1
|
Ronat LA, Raucher-Chéné D, Lavigne KM, Chakravarty M, Joober R, Malla A, Shah J, Lepage M. Longitudinal clinical outcomes based on cognitive and hippocampal clusters of first episode psychosis. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111392. [PMID: 40320230 DOI: 10.1016/j.pnpbp.2025.111392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND In first episode psychosis (FEP), cognitive impairments are core features contributing to clinical and functional heterogeneity. Significant impairment indicates greater clinical severity throughout the course of the illness, particularly for negative symptoms. Hippocampal volume is smaller in FEP than in healthy controls (notably subfields like Cornu Ammonis 1-3 and subiculum), and is related to cognitive impairments and negative symptoms. The aim of this study was to compare the clinical and functional trajectories of FEP subgroups as a function of cognitive performance and hippocampal volumes. METHODS One hundred FEP patients and sixty healthy controls initially assessed using the CogState research battery, underwent 3 T MRI to extract hippocampal subfields and adjacent structures using the MAGeT brain algorithm. Clinical assessments were carried out for negative (Motivational and Pleasure - MAP, and diminished expression - EXP) and depressive symptoms, and global functioning. Measurements were taken at 4 time points (3, 9, 15, 21 months following program entry). Based on available first timepoint standardized cognitive and hippocampal features, using healthy controls as reference, clusters were determined by a hierarchical ascending classification. Their clinical and functional longitudinal trajectories were analyzed using linear mixed-effects models. RESULTS Three baseline clusters were revealed: normal-range hippocampal volume with low attention, working and verbal memory (FEP 0), small hippocampus with low verbal memory and social cognition (FEP 1), and large hippocampus with low verbal memory (FEP 2). At baseline, the clusters did not differ on symptoms severity and global functioning. Longitudinally, MAP, EXP and depressive symptoms decreased over time in FEP 0. Global functioning improved in FEP 0 and FEP 1, while FEP 2 was clinically and functionally stable over time. Longitudinal inter-group comparisons did not yield any significant differences. CONCLUSION The clusters were dissociated between hippocampus and cognition, but their trajectories suggest the importance of hippocampal integrity in the clinical and/or functional outcome. Future studies are needed to understand intervention efficiency depending on hippocampal integrity.
Collapse
Affiliation(s)
- Lucas A Ronat
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Delphine Raucher-Chéné
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Katie M Lavigne
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - Mallar Chakravarty
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Computional Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Ridha Joober
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, McGill University, Montreal, QC, Canada; Prevention and Early Intervention Program for Psychoses (PEPP-Montreal), Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Ashok Malla
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, McGill University, Montreal, QC, Canada; Prevention and Early Intervention Program for Psychoses (PEPP-Montreal), Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Jai Shah
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Martin Lepage
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Zhang S, Xia J, He W, Zou Y, Liu W, Li L, Huang Z, Li Q, Qi Z, Liu W. From energy metabolism to mood regulation: The rise of lactate as a therapeutic target. J Adv Res 2025:S2090-1232(25)00262-0. [PMID: 40262720 DOI: 10.1016/j.jare.2025.04.018] [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: 12/17/2024] [Revised: 03/28/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Disruption of cerebral energy metabolism is increasingly recognized as a key factor in the pathophysiology of mood disorders. Lactate, beyond its role as a metabolic byproduct, is now understood to be a critical player in brain energy homeostasis and a modulator of neuronal function. Recent advances in understanding lactate shuttling between astrocytes and neurons have opened new avenues for exploring its multifaceted roles in mood regulation. Exercise, known to modulate brain lactate levels, further underscores the potential of lactate as a therapeutic target in mood disorders. AIM OF REVIEW This review delves into the alterations in cerebral lactate associated with mood disorders, emphasizing their implications for brain energy dynamics and signaling pathways. Additionally, we discuss the therapeutic potential of lactate in mood disorders, particularly through its capacity to remodel cerebral function. We conclude by assessing the promise of exercise-induced lactate production as a novel strategy for mood disorder treatment. KEY SCIENTIFIC CONCEPTS OF THE REVIEW Alterations in brain lactate may contribute to the pathogenesis of mood disorders. In several studies, lactate is not only a substrate for brain energy metabolism, but also a molecule that triggers signaling cascades. Specifically, lactate is involved in the regulation of neurogenesis, neuroplasticity, endothelial cell function, and microglia lysosomal acidification, therefore improving mood disorders. Meanwhile, exercise as a low-risk intervention strategy can improve mood disorders through lactate regulation. Thus, the evidence from this review supports that lactate could be a potential therapeutic target for mood disorder, contributing to a deeper understanding of mood disorder pathogenesis and intervention.
Collapse
Affiliation(s)
- Sen Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Jie Xia
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Wenke He
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Yong Zou
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Wenbin Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; School of Physical Education, Shanxi University, Taiyuan, China
| | - Lingxia Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Zhuochun Huang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Qing Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Zhengtang Qi
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China.
| | - Weina Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China.
| |
Collapse
|
3
|
Lee BH, Eid RS, Hodges TE, Barth C, Galea LAM. Leveraging research into sex differences and steroid hormones to improve brain health. Nat Rev Endocrinol 2025; 21:214-229. [PMID: 39587332 DOI: 10.1038/s41574-024-01061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Sex differences, driven in part by steroid hormones, shape the structure and function of the brain throughout the lifespan and manifest across brain health and disease. The influence of steroid hormones on neuroplasticity, particularly in the adult hippocampus, differs between the sexes, which has important implications for disorders and diseases that compromise hippocampus integrity, such as depression and Alzheimer disease. This Review outlines the intricate relationship between steroid hormones and hippocampal neuroplasticity across the adult lifespan and explores how the unique physiology of male and female individuals can affect health and disease. Despite calls to include sex and gender in research, only 5% of neuroscience studies published in 2019 directly investigated the influence of sex. Drawing on insights from depression, Alzheimer disease and relevant hippocampal plasticity, this Review underscores the importance of considering sex and steroid hormones to achieve a comprehensive understanding of disease susceptibility and mechanisms. Such consideration will enable the discovery of personalized treatments, ultimately leading to improved health outcomes for all.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rand S Eid
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Travis E Hodges
- Department of Psychology and Education, Mount Holyoke College, South Hadley, MA, USA
| | - Claudia Barth
- Division for Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Liisa A M Galea
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Elliott T, Liu KY, Hazan J, Wilson J, Vallipuram H, Jones K, Mahmood J, Gitlin-Leigh G, Howard R. Hippocampal neurogenesis in adult primates: a systematic review. Mol Psychiatry 2025; 30:1195-1206. [PMID: 39558003 DOI: 10.1038/s41380-024-02815-y] [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: 01/08/2024] [Revised: 10/11/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024]
Abstract
It had long been considered that no new neurons are generated in the primate brain beyond birth, but recent studies have indicated that neurogenesis persists in various locations throughout the lifespan. The dentate gyrus of the hippocampus is of particular interest due to the postulated role played by neurogenesis in memory. However, studies investigating the presence of adult hippocampal neurogenesis (AHN) have reported contradictory findings, and no systematic review of the evidence has been conducted to date. We searched MEDLINE, Embase and PsycINFO on 27th June 2023 for studies on hippocampal neurogenesis in adult primates, excluding review papers. Screening, quality assessment and data extraction was done by independent co-raters. We synthesised evidence from 112 relevant papers. We found robust evidence, primarily supported by immunohistochemical examination of tissue samples and neuroimaging, for newly generated neurons, first detected in the subgranular zone of the dentate gyrus, that mature over time and migrate to the granule cell layer, where they become functionally integrated with surrounding neuronal networks. AHN has been repeatedly observed in both humans and other primates and gradually diminishes with age. Transient increases in AHN are observed following acute insults such as stroke and epileptic seizures, and following electroconvulsive therapy, and AHN is diminished in neurodegenerative conditions. Markers of AHN correlate positively with measures of learning and short-term memory, but associations with antidepressant use and mood states are weaker. Heterogeneous outcome measures limited quantitative syntheses. Further research should better characterise the neuropsychological function of neurogenesis in healthy subjects.
Collapse
Affiliation(s)
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London, UK
| | - Jemma Hazan
- Division of Psychiatry, University College London, London, UK
- Camden and Islington NHS Foundation Trust, London, UK
| | - Jack Wilson
- Camden and Islington NHS Foundation Trust, London, UK
| | | | | | | | | | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
5
|
Zhang SQ, Deng Q, Tian C, Zhao HH, Yang LY, Cheng XW, Wang GP, Liu D. Costunolide normalizes neuroinflammation and improves neurogenesis deficits in a mouse model of depression through inhibiting microglial Akt/mTOR/NF-κB pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01506-w. [PMID: 40011631 DOI: 10.1038/s41401-025-01506-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/09/2025] [Indexed: 02/28/2025]
Abstract
Neuroinflammation is crucial for the pathogenesis of major depression. Preclinical studies have shown the potential of anti-inflammatory agents, specifically costunolide (COS), correlate with antidepressant effects. In this study, we investigated the molecular mechanisms underlying the antidepressant actions of COS. Chronic restraint stress (CRS) was induced in male mice. The mice were treated with either intra-DG injection of COS (5 μM, 1 μL per side) or COS (20 mg/kg, i.p.) for 1 week. We showed that administration of COS through the both routes significantly ameliorated the depressive-like behavior in CRS-exposed mice. Furthermore, administration of COS significantly improved chronic stress-induced adult hippocampal neurogenesis deficits in the mice through attenuating microglia-derived neuroinflammation. We demonstrated that COS (5 μM) exerted anti-neuroinflammatory effects in LPS-treated BV2 cells via inhibiting microglial Akt/mTOR/NF-κB pathway; inactivation of mTOR/NF-κB/IL-1β pathway was required for the pro-neurogenic action of COS in CRS-exposed mice. Our results reveal the antidepressant mechanism of COS that is normalizing neuroinflammation to improve neurogenesis deficits, supporting anti-inflammatory agents as a potential therapeutic strategy for depression.
Collapse
Affiliation(s)
- Shao-Qi Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiao Deng
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Cheng Tian
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huan-Huan Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221004, China
| | - Li-Ying Yang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin-Wei Cheng
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guo-Ping Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
6
|
Wang N, Zhu S, Chen S, Zou J, Zeng P, Tan S. Neurological mechanism-based analysis of the role and characteristics of physical activity in the improvement of depressive symptoms. Rev Neurosci 2025:revneuro-2024-0147. [PMID: 39829004 DOI: 10.1515/revneuro-2024-0147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/22/2024] [Indexed: 01/22/2025]
Abstract
Depression is a common mental disorder characterized by a high prevalence and significant adverse effects, making the searching for effective interventions an urgent priority. In recent years, physical activity (PA) has increasingly been recognized as a standard adjunctive treatment for mental disorders owing to its low cost, easy application, and high efficiency. Epidemiological data shows positive preventive and therapeutic effects of PA on mental illnesses such as depression. This article systematically describes the prophylactic and therapeutic effects of PA on depression and its biological basis. A comprehensive literature analysis reveals that PA significantly improves depressive symptoms by upregulating the expression of "exerkines" such as irisin, adiponectin, and BDNF to positively impacting neuropsychiatric conditions. In particular, lactate could also play a critical role in the ameliorating effects of PA on depression due to the findings about protein lactylation as a novel protein post-transcriptional modification. The literature also suggests that in terms of brain structure, PA may improve hippocampal volume, basal ganglia (neostriatum, caudate-crustal nucleus) and PFC density in patients with MDD. In summary, this study elucidates the multifaceted positive effects of PA on depression and its potential biological mechanisms with a particular emphasis on the roles of various exerkines. Future research may further investigate the effects of different types, intensities, and durations of PA on depression, as well as how to better integrate PA interventions into existing treatment strategies to achieve optimal outcomes in mental health interventions.
Collapse
Affiliation(s)
- Nan Wang
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Shanshan Zhu
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Shuyang Chen
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Ju Zou
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Peng Zeng
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Sijie Tan
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China
| |
Collapse
|
7
|
Boldrini M, Xiao Y, Singh T, Zhu C, Jabbi M, Pantazopoulos H, Gürsoy G, Martinowich K, Punzi G, Vallender EJ, Zody M, Berretta S, Hyde TM, Kleinman JE, Marenco S, Roussos P, Lewis DA, Turecki G, Lehner T, Mann JJ. Omics Approaches to Investigate the Pathogenesis of Suicide. Biol Psychiatry 2024; 96:919-928. [PMID: 38821194 PMCID: PMC11563882 DOI: 10.1016/j.biopsych.2024.05.017] [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: 01/08/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Suicide is the second leading cause of death in U.S. adolescents and young adults and is generally associated with a psychiatric disorder. Suicidal behavior has a complex etiology and pathogenesis. Moderate heritability suggests genetic causes. Associations between childhood and recent life adversity indicate contributions from epigenetic factors. Genomic contributions to suicide pathogenesis remain largely unknown. This article is based on a workshop held to design strategies to identify molecular drivers of suicide neurobiology that would be putative new treatment targets. The panel determined that while bulk tissue studies provide comprehensive information, single-nucleus approaches that identify cell type-specific changes are needed. While single-nuclei techniques lack information on cytoplasm, processes, spines, and synapses, spatial multiomic technologies on intact tissue detect cell alterations specific to brain tissue layers and subregions. Because suicide has genetic and environmental drivers, multiomic approaches that combine cell type-specific epigenome, transcriptome, and proteome provide a more complete picture of pathogenesis. To determine the direction of effect of suicide risk gene variants on RNA and protein expression and how these interact with epigenetic marks, single-nuclei and spatial multiomics quantitative trait loci maps should be integrated with whole-genome sequencing and genome-wide association databases. The workshop concluded with a recommendation for the formation of an international suicide biology consortium that will bring together brain banks and investigators with expertise in cutting-edge omics technologies to delineate the biology of suicide and identify novel potential treatment targets to be tested in cellular and animal models for drug and biomarker discovery to guide suicide prevention.
Collapse
Affiliation(s)
- Maura Boldrini
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York.
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Tarjinder Singh
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York; New York Genome Center, New York, New York
| | - Chenxu Zhu
- New York Genome Center, New York, New York; Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York
| | - Mbemba Jabbi
- Department of Psychiatry and Behavioral Sciences, Mulva Clinics for the Neurosciences, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | - Gamze Gürsoy
- New York Genome Center, New York, New York; Departments of Biomedical Informatics and Computer Science, Columbia University, New York, New York
| | - Keri Martinowich
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Giovanna Punzi
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Eric J Vallender
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Sabina Berretta
- Department of Psychiatry, Harvard Brain Tissue Resource Center, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Stefano Marenco
- Human Brain Collection Core, National Institute of Mental Health's (NIMH) Division of Intramural Research Programs, Bethesda, Maryland
| | - Panagiotis Roussos
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, New York
| | - David A Lewis
- Departments of Psychiatry and Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Institute, McGill University, Montréal, Québec, Canada
| | | | - J John Mann
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York
| |
Collapse
|
8
|
Llewellyn J, Baratam R, Culig L, Beerman I. Cellular stress and epigenetic regulation in adult stem cells. Life Sci Alliance 2024; 7:e202302083. [PMID: 39348938 PMCID: PMC11443024 DOI: 10.26508/lsa.202302083] [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: 04/07/2023] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Stem cells are a unique class of cells that possess the ability to differentiate and self-renew, enabling them to repair and replenish tissues. To protect and maintain the potential of stem cells, the cells and the environment surrounding these cells (stem cell niche) are highly responsive and tightly regulated. However, various stresses can affect the stem cells and their niches. These stresses are both systemic and cellular and can arise from intrinsic or extrinsic factors which would have strong implications on overall aging and certain disease states. Therefore, understanding the breadth of drivers, namely epigenetic alterations, involved in cellular stress is important for the development of interventions aimed at maintaining healthy stem cells and tissue homeostasis. In this review, we summarize published findings of epigenetic responses to replicative, oxidative, mechanical, and inflammatory stress on various types of adult stem cells.
Collapse
Affiliation(s)
- Joey Llewellyn
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Rithvik Baratam
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Luka Culig
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
9
|
Lu Y, Yu X, Wang Z, Kong L, Jiang Z, Shang R, Zhong X, Lv S, Zhang G, Gao H, Yang N. Microbiota-gut-brain axis: Natural antidepressants molecular mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:156012. [PMID: 39260135 DOI: 10.1016/j.phymed.2024.156012] [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: 05/01/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe mental health condition characterized by persistent depression, impaired cognition, and reduced activity. Increasing evidence suggests that gut microbiota (GM) imbalance is closely linked to the emergence and advancement of MDD, highlighting the potential significance of regulating the "Microbiota-Gut-Brain" (MGB) axis to impact the development of MDD. Natural products (NPs), characterized by broad biological activities, low toxicity, and multi-target characteristics, offer unique advantages in antidepressant treatment by regulating MGB axis. PURPOSE This review was aimed to explore the intricate relationship between the GM and the brain, as well as host responses, and investigated the mechanisms underlying the MGB axis in MDD development. It also explored the pharmacological mechanisms by which NPs modulate MGB axis to exert antidepressant effects and addressed current research limitations. Additionally, it proposed new strategies for future preclinical and clinical applications in the MDD domain. METHODS To study the effects and mechanism by which NPs exert antidepressant effects through mediating the MGB axis, data were collected from Web of Science, PubMed, ScienceDirect from initial establishment to March 2024. NPs were classified and summarized by their mechanisms of action. RESULTS NPs, such as flavonoids,alkaloids,polysaccharides,saponins, terpenoids, can treat MDD by regulating the MGB axis. Its mechanism includes balancing GM, regulating metabolites and neurotransmitters such as SCAFs, 5-HT, BDNF, inhibiting neuroinflammation, improving neural plasticity, and increasing neurogenesis. CONCLUSIONS NPs display good antidepressant effects, and have potential value for clinical application in the prevention and treatment of MDD by regulating the MGB axis. However, in-depth study of the mechanisms by which antidepressant medications affect MGB axis will also require considerable effort in clinical and preclinical research, which is essential for the development of effective antidepressant treatments.
Collapse
Affiliation(s)
- Yitong Lu
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiaowen Yu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Zhongling Wang
- Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Linghui Kong
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhenyuan Jiang
- Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Ruirui Shang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xia Zhong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing 100191, China
| | - Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Haonan Gao
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
10
|
Li P, Zhao J, Wei X, Luo L, Chu Y, Zhang T, Zhu A, Yan J. Acupuncture may play a key role in anti-depression through various mechanisms in depression. Chin Med 2024; 19:135. [PMID: 39367470 PMCID: PMC11451062 DOI: 10.1186/s13020-024-00990-2] [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: 02/22/2024] [Accepted: 08/28/2024] [Indexed: 10/06/2024] Open
Abstract
Depression has emerged as a significant global health concern, exerting a profound impact on individuals, as evidenced by its high prevalence and associated suicide rates. Considering its pervasive nature, the absence of optimal treatment modalities remains a challenge. Acupuncture has garnered substantial clinical and experimental validation for its efficacy in addressing diverse forms of depression, including postpartum, post-stroke, and adolescent depression. This article endeavors to elucidate the distinctive attributes and underlying mechanisms of acupuncture in the contemporary treatment of depression. Research has demonstrated that acupuncture exerts diverse physiological effects in animal models of depression, encompassing modulation of the brain, serum, and brain-gut axis. These effects are attributed to various mechanisms, including anti-inflammatory and anti-oxidative actions, promotion of neuronal plasticity, neuroprotection, neurotrophic effects, modulation of neurotransmitters, regulation of endocrine and immune functions, and modulation of cell signal pathways. Currently, the therapeutic mechanism of acupuncture involves the engagement of multiple targets, pathways, and bidirectional regulation. Hence, acupuncture emerges as a promising alternative medical modality, exhibiting substantial research prospects and meriting comprehensive worth further study and dissemination.
Collapse
Affiliation(s)
- Peng Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Department of clinical medicine, Xiamen medical college, xiamen, China
| | - Jiangna Zhao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiuxiang Wei
- Rehabilitation Medicine Department, Shenzhen Hospital of Traditional Chinese and Western Medicine , Shenzhen, China
| | - Longfei Luo
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yuzhou Chu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Tao Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Anning Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Juntao Yan
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
11
|
Ma Y, Qiao Y, Gao X. Potential role of hippocampal neurogenesis in spinal cord injury induced post-trauma depression. Neural Regen Res 2024; 19:2144-2156. [PMID: 38488549 PMCID: PMC11034606 DOI: 10.4103/1673-5374.392855] [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: 05/11/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 04/24/2024] Open
Abstract
It has been reported both in clinic and rodent models that beyond spinal cord injury directly induced symptoms, such as paralysis, neuropathic pain, bladder/bowel dysfunction, and loss of sexual function, there are a variety of secondary complications, including memory loss, cognitive decline, depression, and Alzheimer's disease. The large-scale longitudinal population-based studies indicate that post-trauma depression is highly prevalent in spinal cord injury patients. Yet, few basic studies have been conducted to address the potential molecular mechanisms. One of possible factors underlying the depression is the reduction of adult hippocampal neurogenesis which may come from less physical activity, social isolation, chronic pain, and elevated neuroinflammation after spinal cord injury. However, there is no clear consensus yet. In this review, we will first summarize the alteration of hippocampal neurogenesis post-spinal cord injury. Then, we will discuss possible mechanisms underlie this important spinal cord injury consequence. Finally, we will outline the potential therapeutic options aimed at enhancing hippocampal neurogenesis to ameliorate depression.
Collapse
Affiliation(s)
- Ying Ma
- Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yue Qiao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiang Gao
- Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
12
|
Alonso M, Petit AC, Lledo PM. The impact of adult neurogenesis on affective functions: of mice and men. Mol Psychiatry 2024; 29:2527-2542. [PMID: 38499657 DOI: 10.1038/s41380-024-02504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
In most mammals, new neurons are not only produced during embryogenesis but also after birth. Soon after adult neurogenesis was discovered, the influence of recruiting new neurons on cognitive functions, especially on memory, was documented. Likewise, the late process of neuronal production also contributes to affective functions, but this outcome was recognized with more difficulty. This review covers hypes and hopes of discovering the influence of newly-generated neurons on brain circuits devoted to affective functions. If the possibility of integrating new neurons into the adult brain is a commonly accepted faculty in the realm of mammals, the reluctance is strong when it comes to translating this concept to humans. Compiling data suggest now that new neurons are derived not only from stem cells, but also from a population of neuroblasts displaying a protracted maturation and ready to be engaged in adult brain circuits, under specific signals. Here, we discuss the significance of recruiting new neurons in the adult brain circuits, specifically in the context of affective outcomes. We also discuss the fact that adult neurogenesis could be the ultimate cellular process that integrates elements from both the internal and external environment to adjust brain functions. While we must be critical and beware of the unreal promises that Science could generate sometimes, it is important to continue exploring the potential of neural recruitment in adult primates. Reporting adult neurogenesis in humankind contributes to a new vision of humans as mammals whose brain continues to develop throughout life. This peculiar faculty could one day become the target of treatment for mental health, cognitive disorders, and elderly-associated diseases. The vision of an adult brain which never stops integrating new neurons is a real game changer for designing new therapeutic interventions to treat mental disorders associated with substantial morbidity, mortality, and social costs.
Collapse
Affiliation(s)
- Mariana Alonso
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
| | - Anne-Cécile Petit
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
- Pôle Hospitalo-Universitaire Psychiatrie Paris 15, GHU Paris Psychiatry and Neurosciences, Hôpital Sainte-Anne, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France.
| |
Collapse
|
13
|
Liu Y, Zhang J, Gu X, Jia S. Mapping the current trends and hotspots of adult hippocampal neurogenesis from 2004-2023: a bibliometric analysis. Front Neurosci 2024; 18:1416738. [PMID: 38957185 PMCID: PMC11217541 DOI: 10.3389/fnins.2024.1416738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024] Open
Abstract
Objective We utilized bibliometric and data visualization techniques to discern the primary research domains and emerging frontiers in the field of adult hippocampal neurogenesis (AHN). Methods We systematically searched the Web of Science database for AHN-related articles published between 2004 and 2023. The retrieved articles were filtered based on publication types (articles and reviews) and language (English). We employed CiteSpace, VOSviewer, and the online bibliometric platform (bibliometric.com) to visualize and analyze the collected data. Results In total, 1,590 AHN-related publications were discovered, exhibiting a steady increase in yearly publications over time. The United States emerged as the leading contributor in AHN research in terms of both publication quantity and national influence. Among all research institutions in the field of AHN, the University of California System exhibited the highest impact. Kempermann, Gerd was the most active author. The publications of the top three active authors primarily focused on the functions of AHN, and reversing hippocampal damage and cognitive impairment by improving AHN. An analysis of reference co-citation clustering revealed 8 distinct research clusters, and the notable ones included "adult hippocampal neurogenesis," "neurogenesis," "hippocampus," "dentate gyrus," "neural stem cell," and "depression." Additionally, a burst keyword detection indicated that 'anxiety' is a current research hotspot in the field of AHN. Conclusion This in-depth bibliographic assessment of AHN offers a deeper insight into the present research hotspots in the field. The association between AHN and cognitive diseases, such as Alzheimer's disease (AD) and anxiety, has emerged as a prominent research hotspot.
Collapse
Affiliation(s)
- Ye Liu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
- Department of Anesthesiology, Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Jian Zhang
- Department of Anesthesiology, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiyao Gu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
- Department of Anesthesiology, Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shushan Jia
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| |
Collapse
|
14
|
Thibaudeau A, Schmitt K, François L, Chatrousse L, Hoffmann D, Cousin L, Weiss A, Vuidel A, Jacob CB, Sommer P, Benchoua A, Wilbertz JH. Pharmacological modulation of developmental and synaptic phenotypes in human SHANK3 deficient stem cell-derived neuronal models. Transl Psychiatry 2024; 14:249. [PMID: 38858349 PMCID: PMC11165012 DOI: 10.1038/s41398-024-02947-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 06/12/2024] Open
Abstract
Phelan-McDermid syndrome (PMDS) arises from mutations in the terminal region of chromosome 22q13, impacting the SHANK3 gene. The resulting deficiency of the postsynaptic density scaffolding protein SHANK3 is associated with autism spectrum disorder (ASD). We examined 12 different PMDS patient and CRISPR-engineered stem cell-derived neuronal models and controls and found that reduced expression of SHANK3 leads to neuronal hyperdifferentiation, increased synapse formation, and decreased neuronal activity. We performed automated imaging-based screening of 7,120 target-annotated small molecules and identified three compounds that rescued SHANK3-dependent neuronal hyperdifferentiation. One compound, Benproperine, rescued the decreased colocalization of Actin Related Protein 2/3 Complex Subunit 2 (ARPC2) with ß-actin and rescued increased synapse formation in SHANK3 deficient neurons when administered early during differentiation. Neuronal activity was only mildly affected, highlighting Benproperine's effects as a neurodevelopmental modulator. This study demonstrates that small molecular compounds that reverse developmental phenotypes can be identified in human neuronal PMDS models.
Collapse
|
15
|
Haniff ZR, Bocharova M, Mantingh T, Rucker JJ, Velayudhan L, Taylor DM, Young AH, Aarsland D, Vernon AC, Thuret S. Psilocybin for dementia prevention? The potential role of psilocybin to alter mechanisms associated with major depression and neurodegenerative diseases. Pharmacol Ther 2024; 258:108641. [PMID: 38583670 PMCID: PMC11847495 DOI: 10.1016/j.pharmthera.2024.108641] [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: 09/05/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Major depression is an established risk factor for subsequent dementia, and depression in late life may also represent a prodromal state of dementia. Considering current challenges in the clinical development of disease modifying therapies for dementia, the focus of research is shifting towards prevention and modification of risk factors to alter the neurodegenerative disease trajectory. Understanding mechanistic commonalities underlying affective symptoms and cognitive decline may reveal biomarkers to aid early identification of those at risk of progressing to dementia during the preclinical phase of disease, thus allowing for timely intervention. Adult hippocampal neurogenesis (AHN) is a phenomenon that describes the birth of new neurons in the dentate gyrus throughout life and it is associated with spatial learning, memory and mood regulation. Microglia are innate immune system macrophages in the central nervous system that carefully regulate AHN via multiple mechanisms. Disruption in AHN is associated with both dementia and major depression and microgliosis is a hallmark of several neurodegenerative diseases. Emerging evidence suggests that psychedelics promote neuroplasticity, including neurogenesis, and may also be immunomodulatory. In this context, psilocybin, a serotonergic agonist with rapid-acting antidepressant properties has the potential to ameliorate intersecting pathophysiological processes relevant for both major depression and neurodegenerative diseases. In this narrative review, we focus on the evidence base for the effects of psilocybin on adult hippocampal neurogenesis and microglial form and function; which may suggest that psilocybin has the potential to modulate multiple mechanisms of action, and may have implications in altering the progression from major depression to dementia in those at risk.
Collapse
Affiliation(s)
- Zarah R Haniff
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - Mariia Bocharova
- Department of Old Age Psychiatry, Division of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - Tim Mantingh
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - James J Rucker
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, Denmark Hill, London, United Kingdom
| | - Latha Velayudhan
- Department of Old Age Psychiatry, Division of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - David M Taylor
- South London and Maudsley NHS Foundation Trust, Maudsley Hospital, Denmark Hill, London, United Kingdom
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, United Kingdom
| | - Dag Aarsland
- Department of Old Age Psychiatry, Division of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Wolfson Centre for Age Related Diseases, Division of Neuroscience of the Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Stavanger University Hospital, Stavanger, Norway
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, United Kingdom.
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| |
Collapse
|
16
|
Caruso MG, Nicolas S, Lucassen PJ, Mul JD, O’Leary OF, Nolan YM. Ageing, Cognitive Decline, and Effects of Physical Exercise: Complexities, and Considerations from Animal Models. Brain Plast 2024; 9:43-73. [PMID: 38993577 PMCID: PMC11234681 DOI: 10.3233/bpl-230157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 07/13/2024] Open
Abstract
In our ageing global population, the cognitive decline associated with dementia and neurodegenerative diseases represents a major healthcare problem. To date, there are no effective treatments for age-related cognitive impairment, thus preventative strategies are urgently required. Physical exercise is gaining traction as a non-pharmacological approach to promote brain health. Adult hippocampal neurogenesis (AHN), a unique form of brain plasticity which is necessary for certain cognitive functions declines with age and is enhanced in response to exercise. Accumulating evidence from research in rodents suggests that physical exercise has beneficial effects on cognition through its proneurogenic capabilities. Given ethical and technical limitations in human studies, preclinical research in rodents is crucial for a better understanding of such exercise-induced brain and behavioural changes. In this review, exercise paradigms used in preclinical research are compared. We provide an overview of the effects of different exercise paradigms on age-related cognitive decline from middle-age until older-age. We discuss the relationship between the age-related decrease in AHN and the potential impact of exercise on mitigating this decline. We highlight the emerging literature on the impact of exercise on gut microbiota during ageing and consider the role of the gut-brain axis as a future possible strategy to optimize exercise-enhanced cognitive function. Finally, we propose a guideline for designing optimal exercise protocols in rodent studies, which would inform clinical research and contribute to developing preventative strategies for age-related cognitive decline.
Collapse
Affiliation(s)
- Maria Giovanna Caruso
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Sarah Nicolas
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Paul J. Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Joram D. Mul
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Olivia F. O’Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Yvonne M. Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| |
Collapse
|
17
|
Beltran-Casanueva R, Hernández-García A, Serrano-Castro PJ, Sánchez-Pérez JA, Barbancho-Fernández MA, García-Casares N, Fuxe K, Borroto-Escuela DO, Narváez M. Long-term enhancements in antidepressant efficacy and neurogenesis: Effects of intranasal co-administration of neuropeptide Y 1 receptor (NPY1R) and galanin receptor 2 (GALR2) agonists in the ventral hippocampus. FASEB J 2024; 38:e23595. [PMID: 38572811 DOI: 10.1096/fj.202400087r] [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: 01/13/2024] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
This study evaluates the sustained antidepressant-like effects and neurogenic potential of a 3-day intranasal co-administration regimen of galanin receptor 2 (GALR2) agonist M1145 and neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31, Pro34]NPY in the ventral hippocampus of adult rats, with outcomes analyzed 3 weeks post-treatment. Utilizing the forced swimming test (FST), we found that this co-administration significantly enhances antidepressant-like behaviors, an effect neutralized by the GALR2 antagonist M871, highlighting the synergistic potential of these neuropeptides in modulating mood-related behaviors. In situ proximity ligation assay (PLA) indicated a significant increase in GALR2/NPYY1R heteroreceptor complexes in the ventral hippocampal dentate gyrus, suggesting a molecular basis for the behavioral outcomes observed. Moreover, proliferating cell nuclear antigen (PCNA) immunolabeling revealed increased cell proliferation in the subgranular zone of the dentate gyrus, specifically in neuroblasts as evidenced by co-labeling with doublecortin (DCX), without affecting quiescent neural progenitors or astrocytes. The study also noted a significant uptick in the number of DCX-positive cells and alterations in dendritic morphology in the ventral hippocampus, indicative of enhanced neuronal differentiation and maturation. These morphological changes highlight the potential of these agonists to facilitate the functional integration of new neurons into existing neural circuits. By demonstrating the long-lasting effects of a brief, 3-day intranasal administration of GALR2 and NPY1R agonists, our findings contribute significantly to the understanding of neuropeptide-mediated neuroplasticity and herald novel therapeutic strategies for the treatment of depression and related mood disorders, emphasizing the therapeutic promise of targeting neurogenesis and neuronal maturation processes.
Collapse
Affiliation(s)
- Rasiel Beltran-Casanueva
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Aracelis Hernández-García
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Departamento de Docencia e Investigación, Universidad de Ciencias Médicas de Holguín, Hospital Pedíatrico Universitario Octavio de la Concepción de la Pedraja, Holguín, Cuba
| | - Pedro Jesús Serrano-Castro
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| | - Jose Andrés Sánchez-Pérez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Psychiatry, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | | | - Natalia García-Casares
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Manuel Narváez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| |
Collapse
|
18
|
Borroto-Escuela D, Serrano-Castro P, Sánchez-Pérez JA, Barbancho-Fernández MA, Fuxe K, Narváez M. Enhanced neuronal survival and BDNF elevation via long-term co-activation of galanin 2 (GALR2) and neuropeptide Y1 receptors (NPY1R): potential therapeutic targets for major depressive disorder. Expert Opin Ther Targets 2024; 28:295-308. [PMID: 38622072 DOI: 10.1080/14728222.2024.2342517] [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: 03/01/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Major Depressive Disorder (MDD) is a prevalent and debilitating condition, necessitating novel therapeutic strategies due to the limited efficacy and adverse effects of current treatments. We explored how galanin receptor 2 (GALR2) and Neuropeptide Y1 Receptor (NPYY1R) agonists, working together, can boost brain cell growth and increase antidepressant-like effects in rats. This suggests new ways to treat Major Depressive Disorder (MDD). RESEARCH DESIGN AND METHODS In a controlled laboratory setting, adult naive Sprague-Dawley rats were administered directly into the brain's ventricles, a method known as intracerebroventricular (ICV) administration, with GALR2 agonist (M1145), NPYY1R agonist, both, or in combination with a GALR2 antagonist (M871). Main outcome measures included long-term neuronal survival, differentiation, and behavioral. RESULTS Co-administration of M1145 and NPYY1R agonist significantly enhanced neuronal survival and maturation in the ventral dentate gyrus, with a notable increase in Brain-Derived Neurotrophic Factor (BDNF) expression. This neurogenic effect was associated with an antidepressant-like effect, an outcome partially reversed by M871. CONCLUSIONS GALR2 and NPYY1R agonists jointly promote hippocampal neurogenesis and exert antidepressant-like effects in rats without adverse outcomes, highlighting their therapeutic potential for MDD. The study's reliance on an animal model and intracerebroventricular delivery warrants further clinical exploration to confirm these promising results.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Antidepressive Agents/pharmacology
- Antidepressive Agents/administration & dosage
- Brain-Derived Neurotrophic Factor/metabolism
- Cell Survival/drug effects
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Major/physiopathology
- Disease Models, Animal
- Neurons/drug effects
- Neurons/metabolism
- Peptides
- Rats, Sprague-Dawley
- Receptor, Galanin, Type 2/metabolism
- Receptors, G-Protein-Coupled
- Receptors, Neuropeptide
- Receptors, Neuropeptide Y/metabolism
- Receptors, Neuropeptide Y/antagonists & inhibitors
Collapse
Affiliation(s)
- Dasiel Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Pedro Serrano-Castro
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| | - Jose Andrés Sánchez-Pérez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Psychiatry, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | | | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Narváez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| |
Collapse
|
19
|
Meda N, Miola A, Cattarinussi G, Sambataro F. Whole-brain structural and functional neuroimaging of individuals who attempted suicide and people who did not: A systematic review and exploratory coordinate-based meta-analysis. Eur Neuropsychopharmacol 2024; 79:66-77. [PMID: 38237538 DOI: 10.1016/j.euroneuro.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 02/06/2024]
Abstract
Suicide is the cause of death of approximately 800,000 people a year. Despite the relevance of this behaviour, risk assessment tools rely on clinician experience and subjective ratings. Given that previous suicide attempts are the single strongest predictors of future attempts, we designed a systematic review and coordinate-based meta-analysis to demonstrate whether neuroimaging features can help distinguish individuals who attempted suicide from subjects who did not. Out of 5,659 publications from PubMed, Scopus, and Web of Science, we summarised 102 experiments and meta-analysed 23 of them. A cluster in the right superior temporal gyrus, a region implicated in emotional processing, might be functionally hyperactive in individuals who attempted suicide. No statistically significant differences in brain morphometry were evidenced. Furthermore, we used JuSpace to show that this cluster is enriched in 5-HT1A heteroreceptors in the general population. This exploratory meta-analysis provides a putative neural substrate linked to previous suicide attempts. Heterogeneity in the analytical techniques and weak or absent power analysis of the studies included in this review currently limit the applicability of the findings, the replication of which should be prioritised.
Collapse
Affiliation(s)
- Nicola Meda
- Department of Neuroscience, University of Padova, Via Giustiniani, 3, Padua, Italy; Padova University Hospital, Padua, Italy
| | - Alessandro Miola
- Department of Neuroscience, University of Padova, Via Giustiniani, 3, Padua, Italy; Padova Neuroscience Center, University of Padova, Padua, Italy; Casa di Cura Parco dei Tigli, Padova, Italy
| | - Giulia Cattarinussi
- Department of Neuroscience, University of Padova, Via Giustiniani, 3, Padua, Italy; Padova Neuroscience Center, University of Padova, Padua, Italy
| | - Fabio Sambataro
- Department of Neuroscience, University of Padova, Via Giustiniani, 3, Padua, Italy; Padova University Hospital, Padua, Italy; Padova Neuroscience Center, University of Padova, Padua, Italy.
| |
Collapse
|
20
|
Dong Y, Lu Z, Gao T, Wei Z, Ou Z, Shi Z, Shen J. A polypeptide derived from pilose antler ameliorates CUMS-induced depression-like behavior by SENP2-PLCβ4 signaling axis. Eur J Pharmacol 2024; 963:176247. [PMID: 38056617 DOI: 10.1016/j.ejphar.2023.176247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Neurogenesis is known to be closely associated with depression. We aimed to investigate whether a polypeptide monomer derived from pilose antler (polypeptide sequence LSALEGVFYP, PAP) exerts an antidepressant effect by influencing neurogenesis, and to elucidate the mechanism of its antidepressant action. Behavioral tests were performed to observe the antidepressant effect of PAP. Neurogenesis in the dentate gyrus (DG) region of hippocampus was observed by immunofluorescence. The expression of key proteins of Sentrin/SUMO-specific proteases 2 (SENP2)- Phosphoinositide-specific phospholipase C beta 4 (PLCβ4) pathway was accessed by co-immunoprecipitation (Co-IP), and the calcium homeostasis associated proteins were observed via Western blot (WB). Subsequently, temozolomide (TMZ) pharmacologically blocked neurogenesis to verify the antidepressant effect of PAP on neurogenesis. The mechanism of PAP antidepressant effect was verified by constructing a sh-SENP2 virus vector to silence SENP2 protein. Finally, corticosterone (CORT)-induced PC12 cell model was used to verify whether PAP was involved in the process of deconjugated PLCβ4 SUMOylated. The results showed that PAP improved depression-like behavior and neurogenesis induced by chronic unpredictable mild stimulation (CUMS). In addition, PAP acted on SENP2-PLCβ4 pathway to deconjugate the SUMOylation of PLCβ4 and affect calcium homeostasis. Pharmacological blockade of neurogenesis by TMZ treatment impaired the antidepressant efficacy of PAP. Knockout of SENP2 in the CUMS model attenuated the antidepressant response of PAP, and the impaired neurogenesis was not ameliorated by PAP treatment. In summary, PAP acted on the SENP2-PLCβ4 signaling pathway to inhibit the SUMOylation of PLCβ4 and maintain calcium homeostasis, thereby protecting neurogenesis and playing an antidepressant role.
Collapse
Affiliation(s)
- Yu Dong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zihan Lu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, 210009, China
| | - Tiantian Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhijie Ou
- Neurology Department, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, 215500, China.
| | - Zheng Shi
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jie Shen
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
21
|
Valeri J, Stiplosek C, O’Donovan SM, Sinclair D, Grant K, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular Matrix Abnormalities in the Hippocampus of Subjects with Substance Use Disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.09.07.23295222. [PMID: 37732207 PMCID: PMC10508799 DOI: 10.1101/2023.09.07.23295222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH
| | | | - Ratna Bollavarapu
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Donna M. Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
22
|
Davydova YD, Kazantseva AV, Khusnutdinova EK. [A perspective on the application of CRISPR/CAS9 genome editing system to study of molecular-genetic basis of mental disorders]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:27-33. [PMID: 38529860 DOI: 10.17116/jnevro202412403127] [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: 03/27/2024]
Abstract
Schizophrenia, depression, bipolar disorder and autism spectrum disorders are common mental disorders that are among the leading causes of disability worldwide. The major complication to effective therapies for mental disorders is the poor understanding of their pathogenic mechanisms. Currently, an increasing number of research groups are focusing on uncovering the molecular mechanisms of mental disorders and developing novel therapies using the CRISPR/Cas9 (Clustered, Regularly Interspaced, Short Palindromic Repeats (CRISPR) - CRISPR-associated system 9 (Cas9)) system to determine the molecular mechanisms of developing mental disorders and novel therapy. The CRISPR/Cas9 system is the most promising among genome editing tools. Numerous advantages of the CRISPR/Cas9 system and its successful application in some studies provide wide opportunities for genome therapy and regeneration medicine. In this review we shortly describe structure and function of the CRISPR/Cas9 system and its application to study the molecular-genetic basis of mental disorders in human.
Collapse
Affiliation(s)
- Yu D Davydova
- Institute of Biochemistry and Genetics - Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
- Ufa University of Science and Technology, Ufa, Russia
| | - A V Kazantseva
- Institute of Biochemistry and Genetics - Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
- Ufa State Petroleum Technical University, Ufa, Russia
| | - E K Khusnutdinova
- Institute of Biochemistry and Genetics - Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
- Ufa University of Science and Technology, Ufa, Russia
| |
Collapse
|
23
|
Ben-Zion Z, Korem N, Fine NB, Katz S, Siddhanta M, Funaro MC, Duek O, Spiller TR, Danböck SK, Levy I, Harpaz-Rotem I. Structural Neuroimaging of Hippocampus and Amygdala Subregions in Posttraumatic Stress Disorder: A Scoping Review. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:120-134. [PMID: 38298789 PMCID: PMC10829655 DOI: 10.1016/j.bpsgos.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 02/02/2024] Open
Abstract
Numerous studies have explored the relationship between posttraumatic stress disorder (PTSD) and the hippocampus and the amygdala because both regions are implicated in the disorder's pathogenesis and pathophysiology. Nevertheless, those key limbic regions consist of functionally and cytoarchitecturally distinct substructures that may play different roles in the etiology of PTSD. Spurred by the availability of automatic segmentation software, structural neuroimaging studies of human hippocampal and amygdala subregions have proliferated in recent years. Here, we present a preregistered scoping review of the existing structural neuroimaging studies of the hippocampus and amygdala subregions in adults diagnosed with PTSD. A total of 3513 studies assessing subregion volumes were identified, 1689 of which were screened, and 21 studies were eligible for this review (total N = 2876 individuals). Most studies examined hippocampal subregions and reported decreased CA1, CA3, dentate gyrus, and subiculum volumes in PTSD. Fewer studies investigated amygdala subregions and reported altered lateral, basal, and central nuclei volumes in PTSD. This review further highlights the conceptual and methodological limitations of the current literature and identifies future directions to increase understanding of the distinct roles of hippocampal and amygdalar subregions in posttraumatic psychopathology.
Collapse
Affiliation(s)
- Ziv Ben-Zion
- Yale School of Medicine, Yale University, New Haven, Connecticut
- US Department of Veterans Affairs National Center for PTSD, Clinical Neuroscience Division, VA Connecticut Healthcare System, West Haven, Connecticut
- Wu Tsai Institute, Yale University, New Haven, Connecticut
- Department of Psychology, Yale University, New Haven, Connecticut
| | - Nachshon Korem
- Yale School of Medicine, Yale University, New Haven, Connecticut
- US Department of Veterans Affairs National Center for PTSD, Clinical Neuroscience Division, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Naomi B. Fine
- Sagol Brain Institute Tel-Aviv, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Social Sciences, School of Psychological Science, Tel Aviv University, Tel Aviv, Israel
| | - Sophia Katz
- Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Megha Siddhanta
- Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Melissa C. Funaro
- Harvey Cushing/John Hay Whitney Medical Library, Yale University, New Haven, Connecticut
| | - Or Duek
- Yale School of Medicine, Yale University, New Haven, Connecticut
- US Department of Veterans Affairs National Center for PTSD, Clinical Neuroscience Division, VA Connecticut Healthcare System, West Haven, Connecticut
- Department of Epidemiology, Biostatistics and Community Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tobias R. Spiller
- Yale School of Medicine, Yale University, New Haven, Connecticut
- US Department of Veterans Affairs National Center for PTSD, Clinical Neuroscience Division, VA Connecticut Healthcare System, West Haven, Connecticut
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Sarah K. Danböck
- Yale School of Medicine, Yale University, New Haven, Connecticut
- Division of Clinical Psychology and Psychopathology, Department of Psychology, Paris London University of Salzburg, Salzburg, Austria
| | - Ifat Levy
- Yale School of Medicine, Yale University, New Haven, Connecticut
- Wu Tsai Institute, Yale University, New Haven, Connecticut
- Department of Psychology, Yale University, New Haven, Connecticut
| | - Ilan Harpaz-Rotem
- Yale School of Medicine, Yale University, New Haven, Connecticut
- US Department of Veterans Affairs National Center for PTSD, Clinical Neuroscience Division, VA Connecticut Healthcare System, West Haven, Connecticut
- Wu Tsai Institute, Yale University, New Haven, Connecticut
- Department of Psychology, Yale University, New Haven, Connecticut
| |
Collapse
|
24
|
Matar D, Serhan A, El Bilani S, Faraj RA, Hadi BA, Fakhoury M. Psychopharmacological Approaches for Neural Plasticity and Neurogenesis in Major Depressive Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1456:27-48. [PMID: 39261422 DOI: 10.1007/978-981-97-4402-2_2] [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: 09/13/2024]
Abstract
Major depressive disorder (MDD) is a mental health disorder associated with cognitive impairment, dysregulated appetite, fatigue, insomnia or hypersomnia, and severe mood changes that significantly impact the ability of the affected individual to perform day-to-day tasks, leading to suicide in the worst-case scenario. As MDD is becoming more prevalent, affecting roughly 300 million individuals worldwide, its treatment has become a major point of interest. Antidepressants acting as selective serotonin reuptake inhibitors (SSRIs) are currently used as the first line of treatment for MDD. Other antidepressants currently used for the treatment of MDD include the serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs). However, although effective in alleviating symptoms of MDD, most antidepressants require weeks or even months of regular administration prior to eliciting a rational clinical effect. Owing to the strong evidence showing a relationship between neural plasticity, neurogenesis, and MDD, researchers have also looked at the possibility of using treatment modalities that target these processes in an attempt to improve clinical outcome. The overarching aim of this chapter is to highlight the role of neural plasticity and neurogenesis in the pathophysiology of MDD and discuss the most recently studied treatment strategies that target these processes by presenting supporting evidence from both animal and human studies.
Collapse
Affiliation(s)
- Dina Matar
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Aya Serhan
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Sabah El Bilani
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Rashel Abi Faraj
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Bayan Ali Hadi
- School of Pharmacy, Lebanese American University, Beirut, Lebanon
| | - Marc Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
25
|
Barth C, Nerland S, Jørgensen KN, Haatveit B, Wortinger LA, Melle I, Haukvik UK, Ueland T, Andreassen OA, Agartz I. Altered Sex Differences in Hippocampal Subfield Volumes in Schizophrenia. Schizophr Bull 2024; 50:107-119. [PMID: 37354490 PMCID: PMC10754184 DOI: 10.1093/schbul/sbad091] [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] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND HYPOTHESIS The hippocampus is a heterogenous brain structure that differs between the sexes and has been implicated in the pathophysiology of psychiatric illnesses. Here, we explored sex and diagnostic group differences in hippocampal subfield volumes, in individuals with schizophrenia spectrum disorder (SZ), bipolar disorders (BD), and healthy controls (CTL). STUDY DESIGN One thousand and five hundred and twenty-one participants underwent T1-weighted magnetic resonance imaging (SZ, n = 452, mean age 30.7 ± 9.2 [SD] years, males 59.1%; BD, n = 316, 33.7 ± 11.4, 41.5%; CTL, n = 753, 34.1 ± 9.1, 55.6%). Total hippocampal, subfield, and intracranial volumes were estimated with Freesurfer (v6.0.0). Analysis of covariance and multiple regression models were fitted to examine sex-by-diagnostic (sub)group interactions in volume. In SZ and BD, separately, associations between volumes and clinical as well as cognitive measures were examined between the sexes using regression models. STUDY RESULTS Significant sex-by-group interactions were found for the total hippocampus, dentate gyrus, molecular layer, presubiculum, fimbria, hippocampal-amygdaloid transition area, and CA4, indicating a larger volumetric deficit in male patients relative to female patients when compared with same-sex CTL. Subgroup analyses revealed that this interaction was driven by males with schizophrenia. Effect sizes were overall small (partial η < 0.02). We found no significant sex differences in the associations between hippocampal volumes and clinical or cognitive measures in SZ and BD. CONCLUSIONS Using a well-powered sample, our findings indicate that the pattern of morphological sex differences in hippocampal subfields is altered in individuals with schizophrenia relative to CTL, due to higher volumetric deficits in males.
Collapse
Affiliation(s)
- Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
| | - Stener Nerland
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
| | - Kjetil N Jørgensen
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Department of Psychiatry, Telemark Hospital, Skien, Norway
| | - Beathe Haatveit
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
| | - Laura A Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
| | - Ingrid Melle
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
| | - Unn K Haukvik
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
- Department of Adult Mental Health, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Torill Ueland
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, NORMENT, Oslo, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, NORMENT, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
26
|
Chang WL, Hen R. Adult Neurogenesis, Context Encoding, and Pattern Separation: A Pathway for Treating Overgeneralization. ADVANCES IN NEUROBIOLOGY 2024; 38:163-193. [PMID: 39008016 DOI: 10.1007/978-3-031-62983-9_10] [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: 07/16/2024]
Abstract
In mammals, the subgranular zone of the dentate gyrus is one of two brain regions (with the subventricular zone of the olfactory bulb) that continues to generate new neurons throughout adulthood, a phenomenon known as adult hippocampal neurogenesis (AHN) (Eriksson et al., Nat Med 4:1313-1317, 1998; García-Verdugo et al., J Neurobiol 36:234-248, 1998). The integration of these new neurons into the dentate gyrus (DG) has implications for memory encoding, with unique firing and wiring properties of immature neurons that affect how the hippocampal network encodes and stores attributes of memory. In this chapter, we will describe the process of AHN and properties of adult-born cells as they integrate into the hippocampal circuit and mature. Then, we will discuss some methodological considerations before we review evidence for the role of AHN in two major processes supporting memory that are performed by the DG. First, we will discuss encoding of contextual information for episodic memories and how this is facilitated by AHN. Second, will discuss pattern separation, a major role of the DG that reduces interference for the formation of new memories. Finally, we will review clinical and translational considerations, suggesting that stimulation of AHN may help decrease overgeneralization-a common endophenotype of mood, anxiety, trauma-related, and age-related disorders.
Collapse
Affiliation(s)
- Wei-Li Chang
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Rene Hen
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA.
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
27
|
Guilloux JP, Nguyen TML, Gardier AM. [Ketamine: a neuropsychotropic drug with an innovative mechanism of action]. Biol Aujourdhui 2023; 217:133-144. [PMID: 38018940 DOI: 10.1051/jbio/2023026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Indexed: 11/30/2023]
Abstract
Ketamine, a non-competitive antagonist of the N-methyl-D-aspartate-glutamate receptor (R-NMDA), has a rapid (from 24 h post-dose) and prolonged (up to one week) antidepressant effect in treatment resistant depression and in rodent models of anxiety/depression. Arguments regarding its cellular and molecular mechanisms underlying its antidepressant activity mainly come from animal studies. However, debates still persist on the structural remodeling of frontocortical/hippocampal neurons and the role of excitatory/inhibitory neurotransmitters involved in its behavioral effect. Neurochemical and behavioral changes are maintained 24 h after administration of ketamine, well beyond its plasma elimination half-life. The glutamatergic pyramidal cells of the medial prefrontal cortex are primarily implicated in the therapeutic effects of ketamine. Advances in knowledge of the consequences of R-NMDA blockade allowed to specify the underlying mechanisms involving the activation of AMPA glutamate receptors, which triggers a cascade of intracellular events dependent on the mechanistic target of rapamycin, brain-derived neurotrophic factor, and synaptic protein synthesis facilitating synaptic plasticity (number of dendritic spines, synaptogenesis). This review focuses on abnormalities of neurotransmitter systems involved in major depressive disorders, their potential impact on neural circuitry and beneficial effects of ketamine. Recent preclinical data pave the way for future studies to better clarify the mechanism of action of fast-acting antidepressant drugs for the development of novel, more effective therapies.
Collapse
Affiliation(s)
- Jean-Philippe Guilloux
- Laboratoire de Neuropharmacologie, Université Paris-Saclay, Faculté de Pharmacie, Inserm CESP/UMR 1018, Équipe MOODS, F-91400 Orsay, France
| | - Thi Mai Loan Nguyen
- Laboratoire de Neuropharmacologie, Université Paris-Saclay, Faculté de Pharmacie, Inserm CESP/UMR 1018, Équipe MOODS, F-91400 Orsay, France
| | - Alain M Gardier
- Laboratoire de Neuropharmacologie, Université Paris-Saclay, Faculté de Pharmacie, Inserm CESP/UMR 1018, Équipe MOODS, F-91400 Orsay, France
| |
Collapse
|
28
|
Loef D, Tendolkar I, van Eijndhoven PFP, Hoozemans JJM, Oudega ML, Rozemuller AJM, Lucassen PJ, Dols A, Dijkstra AA. Electroconvulsive therapy is associated with increased immunoreactivity of neuroplasticity markers in the hippocampus of depressed patients. Transl Psychiatry 2023; 13:355. [PMID: 37981649 PMCID: PMC10658169 DOI: 10.1038/s41398-023-02658-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Electroconvulsive therapy (ECT) is an effective therapy for depression, but its cellular effects on the human brain remain elusive. In rodents, electroconvulsive shocks increase proliferation and the expression of plasticity markers in the hippocampal dentate gyrus (DG), suggesting increased neurogenesis. Furthermore, MRI studies in depressed patients have demonstrated increases in DG volume after ECT, that were notably paralleled by a decrease in depressive mood scores. Whether ECT also triggers cellular plasticity, inflammation or possibly injury in the human hippocampus, was unknown. We here performed a first explorative, anatomical study on the human post-mortem hippocampus of a unique, well-documented cohort of bipolar or unipolar depressed patients, who had received ECT in the 5 years prior to their death. They were compared to age-matched patients with a depressive disorder who had not received ECT and to matched healthy controls. Upon histopathological examination, no indications were observed for major hippocampal cell loss, overt cytoarchitectural changes or classic neuropathology in these 3 groups, nor were obvious differences present in inflammatory markers for astrocytes or microglia. Whereas the numbers of proliferating cells expressing Ki-67 was not different, we found a significantly higher percentage of cells positive for Doublecortin, a marker commonly used for young neurons and cellular plasticity, in the subgranular zone and CA4 / hilus of the hippocampus of ECT patients. Also, the percentage of positive Stathmin 1 cells was significantly higher in the subgranular zone of ECT patients, indicating neuroplasticity. These first post-mortem observations suggest that ECT has no damaging effects but may rather have induced neuroplasticity in the DG of depressed patients.
Collapse
Affiliation(s)
- Dore Loef
- Amsterdam UMC, location VUmc, Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands.
- GGZ inGeest Specialized Mental Health Care, Amsterdam, the Netherlands.
| | - Indira Tendolkar
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Centre for Cognitive Neuroimaging, Nijmegen, the Netherlands
- Department of Psychiatry and Psychotherapy, University Hospital Essen, Essen, Germany
| | - Philip F P van Eijndhoven
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Centre for Cognitive Neuroimaging, Nijmegen, the Netherlands
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Mardien L Oudega
- Amsterdam UMC, location VUmc, Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands
- GGZ inGeest Specialized Mental Health Care, Amsterdam, the Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Annemiek Dols
- Amsterdam UMC, location VUmc, Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Psychiatry, UMC Utrecht Brain Center, University Utrecht, Utrecht, the Netherlands
| | - Anke A Dijkstra
- Molecular Neuroscience Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
29
|
Wu X, Zhang Y, Wang P, Li X, Song Z, Wei C, Zhang Q, Luo B, Liu Z, Yang Y, Ren Z, Liu H. Clinical and preclinical evaluation of miR-144-5p as a key target for major depressive disorder. CNS Neurosci Ther 2023; 29:3598-3611. [PMID: 37308778 PMCID: PMC10580367 DOI: 10.1111/cns.14291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/06/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Neuronal abnormalities are closely associated with major depressive disorder (MDD). Available evidence suggests a role for microRNAs (miRNAs) in regulating the expression of genes involved in MDD. Hence, miRNAs that can be potential therapeutic targets need to be identified. METHODS A mouse model of chronic unpredictable stress (CUS) was used to evaluate the function of miRNAs in MDD. miR-144-5p was screened from the hippocampi of CUS mice based on sequencing results. Adenovirus-associated vectors were used to overexpress or knockdown miR-144-5p in mice. BpV(pic) and LY294002 were used to determine the relationship between miR-144-5p target genes PTEN and TLR4 in neuronal impairment caused by miR-144-5p deficiency. Western blotting, immunofluorescence, ELISA immunosorbent assay, and Golgi staining were used to detect neuronal abnormalities. Serum samples from healthy individuals and patients with MDD were used to detect miR-144-5p levels in the serum and serum exosomes using qRT-PCR. RESULTS miR-144-5p expression was significantly decreased within the hippocampal dentate gyrus (DG) of CUS mice. Upregulation of miR-144-5p in the DG ameliorated depression-like behavior in CUS mice and attenuated neuronal abnormalities by directly targeting PTEN and TLR4 expression. Furthermore, miR-144-5p knockdown in normal mice led to depression-like behavior via inducing neuronal abnormalities, including abnormal neurogenesis, neuronal apoptosis, altered synaptic plasticity, and neuroinflammation. miR-144-5p deficiency-mediated neuronal impairment was mediated by PI3K/Akt/FoxO1 signaling. Furthermore, miR-144-5p levels were downregulated in the sera of patients with MDD and associated with depressive symptoms. Consistently, serum exosome-derived miR-144-5p levels were decreased in patients with MDD. CONCLUSION miR-144-5p plays a vital role in regulating neuronal abnormalities in depression. Our findings provide translational evidence that miR-144-5p is a new potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Xiaodong Wu
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Yulong Zhang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Ping Wang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Xiaohui Li
- Department of AnatomyAnhui Medical UniversityHefeiChina
| | - Zhen Song
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Chuke Wei
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Qing Zhang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Bei Luo
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Zhichun Liu
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Yingying Yang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Zhenhua Ren
- Department of AnatomyAnhui Medical UniversityHefeiChina
| | - Huanzhong Liu
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| |
Collapse
|
30
|
Fang S, Wu Z, Guo Y, Zhu W, Wan C, Yuan N, Chen J, Hao W, Mo X, Guo X, Fan L, Li X, Chen J. Roles of microglia in adult hippocampal neurogenesis in depression and their therapeutics. Front Immunol 2023; 14:1193053. [PMID: 37881439 PMCID: PMC10597707 DOI: 10.3389/fimmu.2023.1193053] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Collapse
Affiliation(s)
- Shaoyi Fang
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhibin Wu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yali Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenjun Zhu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Chunmiao Wan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Shenzhen People’s Hospital, 2Clinical Medical College, Jinan University, Shenzhen, China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhi Hao
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaowei Mo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lili Fan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
31
|
Zhang SQ, Deng Q, Zhu Q, Hu ZL, Long LH, Wu PF, He JG, Chen HS, Yue Z, Lu JH, Wang F, Chen JG. Cell type-specific NRBF2 orchestrates autophagic flux and adult hippocampal neurogenesis in chronic stress-induced depression. Cell Discov 2023; 9:90. [PMID: 37644025 PMCID: PMC10465581 DOI: 10.1038/s41421-023-00583-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/22/2023] [Indexed: 08/31/2023] Open
Abstract
Dysfunctional autophagy and impairment of adult hippocampal neurogenesis (AHN) each contribute to the pathogenesis of major depressive disorder (MDD). However, whether dysfunctional autophagy is linked to aberrant AHN underlying MDD remains unclear. Here we demonstrate that the expression of nuclear receptor binding factor 2 (NRBF2), a component of autophagy-associated PIK3C3/VPS34-containing phosphatidylinositol 3-kinase complex, is attenuated in the dentate gyrus (DG) under chronic stress. NRBF2 deficiency inhibits the activity of the VPS34 complex and impairs autophagic flux in adult neural stem cells (aNSCs). Moreover, loss of NRBF2 disrupts the neurogenesis-related protein network and causes exhaustion of aNSC pool, leading to the depression-like phenotype. Strikingly, overexpressing NRBF2 in aNSCs of the DG is sufficient to rescue impaired AHN and depression-like phenotype of mice. Our findings reveal a significant role of NRBF2-dependent autophagy in preventing chronic stress-induced AHN impairment and suggest the therapeutic potential of targeting NRBF2 in MDD treatment.
Collapse
Affiliation(s)
- Shao-Qi Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiao Deng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Zhuhai, Macau SAR, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| | - Li-Hong Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| | - Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| | - Jin-Gang He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| | - Hong-Sheng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Zhuhai, Macau SAR, China.
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.
| |
Collapse
|
32
|
AO YAWEN, LI YUSHUANG, ZHAO YILIN, ZHANG LIANG, YANG RENJIE, ZHA YUNFEI. Hippocampal Subfield Volumes in Amateur Marathon Runners. Med Sci Sports Exerc 2023; 55:1208-1217. [PMID: 36878015 PMCID: PMC10241426 DOI: 10.1249/mss.0000000000003144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
PURPOSE Numerous studies have implicated the involvement of structure and function of the hippocampus in physical exercise, and the larger hippocampal volume is one of the relevant benefits reported in exercise. It remains to be determined how the different subfields of hippocampus respond to physical exercise. METHODS A 3D T1-weighted magnetic resonance imaging was acquired in 73 amateur marathon runners (AMR) and 52 healthy controls (HC) matched with age, sex, and education. The Montreal Cognitive Assessment, the Pittsburgh Sleep Quality Index (PSQI), and the Fatigue Severity Scale were assessed in all participants. We obtained hippocampal subfield volumes using FreeSurfer 6.0. We compared the volumes of the hippocampal subfield between the two groups and ascertained correlation between the significant subfield metrics and the significant behavioral measure in AMR group. RESULTS The AMR had significantly better sleep than HC, manifested as with lower score of PSQI. Sleep duration in AMR and HC was not significantly different from each other. In the AMR group, the left and right hippocampus, cornu ammonis 1 (CA1), CA4, granule cell and molecular layers of the dentate gyrus, molecular layer, left CA2-3, and left hippocampal-amygdaloid transition area volumes were significantly larger compared with those in the HC group. In AMR group, the correlations between the PSQI and the hippocampal subfield volumes were not significant. No correlations were found between hippocampal subfield volumes and sleep duration in AMR group. CONCLUSIONS We reported larger volumes of specific hippocampal subfields in AMR, which may provide a hippocampal volumetric reserve that protects against age-related hippocampal deterioration. These findings should be further investigated in longitudinal studies.
Collapse
|
33
|
Musaelyan K, Horowitz MA, McHugh S, Szele FG. Fluoxetine Can Cause Epileptogenesis and Aberrant Neurogenesis in Male Wild-Type Mice. Dev Neurosci 2023; 46:158-166. [PMID: 37302394 DOI: 10.1159/000531478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Antidepressants in general, and fluoxetine in particular, increase adult hippocampal neurogenesis (AHN) in mice. Here we asked how the antidepressant fluoxetine affects behavior and AHN in a corticosterone model of depression. In three groups of adult male C57BL/6j mice, we administered either vehicle (VEH), corticosterone (CORT) treatment to induce a depression-like state, or corticosterone plus a standard dose of fluoxetine (CORT+FLX). Following treatment, mice performed the open field test, the novelty suppressed feeding (NSF) test, and the splash test. Neurogenesis was assessed by means of immunohistochemistry using BrdU and neuronal maturation markers. Unexpectedly, 42% of the CORT+FLX-treated mice exhibited severe weight loss, seizures, and sudden death. As expected, the CORT-treated group had altered behaviors compared to the VEH group, but the CORT+FLX mice that survived did not show any behavioral improvement compared to the CORT group. Antidepressants generally increase neurogenesis and here we also found that compared to CORT mice, CORT+FLX mice that survived had a significantly greater density of BrdU+, BrdU+DCX+, and BrdU+NeuN+ cells, suggesting increased neurogenesis. Moreover, the density of BrdU+NeuN+ cells was increased in an aberrant location, the hilus, of CORT+FLX mice, similar to previous studies describing aberrant neurogenesis following seizures. In conclusion, fluoxetine could induce considerable adverse effects in wild-type mice, including seizure-like activity. Fluoxetine-induced neurogenesis increases could be related to this activity; therefore, proneurogenic effects of fluoxetine and other antidepressants, especially in the absence of any behavioral therapeutic effects, should be interpreted with caution.
Collapse
Affiliation(s)
- Ksenia Musaelyan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Mark A Horowitz
- Research and Development Department, North East London NHS Foundation Trust, Ilford, UK
- Department of Psychiatry, University College London, London, UK
| | - Stephen McHugh
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Atwood B, Yassin W, Chan SY, Hall MH. Subfield-specific longitudinal changes of hippocampal volumes in patients with early-stage bipolar disorder. Bipolar Disord 2023; 25:301-311. [PMID: 36855850 PMCID: PMC10330583 DOI: 10.1111/bdi.13315] [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] [Indexed: 03/02/2023]
Abstract
BACKGROUND The hippocampus is a heterogeneous structure composed of biologically and functionally distinct subfields. Hippocampal aberrations are proposed to play a fundamental role in the etiology of psychotic symptoms. Bipolar disorder (BPD) has substantial overlap in symptomatology and genetic liability with schizophrenia (SZ), and reduced hippocampal volumes, particularly at the chronic illness stages, are documented in both disorders. Studies of hippocampal subfields in the early stage of BPD are limited and cross-sectional findings to date report no reduction in hippocampal volumes. To our knowledge, there have been no longitudinal studies of BPD evaluating hippocampal volumes in the early phase of illness. We investigated the longitudinal changes in hippocampal regions and subfields in BPD mainly and in early stage of psychosis (ESP) patients more broadly and compared them to those in controls (HC). METHODS Baseline clinical and structural MRI data were acquired from 88 BPD, from a total of 143 ESP patients, and 74 HCs. Of those, 66 participants (23 HC, 43 patients) completed a 12-month follow-up visit. The hippocampus regions and subfields were segmented using Freesurfer automated pipeline. RESULTS We found general baseline deficits in hippocampal volumes among BPD and ESP cohorts. Both cohorts displayed significant increases in the anterior hippocampal region and dentate gyrus compared with controls. Additionally, antipsychotic medications were positively correlated with the posterior region at baseline. CONCLUSION These findings highlight brain plasticity in BPD and in ESP patients providing evidence that deviations in hippocampal volumes are adaptive responses to atypical signaling rather than progressive degeneration.
Collapse
Affiliation(s)
- Bruce Atwood
- Psychosis Neurobiology Laboratory, McLean Hospital, Belmont, MA, USA
- Schizophrenia and Bipolar Disorders Program, McLean Hospital, Belmont, MA, USA
| | - Walid Yassin
- Psychosis Neurobiology Laboratory, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Shi Yu Chan
- Psychosis Neurobiology Laboratory, McLean Hospital, Belmont, MA, USA
- Schizophrenia and Bipolar Disorders Program, McLean Hospital, Belmont, MA, USA
| | - Mei-Hua Hall
- Psychosis Neurobiology Laboratory, McLean Hospital, Belmont, MA, USA
- Schizophrenia and Bipolar Disorders Program, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Fung CCA, Fukai T. Competition on presynaptic resources enhances the discrimination of interfering memories. PNAS NEXUS 2023; 2:pgad161. [PMID: 37275260 PMCID: PMC10235910 DOI: 10.1093/pnasnexus/pgad161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023]
Abstract
Evidence suggests that hippocampal adult neurogenesis is critical for discriminating considerably interfering memories. During adult neurogenesis, synaptic competition modifies the weights of synaptic connections nonlocally across neurons, thus providing a different form of unsupervised learning from Hebb's local plasticity rule. However, how synaptic competition achieves separating similar memories largely remains unknown. Here, we aim to link synaptic competition with such pattern separation. In synaptic competition, adult-born neurons are integrated into the existing neuronal pool by competing with mature neurons for synaptic connections from the entorhinal cortex. We show that synaptic competition and neuronal maturation play distinct roles in separating interfering memory patterns. Furthermore, we demonstrate that a feedforward neural network trained by a competition-based learning rule can outperform a multilayer perceptron trained by the backpropagation algorithm when only a small number of samples are available. Our results unveil the functional implications and potential applications of synaptic competition in neural computation.
Collapse
Affiliation(s)
| | - Tomoki Fukai
- To whom correspondence should be addressed: (C.C.A. Fung); (T. Fukai)
| |
Collapse
|
36
|
Meyers KT, Damphousse CC, Ozols AB, Campbell JM, Newbern JM, Hu C, Marrone DF, Gallitano AL. Serial electroconvulsive Seizure alters dendritic complexity and promotes cellular proliferation in the mouse dentate gyrus; a role for Egr3. Brain Stimul 2023; 16:889-900. [PMID: 37146791 PMCID: PMC10776161 DOI: 10.1016/j.brs.2023.04.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Despite being one of the safest, most effective treatments for severe mood disorders, the therapeutic mechanisms of electroconvulsive therapy remain unknown. Electroconvulsive seizure (ECS) induces rapid, high-level expression of immediate early genes (IEGs) and brain-derived neurotrophic factor (BDNF), in addition to stimulation of neurogenesis and dendritic remodeling of dentate gyrus (DG) neurons. We have previously shown that this upregulation of BDNF fails to occur in the hippocampus of mice lacking the IEG Egr3. Since BDNF influences neurogenesis and dendritic remodeling, we hypothesized that Egr3-/- mice will exhibit deficits in neurogenesis and dendritic remodeling in response to ECS. OBJECTIVE To test this hypothesis, we examined dendritic remodeling and cellular proliferation in the DG of Egr3-/- and wild-type mice following repeated ECS. METHODS Mice received 10 daily ECSs. Dendritic morphology was examined in Golgi-Cox-stained tissue and cellular proliferation was analyzed through bromodeoxyuridine (BrdU) immunohistochemistry and confocal imaging. RESULTS Serial ECS in mice results in dendritic remodeling, increased spine density, and cellular proliferation in the DG. Loss of Egr3 alters the dendritic remodeling induced by serial ECS but does not change the number of dendritic spines or cellular proliferation consequences of ECS. CONCLUSION Egr3 influences the dendritic remodeling induced by ECS but is not required for ECS-induced proliferation of hippocampal DG cells.
Collapse
Affiliation(s)
- K T Meyers
- Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ, 85281, USA; Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - C C Damphousse
- Psychology, Wilfrid Laurier University, Waterloo, ON, N2L 3C5, Canada
| | - A B Ozols
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - J M Campbell
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - J M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - C Hu
- Epidemiology and Biostatistics, University of Arizona Mel and Enid Zuckerman College of Public Health - Phoenix, 714 E Van Buren St #119, Phoenix, AZ, 85006, USA
| | - D F Marrone
- Psychology, Wilfrid Laurier University, Waterloo, ON, N2L 3C5, Canada.
| | - A L Gallitano
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA.
| |
Collapse
|
37
|
Li YD, Luo YJ, Xie L, Tart DS, Sheehy RN, Zhang L, Coleman LG, Chen X, Song J. Activation of hypothalamic-enhanced adult-born neurons restores cognitive and affective function in Alzheimer's disease. Cell Stem Cell 2023; 30:415-432.e6. [PMID: 37028406 PMCID: PMC10150940 DOI: 10.1016/j.stem.2023.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/29/2022] [Accepted: 02/14/2023] [Indexed: 04/09/2023]
Abstract
Patients with Alzheimer's disease (AD) exhibit progressive memory loss, depression, and anxiety, accompanied by impaired adult hippocampal neurogenesis (AHN). Whether AHN can be enhanced in impaired AD brain to restore cognitive and affective function remains elusive. Here, we report that patterned optogenetic stimulation of the hypothalamic supramammillary nucleus (SuM) enhances AHN in two distinct AD mouse models, 5×FAD and 3×Tg-AD. Strikingly, the chemogenetic activation of SuM-enhanced adult-born neurons (ABNs) rescues memory and emotion deficits in these AD mice. By contrast, SuM stimulation alone or activation of ABNs without SuM modification fails to restore behavioral deficits. Furthermore, quantitative phosphoproteomics analyses reveal activation of the canonical pathways related to synaptic plasticity and microglia phagocytosis of plaques following acute chemogenetic activation of SuM-enhanced (vs. control) ABNs. Our study establishes the activity-dependent contribution of SuM-enhanced ABNs in modulating AD-related deficits and informs signaling mechanisms mediated by the activation of SuM-enhanced ABNs.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dalton S Tart
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Pharmacology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Libo Zhang
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leon G Coleman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
38
|
Terreros-Roncal J, Flor-García M, Moreno-Jiménez EP, Rodríguez-Moreno CB, Márquez-Valadez B, Gallardo-Caballero M, Rábano A, Llorens-Martín M. Methods to study adult hippocampal neurogenesis in humans and across the phylogeny. Hippocampus 2023; 33:271-306. [PMID: 36259116 PMCID: PMC7614361 DOI: 10.1002/hipo.23474] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/09/2022]
Abstract
The hippocampus hosts the continuous addition of new neurons throughout life-a phenomenon named adult hippocampal neurogenesis (AHN). Here we revisit the occurrence of AHN in more than 110 mammalian species, including humans, and discuss the further validation of these data by single-cell RNAseq and other alternative techniques. In this regard, our recent studies have addressed the long-standing controversy in the field, namely whether cells positive for AHN markers are present in the adult human dentate gyrus (DG). Here we review how we developed a tightly controlled methodology, based on the use of high-quality brain samples (characterized by short postmortem delays and ≤24 h of fixation in freshly prepared 4% paraformaldehyde), to address human AHN. We review that the detection of AHN markers in samples fixed for 24 h required mild antigen retrieval and chemical elimination of autofluorescence. However, these steps were not necessary for samples subjected to shorter fixation periods. Moreover, the detection of labile epitopes (such as Nestin) in the human hippocampus required the use of mild detergents. The application of this strictly controlled methodology allowed reconstruction of the entire AHN process, thus revealing the presence of neural stem cells, proliferative progenitors, neuroblasts, and immature neurons at distinct stages of differentiation in the human DG. The data reviewed here demonstrate that methodology is of utmost importance when studying AHN by means of distinct techniques across the phylogenetic scale. In this regard, we summarize the major findings made by our group that emphasize that overlooking fundamental technical principles might have consequences for any given research field.
Collapse
Affiliation(s)
- Julia Terreros-Roncal
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miguel Flor-García
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Elena P Moreno-Jiménez
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carla B Rodríguez-Moreno
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Berenice Márquez-Valadez
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Gallardo-Caballero
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alberto Rábano
- Neuropathology Department, CIEN Foundation, Madrid, Spain
| | - María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
39
|
Correia AS, Cardoso A, Vale N. Oxidative Stress in Depression: The Link with the Stress Response, Neuroinflammation, Serotonin, Neurogenesis and Synaptic Plasticity. Antioxidants (Basel) 2023; 12:470. [PMID: 36830028 PMCID: PMC9951986 DOI: 10.3390/antiox12020470] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Depression is a prevalent, complex, and highly debilitating disease. The full comprehension of this disease is still a global challenge. Indeed, relapse, recurrency, and therapeutic resistance are serious challenges in the fight against depression. Nevertheless, abnormal functioning of the stress response, inflammatory processes, neurotransmission, neurogenesis, and synaptic plasticity are known to underlie the pathophysiology of this mental disorder. The role of oxidative stress in disease and, particularly, in depression is widely recognized, being important for both its onset and development. Indeed, excessive generation of reactive oxygen species and lack of efficient antioxidant response trigger processes such as inflammation, neurodegeneration, and neuronal death. Keeping in mind the importance of a detailed study about cellular and molecular mechanisms that are present in depression, this review focuses on the link between oxidative stress and the stress response, neuroinflammation, serotonergic pathways, neurogenesis, and synaptic plasticity's imbalances present in depression. The study of these mechanisms is important to lead to a new era of treatment and knowledge about this highly complex disease.
Collapse
Affiliation(s)
- Ana Salomé Correia
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Armando Cardoso
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
40
|
Wolter JM, Le BD, Matoba N, Lafferty MJ, Aygün N, Liang D, Courtney K, Song J, Piven J, Zylka MJ, Stein JL. Cellular Genome-wide Association Study Identifies Common Genetic Variation Influencing Lithium-Induced Neural Progenitor Proliferation. Biol Psychiatry 2023; 93:8-17. [PMID: 36307327 PMCID: PMC9982734 DOI: 10.1016/j.biopsych.2022.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 07/22/2022] [Accepted: 08/18/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Bipolar disorder is a highly heritable neuropsychiatric condition affecting more than 1% of the human population. Lithium salts are commonly prescribed as a mood stabilizer for individuals with bipolar disorder. Lithium is clinically effective in approximately half of treated individuals, and their genetic backgrounds are known to influence treatment outcomes. While the mechanism of lithium's therapeutic action is unclear, it stimulates adult neural progenitor cell proliferation, similar to some antidepressant drugs. METHODS To identify common genetic variants that modulate lithium-induced proliferation, we conducted an EdU incorporation assay in a library of 80 genotyped human neural progenitor cells treated with lithium. These data were used to perform a genome-wide association study to identify common genetic variants that influence lithium-induced neural progenitor cell proliferation. We manipulated the expression of a putatively causal gene using CRISPRi/a (clustered regularly interspaced short palindromic repeats interference/activation) constructs to experimentally verify lithium-induced proliferation effects. RESULTS We identified a locus on chr3p21.1 associated with lithium-induced proliferation. This locus is also associated with bipolar disorder risk, schizophrenia risk, and interindividual differences in intelligence. We identified a single gene, GNL3, whose expression temporally increased in an allele-specific fashion following lithium treatment. Experimentally increasing the expression of GNL3 led to increased proliferation under baseline conditions, while experimentally decreasing GNL3 expression suppressed lithium-induced proliferation. CONCLUSIONS Our experiments reveal that common genetic variation modulates lithium-induced neural progenitor proliferation and that GNL3 expression is necessary for the full proliferation-stimulating effects of lithium. These results suggest that performing genome-wide associations in genetically diverse human cell lines is a useful approach to discover context-specific pharmacogenomic effects.
Collapse
Affiliation(s)
- Justin M Wolter
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brandon D Le
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nana Matoba
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael J Lafferty
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nil Aygün
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dan Liang
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kenan Courtney
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Juan Song
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mark J Zylka
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jason L Stein
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
41
|
Codagnone MG, Kara N, Ratsika A, Levone BR, van de Wouw M, Tan LA, Cunningham JI, Sanchez C, Cryan JF, O'Leary OF. Inhibition of FKBP51 induces stress resilience and alters hippocampal neurogenesis. Mol Psychiatry 2022; 27:4928-4938. [PMID: 36104438 PMCID: PMC9763121 DOI: 10.1038/s41380-022-01755-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 07/07/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023]
Abstract
Stress-related psychiatric disorders such as depression are among the leading causes of morbidity and mortality. Considering that many individuals fail to respond to currently available antidepressant drugs, there is a need for antidepressants with novel mechanisms. Polymorphisms in the gene encoding FK506-binding protein 51 (FKBP51), a co-chaperone of the glucocorticoid receptor, have been linked to susceptibility to stress-related psychiatric disorders. Whether this protein can be targeted for their treatment remains largely unexplored. The aim of this work was to investigate whether inhibition of FKBP51 with SAFit2, a novel selective inhibitor, promotes hippocampal neuron outgrowth and neurogenesis in vitro and stress resilience in vivo in a mouse model of chronic psychosocial stress. Primary hippocampal neuronal cultures or hippocampal neural progenitor cells (NPCs) were treated with SAFit2 and neuronal differentiation and cell proliferation were analyzed. Male C57BL/6 mice were administered SAFit2 while concurrently undergoing a chronic stress paradigm comprising of intermittent social defeat and overcrowding, and anxiety and depressive -related behaviors were evaluated. SAFit2 increased neurite outgrowth and number of branch points to a greater extent than brain derived neurotrophic factor (BDNF) in primary hippocampal neuronal cultures. SAFit2 increased hippocampal NPC neurogenesis and increased neurite complexity and length of these differentiated neurons. In vivo, chronic SAFit2 administration prevented stress-induced social avoidance, decreased anxiety in the novelty-induced hypophagia test, and prevented stress-induced anxiety in the open field but did not alter adult hippocampal neurogenesis in stressed animals. These data warrant further exploration of inhibition of FKBP51 as a strategy to treat stress-related disorders.
Collapse
Affiliation(s)
- Martin G Codagnone
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Instituto de Biología Celular y Neurociencia "de Robertis" IBCN (UBA-CONICET), Buenos Aires, Argentina
| | - Nirit Kara
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Anna Ratsika
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Brunno Rocha Levone
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | | | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
42
|
Guzzetta KE, Cryan JF, O’Leary OF. Microbiota-Gut-Brain Axis Regulation of Adult Hippocampal Neurogenesis. Brain Plast 2022; 8:97-119. [PMID: 36448039 PMCID: PMC9661352 DOI: 10.3233/bpl-220141] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 11/15/2022] Open
Abstract
The birth, maturation, and integration of new neurons in the adult hippocampus regulates specific learning and memory processes, responses to stress, and antidepressant treatment efficacy. This process of adult hippocampal neurogenesis is sensitive to environmental stimuli, including peripheral signals from certain cytokines, hormones, and metabolites, which can promote or hinder the production and survival of new hippocampal neurons. The trillions of microorganisms resident to the gastrointestinal tract, collectively known as the gut microbiota, also demonstrate the ability to modulate adult hippocampal neurogenesis. In doing so, the microbiota-gut-brain axis can influence brain functions regulated by adult hippocampal neurogenesis. Unlike the hippocampus, the gut microbiota is highly accessible to direct interventions, such as prebiotics, probiotics, and antibiotics, and can be manipulated by lifestyle choices including diet. Therefore, understanding the pathways by which the gut microbiota shapes hippocampal neurogenesis may reveal novel targets for non-invasive therapeutics to treat disorders in which alterations in hippocampal neurogenesis have been implicated. This review first outlines the factors which influence both the gut microbiome and adult hippocampal neurogenesis, with cognizance that these effects might happen either independently or due to microbiota-driven mechanisms. We then highlight approaches for investigating the regulation of adult hippocampal neurogenesis by the microbiota-gut-brain axis. Finally, we summarize the current evidence demonstrating the gut microbiota's ability to influence adult hippocampal neurogenesis, including mechanisms driven through immune pathways, microbial metabolites, endocrine signalling, and the nervous system, and postulate implications for these effects in disease onset and treatment.
Collapse
Affiliation(s)
- Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Olivia F. O’Leary
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
43
|
Hao K, Wang H, Zhang Y, Xie X, Huang H, Chen C, Xu S, Xu R, Shu C, Liu Z, Zhou Y, Reynolds GP, Wang G. Nicotinamide reverses deficits in puberty-born neurons and cognitive function after maternal separation. J Neuroinflammation 2022; 19:232. [PMID: 36131290 PMCID: PMC9494869 DOI: 10.1186/s12974-022-02591-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 09/04/2022] [Indexed: 01/09/2023] Open
Abstract
Background Early life stress (ELS) is associated with the development of schizophrenia later in life. The hippocampus develops significantly during childhood and is extremely reactive to stress. In rodent models, ELS can induce neuroinflammation, hippocampal neuronal loss, and schizophrenia-like behavior. While nicotinamide (NAM) can inhibit microglial inflammation, it is unknown whether NAM treatment during adolescence reduces hippocampal neuronal loss and abnormal behaviors induced by ELS. Methods Twenty-four hours of maternal separation (MS) of Wistar rat pups on post-natal day (PND)9 was used as an ELS. On PND35, animals received a single intraperitoneal injection of BrdU to label dividing neurons and were given NAM from PND35 to PND65. Behavioral testing was performed. Western blotting and immunofluorescence staining were used to detect nicotinamide adenine dinucleotide (NAD+)/Sirtuin3 (Sirt3)/superoxide dismutase 2 (SOD2) pathway-related proteins. Results Compared with controls, only MS animals in the adult stage (PND56–65) but not the adolescent stage (PND31–40) exhibited pre-pulse inhibition deficits and cognitive impairments mimicking schizophrenia symptoms. MS decreased the survival and activity of puberty-born neurons and hippocampal NAD+ and Sirt3 expression in adulthood. These observations were related to an increase in acetylated SOD2, microglial activation, and significant increases in pro-inflammatory IL-1β, TNF-α, and IL-6 expression. All the effects of MS at PND9 were reversed by administering NAM in adolescence (PND35–65). Conclusions MS may lead to schizophrenia-like phenotypes and persistent hippocampal abnormalities. NAM may be a safe and effective treatment in adolescence to restore normal hippocampal function and prevent or ameliorate schizophrenia-like behavior. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02591-y.
Collapse
Affiliation(s)
- Keke Hao
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China. .,Department of Psychiatry, Zhongxiang Hospital of Renmin Hospital of Wuhan University, Zhongxiang, 431900, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Yuejin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, China
| | - Xinhui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Huan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Cheng Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Shilin Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Rui Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Chang Shu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yuan Zhou
- Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China. .,Hubei Institute of Neurology and Psychiatry Research, Wuhan, 430060, China.
| |
Collapse
|
44
|
Zhang S, Lu Y, Shi W, Ren Y, Xiao K, Chen W, Li L, Zhao J. SIRT1/FOXO1 Axis-Mediated Hippocampal Angiogenesis is Involved in the Antidepressant Effect of Chaihu Shugan San. Drug Des Devel Ther 2022; 16:2783-2801. [PMID: 36039087 PMCID: PMC9419814 DOI: 10.2147/dddt.s370825] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Methods Results Conclusion
Collapse
Affiliation(s)
- Shan Zhang
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Yujia Lu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Wei Shi
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Yi Ren
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Kaihui Xiao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Li Li
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Jingjie Zhao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
- Department of Integrated Traditional and Western Medicine, Capital Medical University, Beijing, 100050, People’s Republic of China
- Correspondence: Jingjie Zhao, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Beijing, 100050, People’s Republic of China, Tel/Fax +86 10-63139096, Email
| |
Collapse
|
45
|
Shi LS, Ji CH, Liu Y, Gu JH, Tang WQ, Zhang W, Guan W. Ginsenoside Rh2 administration produces crucial antidepressant-like effects in a CUMS-induced mice model of depression. Brain Behav 2022; 12:e2705. [PMID: 35848938 PMCID: PMC9392527 DOI: 10.1002/brb3.2705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/26/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022] Open
Abstract
INTRODUCTION The most striking feature of depression is sadness and a loss of interest in activities, which represents a major cause of disability globally. Therefore, it is necessary to identify novel antidepressants for clinical practice. Ginsenoside Rh2 (Rh2) is one of the major bioactive ginsenosides that can be extracted from Panax ginseng and has been demonstrated to improve both memory and learning. The purpose of this study was to provide broad insight into the mechanisms underlying depression and gain greater insights into antidepressant therapy. METHODS In this study, we first established an effective and feasible depression animal model of chronic unpredictable mild stress (CUMS) and behavioral testing was evaluated by the forced swim test (FST), the tail suspension test (TST) and the sucrose preference test. Following pretreatment with Rh2 (10 and 20 mg/kg), the immobility time of mice was reduced without affecting locomotor activity in both the FST and TST. Western blotting and immunofluorescence were used to investigate the activation of the hippocampal BDNF signaling pathway and hippocampal neurogenesis. RESULTS Different concentrations of Rh2 significantly reduced depressive-like symptoms in CUMS-induced mice and downregulated the effects of the BDNF signaling cascade and neurogenesis in the hippocampus. Furthermore, the administration of K252a completely prevented the antidepressant-like activity of Rh2 in mice. CONCLUSION The results indicated that Rh2 possesses the antidepression action via the positive regulation of the BDNF-TrkB pathway.
Collapse
Affiliation(s)
- Lin-Sheng Shi
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Chun-Hui Ji
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Yue Liu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Jiang-Hong Gu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wen-Qian Tang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wei Zhang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
46
|
Jones KL, Zhou M, Jhaveri DJ. Dissecting the role of adult hippocampal neurogenesis towards resilience versus susceptibility to stress-related mood disorders. NPJ SCIENCE OF LEARNING 2022; 7:16. [PMID: 35842419 PMCID: PMC9288448 DOI: 10.1038/s41539-022-00133-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/01/2022] [Indexed: 05/13/2023]
Abstract
Adult hippocampal neurogenesis in the developmental process of generating and integrating new neurons in the hippocampus during adulthood and is a unique form of structural plasticity with enormous potential to modulate neural circuit function and behaviour. Dysregulation of this process is strongly linked to stress-related neuropsychiatric conditions such as anxiety and depression, and efforts have focused on unravelling the contribution of adult-born neurons in regulating stress response and recovery. Chronic stress has been shown to impair this process, whereas treatment with clinical antidepressants was found to enhance the production of new neurons in the hippocampus. However, the precise role of adult hippocampal neurogenesis in mediating the behavioural response to chronic stress is not clear and whether these adult-born neurons buffer or increase susceptibility to stress-induced mood-related maladaptation remains one of the controversial issues. In this review, we appraise evidence probing the causal role of adult hippocampal neurogenesis in the regulation of emotional behaviour in rodents. We find that the relationship between adult-born hippocampal neurons and stress-related mood disorders is not linear, and that simple subtraction or addition of these neurons alone is not sufficient to lead to anxiety/depression or have antidepressant-like effects. We propose that future studies examining how stress affects unique properties of adult-born neurons, such as the excitability and the pattern of connectivity during their critical period of maturation will provide a deeper understanding of the mechanisms by which these neurons contribute to functional outcomes in stress-related mood disorders.
Collapse
Affiliation(s)
- Katherine L Jones
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Mei Zhou
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
47
|
Cai MY, Yang Z, Huang XJ, Li J, Bao WY, Cui JW, Ma LQ, Tong HY. Mongolian Medicine Areca Thirteen Pill (GY-13) Improved Depressive Syndrome via upregulating cAMP/PKA/CREB/BDNF signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115310. [PMID: 35452773 DOI: 10.1016/j.jep.2022.115310] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/08/2022] [Accepted: 04/16/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Areca Thirteen Pill, also called Gao You-13 (GY-13), is a traditional Mongolian herbal formula and has been extensively used to treat depression in Mongolian areas, which belongs to Heyi disease in Mongolian medicine. Major depressive disorder is a serious psychiatric disease, only one-third of individuals with depression are responsive to current antidepressants in clinic. Growing attention has been attracted by traditional herbal medicines in fighting depression because they are considered safer alternatives to pharmacotherapy. AIM OF THE STUDY To reveal the mechanism of GY-13 in the treatment of depression. MATERIALS AND METHODS The rat depression model was established by chronic unpredictable mild stress (CUMS), and primary hippocampal neurons were used to construct a glutamate-induced excitotoxicity model. The antidepressant effect of GY-13 was then assessed by performing sucrose preference tests, open field tests, and body weight measurements on rats. The expression of cAMP and PKA, mRNA levels of brain-derived neurotrophic factor (BDNF) and cAMP response element binding protein (CREB), and hippocampal neuronal apoptosis were measured. RESULTS The results indicate that GY-13 significantly improves depression-like behavior, rescues decreased cAMP, PKA, recovers the mRNA levels of CREB and BDNF, and increases the proliferative activity of hippocampus. In addition, blockade of PKA reverses the effects of GY-13 treatment on CREB mRNA, BDNF mRNA levels. In vitro, GY-13 treatment increased hippocampal proliferative activity and attenuated Glu-induced apoptosis of hippocampal neurons as well as reduced CREB mRNA and BDNF mRNA expression levels. CONCLUSIONS Our research demonstrated that GY-13 treatment exerted a potent antidepressant action via activation of cAMP/CREB/BDNF signaling pathway, promoting proliferation, and suppressing apoptosis. This research provides molecular biological ground for developing GY-13 into a potent alternative for the intervention of depression.
Collapse
Affiliation(s)
- Ming-Yang Cai
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Zhen Yang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Xian-Ju Huang
- South-Central University for Nationalities, Wuhan, 430074, PR China.
| | - Jing Li
- Mongolian Hospital of Fuxin Mongolian Autonomous County, Liaoning Province, Fuxin, 123199, PR China.
| | - Wu-Ye Bao
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, 028007, PR China.
| | - Jing-Wen Cui
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Li-Qun Ma
- South-Central University for Nationalities, Wuhan, 430074, PR China.
| | - Hai-Ying Tong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
48
|
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78:101636. [PMID: 35490966 PMCID: PMC9168971 DOI: 10.1016/j.arr.2022.101636] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Adult neurogenesis, the process by which neurons are generated in certain areas of the adult brain, declines in an age-dependent manner and is one potential target for extending cognitive healthspan. Aging is a major risk factor for neurodegenerative diseases and, as lifespans are increasing, these health challenges are becoming more prevalent. An age-associated loss in neural stem cell number and/or activity could cause this decline in brain function, so interventions that reverse aging in stem cells might increase the human cognitive healthspan. In this review, we describe the involvement of adult neurogenesis in neurodegenerative diseases and address the molecular mechanistic aspects of neurogenesis that involve some of the key aggregation-prone proteins in the brain (i.e., tau, Aβ, α-synuclein, …). We summarize the research pertaining to interventions that increase neurogenesis and regulate known targets in aging research, such as mTOR and sirtuins. Lastly, we share our outlook on restoring the levels of neurogenesis to physiological levels in elderly individuals and those with neurodegeneration. We suggest that modulating neurogenesis represents a potential target for interventions that could help in the fight against neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Luka Culig
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xixia Chu
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
49
|
Zhuang W, Liu H, He Z, Ju J, Gao Q, Shan Z, Lei L. miR-92a-2-5p Regulates the Proliferation and Differentiation of ASD-Derived Neural Progenitor Cells. Curr Issues Mol Biol 2022; 44:2431-2442. [PMID: 35735607 PMCID: PMC9222067 DOI: 10.3390/cimb44060166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex neurodevelopmental disorders with abnormal behavior. However, the pathogenesis of ASD remains to be clarified. It has been demonstrated that miRNAs are essential regulators of ASD. However, it is still unclear how miR-92a-2-5p acts on the developing brain and the cell types directly. In this study, we used neural progenitor cells (NPCs) derived from ASD-hiPSCs as well as from neurotypical controls to examine the effects of miR-92a-2-5p on ASD-NPCs proliferation and neuronal differentiation, and whether miR-92a-2-5p could interact with genetic risk factor, DLG3 for ASD. We observed that miR-92a-2-5p upregulated in ASD-NPCs results in decreased proliferation and neuronal differentiation. Inhibition of miR-92a-2-5p could promote proliferation and neuronal differentiation of ASD-NPCs. DLG3 was negatively regulated by miR-92a-2-5p in NPCs. Our results suggest that miR-92a-2-5p is a strong risk factor for ASD and potentially contributes to neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lei Lei
- Correspondence: (Z.S.); (L.L.)
| |
Collapse
|
50
|
Zeng J, Ji Y, Luan F, Hu J, Rui Y, Liu Y, Rao Z, Liu R, Zeng N. Xiaoyaosan ethyl acetate fraction alleviates depression-like behaviors in CUMS mice by promoting hippocampal neurogenesis via modulating the IGF-1Rβ/PI3K/Akt signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:115005. [PMID: 35051601 DOI: 10.1016/j.jep.2022.115005] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/21/2021] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoyaosan (XYS), a representative and classic traditional Chinese medicine (TCM) prescription with function of dispersing stagnated liver and strengthening spleen, has been used for thousands of years to treat depression. XYS' anti-depression effect has been demonstrated both clinically and experimentally; however, the material basis for this effect has yet to be elucidated. AIM OF THE STUDY This study aimed to evaluate the impact and underlying action mechanism of XYS' antidepressant active component (Xiaoyaosan ethyl acetate fraction, XYSEF) against chronic unpredictable mild stress (CUMS)-induced depression-like behavior in mice. MATERIALS AND METHODS First, we established a behavioral despair depression mouse model to preliminarily determine the effective antidepressant dose of XYSEF. Then, we created a CUMS mouse model and used various classic behavioral tests, including SPT, ST, NFST, and TST, to assess XYSEF's antidepressant properties. IGF-1 levels in mouse serum and hippocampus were quantified using ELISA. The average optical density of Nissl bodies in the mouse hippocampal CA3 region was determined utilizing toluidine blue staining. Brdu and DCX expression in the hippocampal dentate gyrus (DG) was assayed using the immunofluorescence method. IGF-1Rβ, PI3K, p-PI3K, Akt, p-Akt, Caspase-3, and cleaved Caspase-3 protein levels in the hippocampus were determined with Western blot. RESULTS The behavioral despair mouse model findings showed that 9.1 and 40 g/kg of XYSEF both significantly shortened the immobility time of mice, suggesting that the effective dose range was 9.1-40 g/kg. Compared to the CUMS mouse model, XYSEF at 20 and 40 g/kg markedly increased the sucrose preference percentage in the SPT and grooming time in the ST, shortened the immobility time in the TST and the feeding latency in the NSFT, and reversed the downregulated IGF-1 content in mouse serum and hippocampus. In addition, XYSEF amplified the average optical density of Nissl bodies in the hippocampal CA3 region, promoted Brdu and DCX expression in DG, and diminished IGF-1Rβ, p-PI3K/PI3K, p-Akt/Akt, and cleaved Caspase-3/Caspase-3 protein levels in the hippocampi of CUMS mice. CONCLUSION XYSEF acted as an antidepressant in mice exhibiting CUMS-induced depression-like behaviors, possibly by promoting hippocampal neurogenesis, reducing neuronal apoptosis, and inhibiting the over-activation of the IGF-1Rβ/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yafei Ji
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Fei Luan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jingwen Hu
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yixing Rui
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yao Liu
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Zhili Rao
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rong Liu
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|