hospital, Boston, MA, USA
| | - Peter Dome
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Nyiro Gyula National Institute of Psychiatry and Addictology, Budapest, Hungary
| |
Collapse
|
29
|
Mazurie Z, Branchereau P, Cattaert D, Henkous N, Savona-Baron C, Vouimba RM. Acute stress differently modulates interneurons excitability and synaptic plasticity in the primary motor cortex of wild-type and SOD1 G93A mouse model of ALS. J Physiol 2024; 602:4987-5015. [PMID: 39216080 DOI: 10.1113/jp285210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Primary motor cortex (M1) network stability depends on activity of inhibitory interneurons, for which susceptibility to stress was previously demonstrated in limbic regions. Hyperexcitability in M1 following changes in the excitatory/inhibitory balance is a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Using electrophysiological approaches, we assessed the impact of acute restraint stress on inhibitory interneurons excitability and global synaptic plasticity in M1 of the SOD1G93A ALS mouse model at a late pre-symptomatic stage (10-12.5 weeks). Based on their firing type (continuous, discontinuous, with accommodation or not) and electrophysiological characteristics (resting potential, rheobase, firing frequency), interneurons from M1 slices were separated into four clusters, labelled from 1 to 4. Among them, only interneurons from the first cluster, presenting continuous firing with few accommodations, tended to show increased excitability in wild-type (WT) and decreased excitability in SOD1G93A animals following stress. In vivo analyses of evoked field potentials showed that stress suppressed the theta burst-induced plasticity of an excitatory component (N1) recorded in the superficial layers of M1 in WT, with no impact on an inhibitory complex (N2-P1) from the deeper layers. In SOD1G93A mice, stress did not affect N1 but suppressed the N2-P1 plasticity. These data suggest that stress can alter M1 network functioning in a different manner in WT and SOD1G93A mice, possibly through changes of inhibitory interneurons excitability and synaptic plasticity. This suggests that stress-induced activity changes in M1 may therefore influence ALS outcomes. KEY POINTS: Disruption of the excitatory/inhibitory balance in the primary motor cortex (M1) has been linked to cortical hyperexcitability development, a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Psychological stress was reported to influence excitatory/inhibitory balance in limbic regions, but very little is known about its influence on the M1 functioning under physiological or pathological conditions. Our study revealed that acute stress influences the excitatory/inhibitory balance within the M1, through changes in interneurons excitability along with network plasticity. Such changes were different in pathological (SOD1G93A ALS mouse model) vs. physiological (wild-type) conditions. The results of our study help us to better understand how stress modulates the M1 and highlight the need to further characterize stress-induced motor cortex changes because it may be of importance when evaluating ALS outcomes.
Collapse
Affiliation(s)
- Zoé Mazurie
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Nadia Henkous
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Catherine Savona-Baron
- Present address: BoRdeaux Institute of onCology (BRIC), INSERM U1312, University of Bordeaux, Bordeaux, France
| | - Rose-Marie Vouimba
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
30
|
King C, Maze T, Plakke B. Altered prefrontal and cerebellar parvalbumin neuron counts are associated with cognitive changes in male rats. Exp Brain Res 2024; 242:2295-2308. [PMID: 39085433 PMCID: PMC12063742 DOI: 10.1007/s00221-024-06902-y] [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: 05/30/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Exposure to valproic acid (VPA), a common anti-seizure medication, in utero is a risk factor for autism spectrum disorder (ASD). People with ASD often display changes in the cerebellum, including volume changes, altered circuitry, and changes in Purkinje cell populations. ASD is also characterized by changes in the medial prefrontal cortex (mPFC), where excitatory/inhibitory balance is often altered. This study exposed rats to a high dose of VPA during gestation and assessed cognition and anxiety-like behaviors during young adulthood using a set-shifting task and the elevated plus maze. Inhibitory parvalbumin-expressing (PV +) neuron counts were assessed in the mPFC and cerebellar lobules VI and VII (Purkinje cell layers), which are known to modulate cognition. VPA males had increased PV + counts in crus I and II of lobule VII. VPA males also had decreased parvalbumin-expressing neuron counts in the mPFC. It was also found that VPA-exposed rats, regardless of sex, had increased parvalbumin-expressing Purkinje cell counts in lobule VI. In males, this was associated with impaired intra-dimensional shifting on a set-shifting task. Purkinje cell over proliferation may be contributing to the previously observed increase in volume of Lobule VI. These findings suggest that altered inhibitory signaling in cerebellar-frontal circuits may contribute to the cognitive deficits that occur within ASD.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, 1114 Mid-Campus Dr., Manhattan, KS, 66506, USA
| | - Tessa Maze
- Psychological Sciences, Kansas State University, 1114 Mid-Campus Dr., Manhattan, KS, 66506, USA
| | - Bethany Plakke
- Psychological Sciences, Kansas State University, 1114 Mid-Campus Dr., Manhattan, KS, 66506, USA.
| |
Collapse
|
31
|
Teixeira-Silva B, de Mattos GVRM, Carvalho VDF, Campello-Costa P. Caffeine intake during lactation has a sex-dependent effect on the hippocampal excitatory/inhibitory balance and pups' behavior. Brain Res 2024; 1846:149247. [PMID: 39304106 DOI: 10.1016/j.brainres.2024.149247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
During early life, disruptions in glutamatergic and GABAergic synapse development in the hippocampus may contribute to several neurodevelopmental disorders, including cognitive deficits and psychiatric disorders. Caffeine is the most consumed psychoactive drug in the world, and previous work from our group has shown that caffeine disrupts visual system connections at different stages of development. This work aimed to investigate the effects of caffeine consumption during lactation in the glutamatergic and GABAergic synaptic markers in the hippocampus and on the behavior of rat offspring. We found that maternal caffeine intake significantly reduced GluN1 subunits of the NMDA receptor, increased the GluA1/GluA2 ratio of AMPA receptor in the dorsal hippocampus, and decreased GAD content in female pups' ventral hippocampus. On the other hand, an increase in GluN1/GluN2b subunits, a decrease in GAD content in the dorsal hippocampus, and a reduction of the GluA1 content in the ventral hippocampus were observed in male pups. In addition, changes in the behavior of the offspring submitted to indirect caffeine consumption were also sex-dependent, with females developing anxiety-like behavior and males showing anxiety-like behavior and hyper-locomotion. These results highlight that maternal caffeine intake promotes changes in the hippocampal excitatory and inhibitory balance and offspring behavior in a sex-dependent manner, suggesting that the population should be alerted to reduced caffeine consumption by breastfeeding mothers.
Collapse
Affiliation(s)
- Bruna Teixeira-Silva
- Programa de Pós-graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | | | - Vinicius de Frias Carvalho
- Programa de Pós-graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Laboratório de Inflamação, Centro de Pesquisa, Inovação e Vigilância em Covid-19 e Emergências Sanitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n° 4036, Manguinhos, CEP 21041-361 Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n° 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil
| | - Paula Campello-Costa
- Programa de Pós-graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|
32
|
Arora I, Mal P, Arora P, Paul A, Kumar M. GABAergic implications in anxiety and related disorders. Biochem Biophys Res Commun 2024; 724:150218. [PMID: 38865810 DOI: 10.1016/j.bbrc.2024.150218] [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: 02/28/2024] [Revised: 05/05/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024]
Abstract
Evidence indicates that anxiety disorders arise from an imbalance in the functioning of brain circuits that govern the modulation of emotional responses to possibly threatening stimuli. The circuits under consideration in this context include the amygdala's bottom-up activity, which signifies the existence of stimuli that may be seen as dangerous. Moreover, these circuits encompass top-down regulatory processes that originate in the prefrontal cortex, facilitating the communication of the emotional significance associated with the inputs. Diverse databases (e.g., Pubmed, ScienceDirect, Web of Science, Google Scholar) were searched for literature using a combination of different terms e.g., "anxiety", "stress", "neuroanatomy", and "neural circuits", etc. A decrease in GABAergic activity is present in both anxiety disorders and severe depression. Research on cerebral functional imaging in depressive individuals has shown reduced levels of GABA within the cortical regions. Additionally, animal studies demonstrated that a reduction in the expression of GABAA/B receptors results in a behavioral pattern resembling anxiety. The amygdala consists of inhibitory networks composed of GABAergic interneurons, responsible for modulating anxiety responses in both normal and pathological conditions. The GABAA receptor has allosteric sites (e.g., α/γ, γ/β, and α/β) which enable regulation of neuronal inhibition in the amygdala. These sites serve as molecular targets for anxiolytic medications such as benzodiazepine and barbiturates. Alterations in the levels of naturally occurring regulators of these allosteric sites, along with alterations to the composition of the GABAA receptor subunits, could potentially act as mechanisms via which the extent of neuronal inhibition is diminished in pathological anxiety disorders.
Collapse
Affiliation(s)
- Indu Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pankaj Mal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Poonam Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Anushka Paul
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
33
|
Boyle N, Betts S, Lu H. Monoaminergic Modulation of Learning and Cognitive Function in the Prefrontal Cortex. Brain Sci 2024; 14:902. [PMID: 39335398 PMCID: PMC11429557 DOI: 10.3390/brainsci14090902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/09/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Extensive research has shed light on the cellular and functional underpinnings of higher cognition as influenced by the prefrontal cortex. Neurotransmitters act as key regulatory molecules within the PFC to assist with synchronizing cognitive state and arousal levels. The monoamine family of neurotransmitters, including dopamine, serotonin, and norepinephrine, play multifaceted roles in the cognitive processes behind learning and memory. The present review explores the organization and signaling patterns of monoamines within the PFC, as well as elucidates the numerous roles played by monoamines in learning and higher cognitive function.
Collapse
Affiliation(s)
| | | | - Hui Lu
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (N.B.); (S.B.)
| |
Collapse
|
34
|
Rodrigues D, Santa C, Manadas B, Monteiro P. Chronic Stress Alters Synaptic Inhibition/Excitation Balance of Pyramidal Neurons But Not PV Interneurons in the Infralimbic and Prelimbic Cortices of C57BL/6J Mice. eNeuro 2024; 11:ENEURO.0053-24.2024. [PMID: 39147579 DOI: 10.1523/eneuro.0053-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024] Open
Abstract
The medial prefrontal cortex (mPFC) plays a pivotal role in regulating working memory, executive function, and self-regulatory behaviors. Dysfunction in the mPFC circuits is a characteristic feature of several neuropsychiatric disorders including schizophrenia, depression, and post-traumatic stress disorder. Chronic stress (CS) is widely recognized as a major triggering factor for the onset of these disorders. Although evidence suggests synaptic dysfunction in mPFC circuits following CS exposure, it remains unclear how different neuronal populations in the infralimbic (IL) and prelimbic (PL) cortices are affected in terms of synaptic inhibition/excitation balance (I/E ratio). Here, using neuroproteomic analysis and whole-cell patch-clamp recordings in pyramidal neurons (PNs) and parvalbumin (PV) interneurons within the PL and IL cortices, we examined the synaptic changes after 21 d of chronic unpredictable stress, in male mice. Our results reveal distinct impacts of CS on PL and IL PNs, resulting in an increased I/E ratio in both subregions but through different mechanisms: CS increases inhibitory synaptic drive in the PL while decreasing excitatory synaptic drive in the IL. Notably, the I/E ratio and excitatory and inhibitory synaptic drive of PV interneurons remained unaffected in both PL and IL circuits following CS exposure. These findings offer novel mechanistic insights into the influence of CS on mPFC circuits and support the hypothesis of stress-induced mPFC hypofunction.
Collapse
Affiliation(s)
- Diana Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Braga 4710-057, Portugal
- Biomedizinisches Centrum München (BMC), Ludwig-Maximilians-Universität München, Munich 82152, Bayern, Germany
| | - Cátia Santa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-517, Portugal
| | - Patrícia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Braga 4710-057, Portugal
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine, University of Porto, Porto 4200-319, Portugal
- RISE-Health, Health Research Network, Porto 4200-319, Portugal
| |
Collapse
|
35
|
Kuhn YA, Egger S, Bugnon M, Lehmann N, Taubert M, Taube W. Age-related decline in GABAergic intracortical inhibition can be counteracted by long-term learning of balance skills. J Physiol 2024; 602:3737-3753. [PMID: 38949035 DOI: 10.1113/jp285706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 06/11/2024] [Indexed: 07/02/2024] Open
Abstract
Ageing induces a decline in GABAergic intracortical inhibition, which seems to be associated not only with decremental changes in well-being, sleep quality, cognition and pain management but also with impaired motor control. So far, little is known regarding whether targeted interventions can prevent the decline of intracortical inhibition in the primary motor cortex in the elderly. Therefore, the present study investigated whether age-related cortical dis-inhibition could be reversed after 6 months of balance learning and whether improvements in postural control correlated with the extent of reversed dis-inhibition. The results demonstrated that intracortical inhibition can be upregulated in elderly subjects after long-term balance learning and revealed a correlation between changes in balance performance and intracortical inhibition. This is the first study to show physical activity-related upregulation of GABAergic inhibition in a population with chronic dis-inhibition and may therefore be seminal for many pathologies in which the equilibrium between inhibitory and excitatory neurotransmitters is disturbed. KEY POINTS: Ageing induces a decline in GABAergic intracortical inhibition. So far, little is known regarding whether targeted interventions can prevent the decline of intracortical inhibition in the primary motor cortex in the elderly. After 6 months of balance learning, intracortical inhibition can be upregulated in elderly subjects. The results of this study also revealed a correlation between changes in balance performance and intracortical inhibition. This is the first study to show physical activity-related upregulation of GABAergic inhibition in a population with chronic dis-inhibition.
Collapse
Affiliation(s)
- Yves-Alain Kuhn
- Department of Neurosciences and Movement Science, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Sven Egger
- Department of Neurosciences and Movement Science, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Matteo Bugnon
- Department of Neurosciences and Movement Science, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Nico Lehmann
- Department of Neurosciences and Movement Science, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Sport Science, Institute III, Faculty of Humanities, Otto von Guericke University, Magdeburg, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Collaborative Research Center 1436 Neural Resources of Cognition, Otto von Guericke University, Magdeburg, Germany
| | - Marco Taubert
- Department of Sport Science, Institute III, Faculty of Humanities, Otto von Guericke University, Magdeburg, Germany
- Collaborative Research Center 1436 Neural Resources of Cognition, Otto von Guericke University, Magdeburg, Germany
- Center for Behavioral and Brain Science (CBBS), Otto von Guericke University, Universitätsplatz 2, Magdeburg, Germany
| | - Wolfgang Taube
- Department of Neurosciences and Movement Science, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
36
|
Prvulovic M, Sokanovic S, Simeunovic V, Vukojevic A, Jovic M, Todorovic S, Mladenovic A. The complex relationship between late-onset caloric restriction and synaptic plasticity in aged Wistar rats. IUBMB Life 2024; 76:548-562. [PMID: 38390757 DOI: 10.1002/iub.2812] [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: 11/06/2023] [Accepted: 01/15/2024] [Indexed: 02/24/2024]
Abstract
Age-related reduction in spine density, synaptic marker expression, and synaptic efficiency are frequently reported. These changes provide the cellular and molecular basis for the cognitive decline characteristic for old age. Nevertheless, there are several approaches that have the potential to ameliorate these processes and improve cognition, caloric restriction being one of the most promising and widely studied. While lifelong caloric restriction is known for its numerous beneficial effects, including improved cognitive abilities and increased expression of proteins essential for synaptic structure and function, the effects of late-onset and/or short-term CR on synaptic plasticity have yet to be investigated. We have previously documented that the effects of CR are strongly dependent on whether CR is initiated in young or old subjects. With this in mind, we conducted a long-term study in aging Wistar rats to examine changes in the expression of several key synaptic markers under the regimen of CR started at different time points in life. We found a significant increase in the expression of both presynaptic and postsynaptic markers. However, taking into account previously reported changes in the behavior detected in these animals, we consider that this increase cannot represent beneficial effect of CR.
Collapse
Affiliation(s)
- Milica Prvulovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Srdjan Sokanovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Simeunovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Andjela Vukojevic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milena Jovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Smilja Todorovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Mladenovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
37
|
Chen M, Wang C, Lin Y, Chen Y, Xie W, Huang X, Zhang F, Fu C, Zhuang K, Zou T, Can D, Li H, Wu S, Luo C, Zhang J. Dorsal raphe nucleus-hippocampus serotonergic circuit underlies the depressive and cognitive impairments in 5×FAD male mice. Transl Neurodegener 2024; 13:34. [PMID: 39044270 PMCID: PMC11267773 DOI: 10.1186/s40035-024-00425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Depressive symptoms often occur in patients with Alzheimer's disease (AD) and exacerbate the pathogenesis of AD. However, the neural circuit mechanisms underlying the AD-associated depression remain unclear. The serotonergic system plays crucial roles in both AD and depression. METHODS We used a combination of in vivo trans-synaptic circuit-dissecting anatomical approaches, chemogenetic manipulations, optogenetic manipulations, pharmacological methods, behavioral testing, and electrophysiological recording to investigate dorsal raphe nucleus serotonergic circuit in AD-associated depression in AD mouse model. RESULTS We found that the activity of dorsal raphe nucleus serotonin neurons (DRN5-HT) and their projections to the dorsal hippocampal CA1 (dCA1) terminals (DRN5-HT-dCA1CaMKII) both decreased in brains of early 5×FAD mice. Chemogenetic or optogenetic activation of the DRN5-HT-dCA1CaMKII neural circuit attenuated the depressive symptoms and cognitive impairments in 5×FAD mice through serotonin receptor 1B (5-HT1BR) and 4 (5-HT4R). Pharmacological activation of 5-HT1BR or 5-HT4R attenuated the depressive symptoms and cognitive impairments in 5×FAD mice by regulating the DRN5-HT-dCA1CaMKII neural circuit to improve synaptic plasticity. CONCLUSIONS These findings provide a new mechanistic connection between depression and AD and provide potential pharmaceutical prevention targets for AD.
Collapse
Affiliation(s)
- Meiqin Chen
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Chenlu Wang
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, 361000, China
| | - Yinan Lin
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, 361000, China
| | - Yanbing Chen
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wenting Xie
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xiaoting Huang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Fan Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Congrui Fu
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Kai Zhuang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Tingting Zou
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Dan Can
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Huifang Li
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China.
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China.
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Institute of Neuroscience, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
38
|
Zhao M, Li X, Li F, Hu X, Wang J, Liu Y, Zhang C, Bai J, Edden RAE, Gao F, Su M, Ren F. Identification of neurotransmitter imbalances in the cingulate cortex of NMOSD patients using magnetic resonance spectroscopy. Cereb Cortex 2024; 34:bhae304. [PMID: 39073381 PMCID: PMC11284173 DOI: 10.1093/cercor/bhae304] [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: 12/23/2023] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
Cognitive impairment affects 29-67% of patients with neuromyelitis optica spectrum disorder. Previous studies have reported glutamate homeostasis disruptions in astrocytes, leading to imbalances in gamma-aminobutyric acid levels. However, the association between these neurotransmitter changes and cognitive deficits remains inadequately elucidated. Point RESolved Spectroscopy and Hadamard Encoding and Reconstruction of MEGA-Edited Spectroscopy techniques were utilized to evaluate gamma-aminobutyric acid, glutamate, glutathione levels, and excitation/inhibition balance in the anterior cingulate cortex, posterior cingulate cortex, and occipital cortex of 39 neuromyelitis optica spectrum disorder patients and 41 healthy controls. Cognitive function was assessed using neurocognitive scales. Results showed decreased gamma-aminobutyric acid levels alongside increased glutamate, glutathione, and excitation/inhibition ratio in the anterior cingulate cortex and posterior cingulate cortex of neuromyelitis optica spectrum disorder patients. Specifically, within the posterior cingulate cortex of neuromyelitis optica spectrum disorder patients, decreased gamma-aminobutyric acid levels and increased excitation/inhibition ratio correlated significantly with anxiety scores, whereas glutathione levels predicted diminished executive function. The results suggest that neuromyelitis optica spectrum disorder patients exhibit dysregulation in the GABAergic and glutamatergic systems in their brains, where the excitation/inhibition imbalance potentially acts as a neuronal metabolic factor contributing to emotional disorders. Additionally, glutathione levels in the posterior cingulate cortex region may serve as predictors of cognitive decline, highlighting the potential benefits of reducing oxidative stress to safeguard cognitive function in neuromyelitis optica spectrum disorder patients.
Collapse
Affiliation(s)
- Min Zhao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Department of Radiology, Linyi Central Hospital, Linyi, China
| | - Xiao Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Fuyan Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Xin Hu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Jing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yuxi Liu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Chuanchen Zhang
- Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Jie Bai
- Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Fei Gao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Meixia Su
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Fuxin Ren
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| |
Collapse
|
39
|
Zhang BB, Ling XY, Shen QY, Zhang YX, Li QX, Xie ST, Li HZ, Zhang QP, Yung WH, Wang JJ, Ke Y, Zhang XY, Zhu JN. Suppression of excitatory synaptic transmission in the centrolateral amygdala via presynaptic histamine H3 heteroreceptors. J Physiol 2024. [PMID: 38953534 DOI: 10.1113/jp286392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
The central histaminergic system has a pivotal role in emotional regulation and psychiatric disorders, including anxiety, depression and schizophrenia. However, the effect of histamine on neuronal activity of the centrolateral amygdala (CeL), an essential node for fear and anxiety processing, remains unknown. Here, using immunostaining and whole-cell patch clamp recording combined with optogenetic manipulation of histaminergic terminals in CeL slices prepared from histidine decarboxylase (HDC)-Cre rats, we show that histamine selectively suppresses excitatory synaptic transmissions, including glutamatergic transmission from the basolateral amygdala, on both PKC-δ- and SOM-positive CeL neurons. The histamine-induced effect is mediated by H3 receptors expressed on VGLUT1-/VGLUT2-positive presynaptic terminals in CeL. Furthermore, optoactivation of histaminergic afferent terminals from the hypothalamic tuberomammillary nucleus (TMN) also significantly suppresses glutamatergic transmissions in CeL via H3 receptors. Histamine neither modulates inhibitory synaptic transmission by presynaptic H3 receptors nor directly excites CeL neurons by postsynaptic H1, H2 or H4 receptors. These results suggest that histaminergic afferent inputs and presynaptic H3 heteroreceptors may hold a critical position in balancing excitatory and inhibitory synaptic transmissions in CeL by selective modulation of glutamatergic drive, which may not only account for the pathophysiology of psychiatric disorders but also provide potential psychotherapeutic targets. KEY POINTS: Histamine selectively suppresses the excitatory, rather than inhibitory, synaptic transmissions on both PKC-δ- and SOM-positive neurons in the centrolateral amygdala (CeL). H3 receptors expressed on VGLUT1- or VGLUT2-positive afferent terminals mediate the suppression of histamine on glutamatergic synaptic transmission in CeL. Optogenetic activation of hypothalamic tuberomammillary nucleus (TMN)-CeL histaminergic projections inhibits glutamatergic transmission in CeL via H3 receptors.
Collapse
Affiliation(s)
- Bei-Bei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xin-Yu Ling
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qing-Yi Shen
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yang-Xun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qian-Xiao Li
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shu-Tao Xie
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hong-Zhao Li
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qi-Peng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Wing-Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, SAR, China
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| |
Collapse
|
40
|
Ng THJ, Sarikahya MH, Hudson R, Szkudlarek HJ, Pérez-Valenzuela E, Uzuneser TC, Proud E, Gummerson D, Youssef M, Machado M, Zhaksylyk K, DeVuono MV, Chen C, Yeung KKC, Rushlow WJ, Laviolette SR. Adolescent nicotine exposure induces long-term, sex-specific disturbances in mood and anxiety-related behavioral, neuronal and molecular phenotypes in the mesocorticolimbic system. Neuropsychopharmacology 2024; 49:1171-1182. [PMID: 38521861 PMCID: PMC11109238 DOI: 10.1038/s41386-024-01853-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
The majority of lifetime smokers begin using nicotine during adolescence, a critical period of brain development wherein neural circuits critical for mood, affect and cognition are vulnerable to drug-related insults. Specifically, brain regions such as the medial prefrontal cortex (mPFC), the ventral tegmental area (VTA), nucleus accumbens (NAc) and hippocampus, are implicated in both nicotine dependence and pathological phenotypes linked to mood and anxiety disorders. Clinical studies report that females experience higher rates of mood/anxiety disorders and are more resistant to smoking cessation therapies, suggesting potential sex-specific responses to nicotine exposure and later-life neuropsychiatric risk. However, the potential neural and molecular mechanisms underlying such sex differences are not clear. In the present study, we compared the impacts of adolescent nicotine exposure in male vs. female rat cohorts. We performed a combination of behavioral, electrophysiological and targeted protein expression analyses along with matrix assisted laser deionization imaging (MALDI) immediately post-adolescent exposure and later in early adulthood. We report that adolescent nicotine exposure induced long-lasting anxiety/depressive-like behaviors, disrupted neuronal activity patterns in the mPFC-VTA network and molecular alterations in various neural regions linked to affect, anxiety and cognition. Remarkably, these phenotypes were only observed in males and/or were expressed in the opposite direction in females. These findings identify a series of novel, sex-selective biomarkers for adolescent nicotine-induced neuropsychiatric risk, persisting into adulthood.
Collapse
Affiliation(s)
- Tsun Hay Jason Ng
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Enzo Pérez-Valenzuela
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Taygun C Uzuneser
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Emma Proud
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Dana Gummerson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Miray Youssef
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Madeline Machado
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Kuralay Zhaksylyk
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Marieka V DeVuono
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Chaochao Chen
- Department of Chemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
- Department of Biochemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Ken K-C Yeung
- Department of Chemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
- Department of Biochemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
- Department of Psychiatry, Western University, London, ON, N6A 3K7, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada.
- Department of Psychiatry, Western University, London, ON, N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada.
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), London, ON, Canada.
| |
Collapse
|
41
|
Gao M, Kirk M, Lash E, Knight H, Michalopoulou M, Guess N, Browning M, Weich S, Burnet P, Jebb SA, Stevens R, Aveyard P. Evaluating the efficacy and mechanisms of a ketogenic diet as adjunctive treatment for people with treatment-resistant depression: A protocol for a randomised controlled trial. J Psychiatr Res 2024; 174:230-236. [PMID: 38653031 DOI: 10.1016/j.jpsychires.2024.04.023] [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: 02/05/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND One-third of people with depression do not respond to antidepressants, and, after two adequate courses of antidepressants, are classified as having treatment-resistant depression (TRD). Some case reports suggest that ketogenic diets (KDs) may improve some mental illnesses, and preclinical data indicate that KDs can influence brain reward signalling, anhedonia, cortisol, and gut microbiome which are associated with depression. To date, no trials have examined the clinical effect of a KD on TRD. METHODS This is a proof-of-concept randomised controlled trial to investigate the efficacy of a six-week programme of weekly dietitian counselling plus provision of KD meals, compared with an intervention involving similar dietetic contact time and promoting a healthy diet with increased vegetable consumption and reduction in saturated fat, plus food vouchers to purchase healthier items. At 12 weeks we will assess whether participants have continued to follow the assigned diet. The primary outcome is the difference between groups in the change in Patient Health Questionnaire-9 (PHQ-9) score from baseline to 6 weeks. PHQ-9 will be measured at weeks 2, 4, 6 and 12. The secondary outcomes are the differences between groups in the change in remission of depression, change in anxiety score, functioning ability, quality of life, cognitive performance, reward sensitivity, and anhedonia from baseline to 6 and 12 weeks. We will also assess whether changes in reward sensitivity, anhedonia, cortisol awakening response and gut microbiome may explain any changes in depression severity. DISCUSSION This study will test whether a ketogenic diet is an effective intervention to reduce the severity of depression, anxiety and improve quality of life and functioning ability for people with treatment-resistant depression.
Collapse
Affiliation(s)
- Min Gao
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK.
| | - Megan Kirk
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK
| | - Eva Lash
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK
| | - Heather Knight
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Moscho Michalopoulou
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nicola Guess
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Scott Weich
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Philip Burnet
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Susan A Jebb
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Richard Stevens
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Paul Aveyard
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford, UK; NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
42
|
Shang Y, Su Q, Ma R, Chen M, Zhao Z, Yao C, Han L, Yao Z, Hu B. Functional Connectome Hierarchy Distortions in Female Nurses With Occupational Burnout and Its Gene Expression Signatures. J Magn Reson Imaging 2024; 59:2124-2136. [PMID: 37728385 DOI: 10.1002/jmri.28985] [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: 05/24/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Burnout has become a serious public health issue worldwide, particularly during the COVID-19 pandemic. Functional connectome impairments associated with occupational burnout were widely distributed, involving both low-level sensorimotor cortices and high-level association cortices. PURPOSE To investigate whether there are hierarchical perturbations in the functional connectomes and if these perturbations are potentially influenced by genetic factors in nurses who feel "burned out." STUDY TYPE Prospective, case control. POPULATION Thirty-three female nurses with occupational burnout (aged 27-40, 32.42 ± 3.37) and 32 matched nurses who were not feeling burned out (aged 27-42, 32.50 ± 4.21). FIELD STRENGTH/SEQUENCE 3.0 T, gradient-echo echo-planar imaging sequence (GE-EPI). ASSESSMENT Gradient-based techniques were used to depict the perturbations in the multi-dimensional hierarchical structure of the macroscale connectome. Gene expression data were acquired from the Allen Human Brain Atlas. STATISTICAL TESTS Cortex-wide multivariate analyses were used for between-group differences in gradients as well as association analyses between the hierarchy distortions and the MBI score (FDR corrected). Partial least squares, spin test and bootstrapping were utilized together to select the gene sets (FDR corrected). Gene enrichment analyses (GO, KEGG and cell-type) were further performed. Significance level: P < 0.05. RESULTS There were significant gradient distortions, with strong between-group effects in the somatosensory network and moderate effects in the higher-order default-mode network, which were significantly correlated with the gene expression profiles (r = 0.3171). The most related genes were broadly involved in the cellular response to minerals, neuronal plasticity, and the circadian rhythm pathway (q value < 0.01). Significant enrichments were found in excitatory (r = 0.2588), inhibitory neurons (r = 0.2610), and astrocytes cells (r = 0.2633). Regions affected by burnout severity were mainly distributed in the association and visual cortices. DATA CONCLUSION By connecting in vivo imaging to genes, cell classes, and clinical data, this study provides a framework to understand functional impairments in occupational burnout and how the microscopic genetic architecture drive macroscopic distortions. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Yingying Shang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Qian Su
- Department of Nursing, Gansu Provincial Hospital, Lanzhou, Gansu Province, China
- The First Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
| | - Rong Ma
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Miao Chen
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Ziyang Zhao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Chaofan Yao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Lin Han
- Department of Nursing, Gansu Provincial Hospital, Lanzhou, Gansu Province, China
- The First Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- School of Nursing, Lanzhou University, Lanzhou, Gansu Province, China
| | - Zhijun Yao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Bin Hu
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
- Joint Research Center for Cognitive Neurosensor Technology of Lanzhou University & Institute of Semiconductors, Chinese Academy of Sciences, Lanzhou, Gansu Province, China
- Engineering Research Center of Open Source Software and Real-Time System (Lanzhou University), Ministry of Education, Lanzhou, Gansu Province, China
| |
Collapse
|
43
|
Qian J, Yu F, Zheng L, Luo D, Zhao M. Comparison of the Protective Effects of Casein Hydrolysate Containing Tyr-Pro-Val-Glu-Pro-Phe and Casein on the Behaviors and Peripheral and Brain Functions in Mice with Chronic-Stress-Induced Anxiety and Insomnia. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:11515-11530. [PMID: 38726599 DOI: 10.1021/acs.jafc.4c01074] [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: 05/23/2024]
Abstract
Chronic stress is a major inducer of anxiety and insomnia. Milk casein has been studied for its stress-relieving effects. We previously prepared a casein hydrolysate (CP) rich in the sleep-enhancing peptide YPVEPF, and this study aims to systemically investigate the different protective effects of CP and casein on dysfunction and anxiety/insomnia behavior and its underlying mechanisms in chronically stressed mice. Behavioral results showed that CP ameliorated stress-induced insomnia and anxiety more effectively than milk casein, and this difference in amelioration was highly correlated with an increase in GABA, 5-HT, GABAA, 5-HT1A receptors, and BDNF and a decrease in IL-6 and NMDA receptors in stressed mice. Furthermore, CP restored these dysfunctions in the brain and colon by activating the HPA response, modulating the ERK/CREB-BDNF-TrκB signaling pathway, and alleviating inflammation. The abundant YPVEPF (1.20 ± 0.04%) and Tyr-based/Trp-containing peptides of CP may be the key reasons for its different effects compared to casein. Thus, this work revealed the main active structures of CP and provided a novel dietary intervention strategy for the prevention and treatment of chronic-stress-induced dysfunction and anxiety/insomnia behaviors.
Collapse
Affiliation(s)
- Jingjing Qian
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Fengjie Yu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Donghui Luo
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521000, China
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521000, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| |
Collapse
|
44
|
Bove M, Morgese MG, Dimonte S, Sikora V, Agosti LP, Palmieri MA, Tucci P, Schiavone S, Trabace L. Increased stress vulnerability in the offspring of socially isolated rats: Behavioural, neurochemical and redox dysfunctions. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110945. [PMID: 38242425 DOI: 10.1016/j.pnpbp.2024.110945] [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: 07/28/2023] [Revised: 12/22/2023] [Accepted: 01/14/2024] [Indexed: 01/21/2024]
Abstract
Stressful events during pregnancy impact on the progeny neurodevelopment. However, little is known about preconceptional stress effects. The rat social isolation represents an animal model of chronic stress inducing a variety of dysfunctions. Moreover, social deprivation during adolescence interferes with key neurodevelopmental processes. Here, we investigated the development of behavioural, neurochemical and redox alterations in the male offspring of socially isolated female rats before pregnancy, reared in group (GRP) or in social isolation (ISO) from weaning until young-adulthood. To this aim, females were reared in GRP or in ISO conditions, from PND21 to PND70, when they were mated. Their male offspring was housed in GRP or ISO conditions through adolescence and until PND70, when passive avoidance-PA, novel object recognition-NOR and open field-OF tests were performed. Levels of noradrenaline (NA), serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), glutamate (GLU) and GABA were assessed in the prefrontal cortex (PFC). Moreover, cortical ROS levels were quantified, as well as NF-kB and the NADPH oxidase NOX2 expression, redox status (expressed as GSH:GSSG ratio) and SOD1 amount. A significant decrease of the latency time in the PA was observed in the offspring of ISO females. In the NOR test, while a significant increase in the exploratory activity towards the novel object was observed in the offspring of GRP females, no significant differences were found in the offspring of ISO females. No significant differences were found in the OF test among experimental groups. Theoffspring of ISO females showed increased NA and 5-HIAA levels, whereas in the offspring persistently housed in isolation condition from weaninguntil adulthood, we detected reduced 5-HT levels and ehnanced 5-HIAA amount. No significant changes in GLU concentrations were detected, while decreased GABA content was observed in the offspring of ISO females exposed to social isolation. Increased ROS levels as well as reduced NF-κB, NOX2 expression were detected in the offspring of ISO females. This was accompanied by reduced redox status and enhanced SOD1 levels. In conclusion, our results suggest that female exposure to chronic social stress before pregnancy might have a profound influence on the offspring neurodevelopment in terms of cognitive, neurochemical and redox-related alterations, identifying this specific time window for possible preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Lisa Pia Agosti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Maria Adelaide Palmieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy.
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, Foggia 71122, Italy
| |
Collapse
|
45
|
Johnstone N, Cohen Kadosh K. Excitatory and inhibitory neurochemical markers of anxiety in young females. Dev Cogn Neurosci 2024; 66:101363. [PMID: 38447470 PMCID: PMC10925933 DOI: 10.1016/j.dcn.2024.101363] [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: 11/06/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
Between the ages of 10-25 years the maturing brain is sensitive to a multitude of changes, including neurochemical variations in metabolites. Of the different metabolites, gamma-aminobutyric acid (GABA) has long been linked neurobiologically to anxiety symptomology, which begins to manifest in adolescence. To prevent persistent anxiety difficulties into adulthood, we need to understand the maturational trajectories of neurochemicals and how these relate to anxiety levels during this sensitive period. We used magnetic resonance spectroscopy in a sample of younger (aged 10-11) and older (aged 18-25) females to estimate GABA and glutamate levels in brain regions linked to emotion regulation processing, as well as a conceptually distinct control region. Within the Bayesian framework, we found that GABA increased and glutamate decreased with age, negative associations between anxiety and glutamate and GABA ratios in the dorsolateral prefrontal cortex, and a positive relationship of GABA with anxiety levels. The results support the neural over-inhibition hypothesis of anxiety based on GABAergic activity.
Collapse
Affiliation(s)
- Nicola Johnstone
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK.
| | - Kathrin Cohen Kadosh
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK.
| |
Collapse
|
46
|
Martinez CA, Pantazopoulos H, Gisabella B, Stephens ET, Garteiser J, Del Arco A. Choice impulsivity after repeated social stress is associated with increased perineuronal nets in the medial prefrontal cortex. Sci Rep 2024; 14:7093. [PMID: 38528075 PMCID: PMC10963730 DOI: 10.1038/s41598-024-57599-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
Repeated stress can predispose to substance abuse. However, behavioral and neurobiological adaptations that link stress to substance abuse remain unclear. This study investigates whether intermittent social defeat (ISD), a stress protocol that promotes drug-seeking behavior, alters intertemporal decision-making and cortical inhibitory function in the medial prefrontal cortex (mPFC). Male long evans rats were trained in a delay discounting task (DDT) where rats make a choice between a fast (1 s) small reward (1 sugar pellet) and a large reward (3 sugar pellets) that comes with a time delay (10 s or 20 s). A decreased preference for delayed rewards was used as an index of choice impulsivity. Rats were exposed to ISD and tested in the DDT 24 h after each stress episode, and one- and two-weeks after the last stress episode. Immunohistochemistry was performed in rat's brains to evaluate perineuronal nets (PNNs) and parvalbumin GABA interneurons (PV) labeling as markers of inhibitory function in mPFC. ISD significantly decreased the preference for delayed large rewards in low impulsive, but not high impulsive, animals. ISD also increased the density of PNNs in the mPFC. These results suggest that increased choice impulsivity and cortical inhibition predispose animals to seek out rewards after stress.
Collapse
Affiliation(s)
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Emily T Stephens
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jacob Garteiser
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alberto Del Arco
- HESRM, School of Applied Sciences, University of Mississippi, Oxford, MS, USA.
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
47
|
Li HQ, Jiang W, Ling L, Pratelli M, Chen C, Gupta V, Godavarthi SK, Spitzer NC. Generalized fear after acute stress is caused by change in neuronal cotransmitter identity. Science 2024; 383:1252-1259. [PMID: 38484078 PMCID: PMC11830151 DOI: 10.1126/science.adj5996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/22/2024] [Indexed: 03/19/2024]
Abstract
Overgeneralization of fear to harmless situations is a core feature of anxiety disorders resulting from acute stress, yet the mechanisms by which fear becomes generalized are poorly understood. In this study, we show that generalized fear in mice results from a transmitter switch from glutamate to γ-aminobutyric acid (GABA) in serotonergic neurons of the lateral wings of the dorsal raphe. Similar change in transmitter identity was found in the postmortem brains of individuals with posttraumatic stress disorder (PTSD). Overriding the transmitter switch in mice prevented the acquisition of generalized fear. Corticosterone release and activation of glucocorticoid receptors mediated the switch, and prompt antidepressant treatment blocked the cotransmitter switch and generalized fear. Our results provide important insight into the mechanisms involved in fear generalization.
Collapse
Affiliation(s)
- Hui-quan Li
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Wuji Jiang
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Li Ling
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Marta Pratelli
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Cong Chen
- Department of Cellular and Molecular Medicine, University of California San Diego; La Jolla, California 92093
| | - Vaidehi Gupta
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Swetha K. Godavarthi
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Nicholas C. Spitzer
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| |
Collapse
|
48
|
Cui L, Li S, Wang S, Wu X, Liu Y, Yu W, Wang Y, Tang Y, Xia M, Li B. Major depressive disorder: hypothesis, mechanism, prevention and treatment. Signal Transduct Target Ther 2024; 9:30. [PMID: 38331979 PMCID: PMC10853571 DOI: 10.1038/s41392-024-01738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
Worldwide, the incidence of major depressive disorder (MDD) is increasing annually, resulting in greater economic and social burdens. Moreover, the pathological mechanisms of MDD and the mechanisms underlying the effects of pharmacological treatments for MDD are complex and unclear, and additional diagnostic and therapeutic strategies for MDD still are needed. The currently widely accepted theories of MDD pathogenesis include the neurotransmitter and receptor hypothesis, hypothalamic-pituitary-adrenal (HPA) axis hypothesis, cytokine hypothesis, neuroplasticity hypothesis and systemic influence hypothesis, but these hypothesis cannot completely explain the pathological mechanism of MDD. Even it is still hard to adopt only one hypothesis to completely reveal the pathogenesis of MDD, thus in recent years, great progress has been made in elucidating the roles of multiple organ interactions in the pathogenesis MDD and identifying novel therapeutic approaches and multitarget modulatory strategies, further revealing the disease features of MDD. Furthermore, some newly discovered potential pharmacological targets and newly studied antidepressants have attracted widespread attention, some reagents have even been approved for clinical treatment and some novel therapeutic methods such as phototherapy and acupuncture have been discovered to have effective improvement for the depressive symptoms. In this work, we comprehensively summarize the latest research on the pathogenesis and diagnosis of MDD, preventive approaches and therapeutic medicines, as well as the related clinical trials.
Collapse
Affiliation(s)
- Lulu Cui
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Shu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Siman Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Xiafang Wu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yingyu Liu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yijun Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, China.
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Centre of Forensic Investigation, Shenyang, China.
| |
Collapse
|
49
|
Yin YY, Yan JZ, Lai SX, Wei QQ, Sun SR, Zhang LM, Li YF. Gamma oscillations in the mPFC: A potential predictive biomarker of depression and antidepressant effects. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110893. [PMID: 37949392 DOI: 10.1016/j.pnpbp.2023.110893] [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: 08/21/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Gamma oscillations have attracted much attention in the field of mood disorders, but their role in depression remains poorly understood. This study aimed to investigate whether gamma oscillations in the medial prefrontal cortex (mPFC) could serve as a predictive biomarker of depression. Chronic restraint stress (CRS) or lipopolysaccharide (LPS) were used to induce depression-like behaviors in mice; local field potentials (LFPs) in the mPFC were recorded by electrophysiological techniques; We found that both CRS and LPS induced significant depression-like behaviors in mice, including increasing immobility durations in the forced swimming test (FST) and tail suspension test (TST) and increasing the latency to feed in the novelty-suppressed feeding test (NSFT). Electrophysiological results suggested that CRS and LPS significantly reduced low and high gamma oscillations in the mPFC. Furthermore, a single injection of ketamine or scopolamine for 24 h significantly increased gamma oscillations and elicited rapid-acting antidepressant-like effects. In addition, fluoxetine treatment for 21 days significantly increased gamma oscillations and elicited antidepressant-like effects. Taken together, our findings suggest that gamma oscillations are strongly associated with depression, yielding new insights into investigating the predictive biomarkers of depression and the time course of antidepressant effects.
Collapse
Affiliation(s)
- Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| | - Jiao-Zhao Yan
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shi-Xin Lai
- School of Medicine, Sun Yat-Sen University, Shenzhen campus, Shenzhen, China
| | - Qian-Qian Wei
- School of Medicine, Nantong University, Nantong, China
| | - Si-Rui Sun
- Beijing Ditan Hospital Capital Medical University, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China; Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|
50
|
Page CE, Coutellier L. Kv3.1 Voltage-gated Potassium Channels Modulate Anxiety-like Behaviors in Female Mice. Neuroscience 2024; 538:68-79. [PMID: 38157976 PMCID: PMC10872248 DOI: 10.1016/j.neuroscience.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Inhibitory parvalbumin (PV) interneurons regulate the activity of neural circuits within brain regions involved in emotional processing, including the prefrontal cortex (PFC). Recently, rodent studies have implicated a stress-induced increase in prefrontal PV neuron activity in the development of anxiety behaviors, particularly in females. However, the mechanisms through which stress increases activity of prefrontal PV neurons remain unknown. The fast-spiking properties of PV neurons in part come from their expression of voltage-gated potassium (K+) ion channels, particularly Kv3.1 channels. We therefore suggest that stress-induced changes in Kv3.1 channels contribute to the appearance of an anxious phenotype following chronic stress in female mice. Here, we first showed that unpredictable chronic mild stress (UCMS) increased expression of Kv3.1 channels on prefrontal PV neurons in female mice, a potential mechanism underlying the previously observed hyperactivity of these neurons after stress. We then showed that female mice deficient in Kv3.1 channels displayed resilience to UCMS-induced anxiety-like behaviors. Altogether, our findings implicate Kv3.1 channels in the development of anxiety-like behaviors following UCMS, particularly in females, providing a novel mechanism to understand sex-specific vulnerabilities to stress-induced psychopathologies.
Collapse
Affiliation(s)
- Chloe E Page
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Laurence Coutellier
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States; Department of Psychology, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|