1
|
Wang D, Fukuda T, Wu T, Xu X, Isaji T, Gu J. Exogenous L-fucose attenuates depression induced by chronic unpredictable stress: Implicating core fucosylation has an antidepressant potential. J Biol Chem 2025; 301:108230. [PMID: 39864626 PMCID: PMC11879694 DOI: 10.1016/j.jbc.2025.108230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Core fucosylation is one of the most essential modifications of the N-glycans, catalyzed by α1,6-fucosyltransferase (Fut8), which transfers fucose from guanosine 5'-diphosphate (GDP)-fucose to the innermost N-acetylglucosamine residue of N-glycans in an α1-6 linkage. Our previous studies demonstrated that lipopolysaccharide (LPS) can induce a more robust neuroinflammatory response in Fut8 homozygous knockout (KO) (Fut8-/-) and heterozygous KO (Fut8+/-) mice contrasted to the wild-type (Fut8+/+) mice. Exogenous administration of L-fucose suppressed LPS-induced neuroinflammation. Numerous studies indicate that neuroinflammation plays a vital role in the development of depression. Here, we investigated whether core fucosylation regulates depression induced by chronic unpredictable stress (CUS), a well-established model for depression. Our results showed that Fut8+/- mice exhibited depressive-like behaviors and increased neuroinflammation earlier than Fut8+/+ mice. Administration of L-fucose significantly reduced CUS-induced depressive-like behaviors and pro-inflammatory cytokine levels in Fut8+/- mice. The L-fucose treatment produced antidepressant effects by attenuating the complex formation between gp130 and the interleukin-6 (IL-6) receptor and the JAK2/STAT3 signaling pathway. Notably, L-fucose treatment increased dendritic spine density and postsynaptic density protein 95 (PSD-95) expression, which were suppressed in CUS-induced depression. Furthermore, the effects of L-fucose on the CUS-induced depression were also observed in Fut8+/+ mice. Our results clearly demonstrate that L-fucose ameliorates neuroinflammation and synaptic defects in CUS-induced depression, implicating that core fucosylation has significant anti-neuroinflammatory activity and an antidepressant potential.
Collapse
Affiliation(s)
- Dan Wang
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan; Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| | - Tiangui Wu
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Xing Xu
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan; Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan; Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|
2
|
Saccenti D, Lauro LJR, Crespi SA, Moro AS, Vergallito A, Grgič RG, Pretti N, Lamanna J, Ferro M. Boosting Psychotherapy With Noninvasive Brain Stimulation: The Whys and Wherefores of Modulating Neural Plasticity to Promote Therapeutic Change. Neural Plast 2024; 2024:7853199. [PMID: 39723244 PMCID: PMC11669434 DOI: 10.1155/np/7853199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
The phenomenon of neural plasticity pertains to the intrinsic capacity of neurons to undergo structural and functional reconfiguration through learning and experiential interaction with the environment. These changes could manifest themselves not only as a consequence of various life experiences but also following therapeutic interventions, including the application of noninvasive brain stimulation (NIBS) and psychotherapy. As standalone therapies, both NIBS and psychotherapy have demonstrated their efficacy in the amelioration of psychiatric disorders' symptoms, with a certain variability in terms of effect sizes and duration. Consequently, scholars suggested the convenience of integrating the two interventions into a multimodal treatment to boost and prolong the therapeutic outcomes. Such an approach is still in its infancy, and the physiological underpinnings substantiating the effectiveness and utility of combined interventions are still to be clarified. Therefore, this opinion paper aims to provide a theoretical framework consisting of compelling arguments as to why adding NIBS to psychotherapy can promote therapeutic change. Namely, we will discuss the physiological effects of the two interventions, thus providing a rationale to explain the potential advantages of a combined approach.
Collapse
Affiliation(s)
- Daniele Saccenti
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Leonor J. Romero Lauro
- Department of Psychology and NeuroMi, University of Milano-Bicocca, Milan, Italy
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
| | - Sofia A. Crespi
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea S. Moro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | | | | | - Novella Pretti
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Clinical Psychology Center, Division of Neurology, Galliera Hospital, Genoa, Italy
| | - Jacopo Lamanna
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Ferro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
3
|
Fakih N, Fakhoury M. Alzheimer Disease-Link With Major Depressive Disorder and Efficacy of Antidepressants in Modifying its Trajectory. J Psychiatr Pract 2024; 30:181-191. [PMID: 38819242 DOI: 10.1097/pra.0000000000000779] [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] [Indexed: 06/01/2024]
Abstract
Alzheimer disease (AD) is a devastating neurodegenerative disorder that affects millions of individuals worldwide, with no effective cure. The main symptoms include learning and memory loss, and the inability to carry out the simplest tasks, significantly affecting patients' quality of life. Over the past few years, tremendous progress has been made in research demonstrating a link between AD and major depressive disorder (MDD). Evidence suggests that MDD is commonly associated with AD and that it can serve as a precipitating factor for this disease. Antidepressants such as selective serotonin reuptake inhibitors, which are the first line of treatment for MDD, have shown great promise in the treatment of depression in AD, although their effectiveness remains controversial. The goal of this review is to summarize current knowledge regarding the association between AD, MDD, and antidepressant treatment. It first provides an overview of the interaction between AD and MDD at the level of genes, brain regions, neurotransmitter systems, and neuroinflammatory markers. The review then presents current evidence regarding the effectiveness of various antidepressants for AD-related pathophysiology and then finally discusses current limitations, challenges, and future directions.
Collapse
Affiliation(s)
- Nour Fakih
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | | |
Collapse
|
4
|
Vilar-Pereira G, Gibaldi D, Castaño-Barrios L, da Silva AA, Resende Pereira I, Cruz Moreira O, Britto C, Mata dos Santos HA, de Oliveira Lopes R, Wanderley Tinoco L, Oliveira W, Lannes-Vieira J. The beneficial effect of fluoxetine on behavioral and cognitive changes in chronic experimental Chagas disease unveils the role of serotonin fueling astrocyte infection by Trypanosoma cruzi. PLoS Negl Trop Dis 2024; 18:e0012199. [PMID: 38776344 PMCID: PMC11149870 DOI: 10.1371/journal.pntd.0012199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 06/04/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND In Chagas disease (CD), a neglected tropical disease caused by the parasite Trypanosoma cruzi, the development of mental disorders such as anxiety, depression, and memory loss may be underpinned by social, psychological, and biological stressors. Here, we investigated biological factors underlying behavioral changes in a preclinical model of CD. METHODOLOGY/PRINCIPAL FINDINGS In T. cruzi-infected C57BL/6 mice, a kinetic study (5 to 150 days postinfection, dpi) using standardized methods revealed a sequential onset of behavioral changes: reduced innate compulsive behavior, followed by anxiety and depressive-like behavior, ending with progressive memory impairments. Hence, T. cruzi-infected mice were treated (120 to 150 dpi) with 10 mg/Kg/day of the selective serotonin reuptake inhibitor fluoxetine (Fx), an antidepressant that favors neuroplasticity. Fx therapy reversed the innate compulsive behavior loss, anxiety, and depressive-like behavior while preventing or reversing memory deficits. Biochemical, histological, and parasitological analyses of the brain tissue showed increased levels of the neurotransmitters GABA/glutamate and lipid peroxidation products and decreased expression of brain-derived neurotrophic factor in the absence of neuroinflammation at 150 dpi. Fx therapy ameliorated the neurochemical changes and reduced parasite load in the brain tissue. Next, using the human U-87 MG astroglioma cell line, we found no direct effect of Fx on parasite load. Crucially, serotonin/5-HT (Ser/5-HT) promoted parasite uptake, an effect increased by prior stimulation with IFNγ and TNF but abrogated by Fx. Also, Fx blocked the cytokine-driven Ser/5-HT-promoted increase of nitric oxide and glutamate levels in infected cells. CONCLUSION/SIGNIFICANCE We bring the first evidence of a sequential onset of behavioral changes in T. cruzi-infected mice. Fx therapy improves behavioral and biological changes and parasite control in the brain tissue. Moreover, in the central nervous system, cytokine-driven Ser/5-HT consumption may favor parasite persistence, disrupting neurotransmitter balance and promoting a neurotoxic environment likely contributing to behavioral and cognitive disorders.
Collapse
Affiliation(s)
- Glaucia Vilar-Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
| | - Daniel Gibaldi
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
| | - Leda Castaño-Barrios
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
| | - Andrea Alice da Silva
- Laboratório Multidisciplinar de Apoio à Pesquisa em Nefrologia e Ciências Médicas, Departamento de Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil
| | - Isabela Resende Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
| | - Otacílio Cruz Moreira
- Laboratório de Virologia e Parasitologia Molecular, IOC/Fiocruz, Rio de Janeiro, Brazil
| | - Constança Britto
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC/Fiocruz, Rio de Janeiro, Brazil
| | - Hílton Antônio Mata dos Santos
- Escola de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Análise e Desenvolvimento de Inibidores Enzimáticos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel de Oliveira Lopes
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório Multiusuário de Análises por Ressonância Magnética Nuclear (LAMAR), Instituto de Pesquisas de Produtos Naturais (IPPN), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luzineide Wanderley Tinoco
- Laboratório Multiusuário de Análises por Ressonância Magnética Nuclear (LAMAR), Instituto de Pesquisas de Produtos Naturais (IPPN), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wilson Oliveira
- Ambulatório de Doença de Chagas e Insuficiência Cardíaca, Pronto Socorro Cardiológico de Pernambuco (PROCAPE)/Universidade de Pernambuco, Recife, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Qian X, Zhong ZD, Zhang Y, Qiu LQ, Tan HJ. Fluoxetine mitigates depressive-like behaviors in mice via anti-inflammation and enhancing neuroplasticity. Brain Res 2024; 1825:148723. [PMID: 38101693 DOI: 10.1016/j.brainres.2023.148723] [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: 10/09/2023] [Revised: 11/22/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Neuroplasticity and inflammation represent a common final pathway for effective antidepressant treatment. SSRIs are the most commonly prescribed medications for depression and have demonstrated efficacy in reducing depressive symptoms. However, the precise impact of SSRIs on neuroplasticity and inflammation remains unclear. In this study, we aimed to investigate the influence of long-term treatment with SSRIs on hippocampal neuron, inflammation, synaptic function and morphology. Our findings revealed that fluoxetine treatment significantly alleviated behavioral despair, anhedonia, and anxiety in reserpine-treated mice. Moreover, fluoxetine mitigated hippocampal neuron impairment, inhibited inflammatory release, and increased the expression of synaptic proteins markers (SYP and PSD95) in mice. Notably, fluoxetine also suppressed reserpine-induced synapse loss in the hippocampus. Based on these results, fluoxetine has been demonstrated effectively to ameliorate depressive mood and cognitive dysfunction, possibly through the enhancement of synaptic plasticity. Overall, our study contributes to a further understanding of the mechanisms underlying the therapeutic effects of fluoxetine and its potential role in improving depressive symptoms and cognitive impairments.
Collapse
Affiliation(s)
- Xu Qian
- School of Chemistry, Guangdong Key Lab of Chiral Molecules and Drug Discovery, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Zuo-Dong Zhong
- School of Pharmacy, Guangzhou Medical University, Guangzhou 510275, China
| | - Yao Zhang
- Department of Respiratory and Critical Medicine, General Hospital of Eastern Theater Command, Nanjing 210016, China
| | - Li-Qin Qiu
- School of Chemistry, Guangdong Key Lab of Chiral Molecules and Drug Discovery, Sun Yat-Sen University, Guangzhou 510275, China
| | - Hui-Jun Tan
- School of Chemistry, Guangdong Key Lab of Chiral Molecules and Drug Discovery, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
6
|
Filipović D, Inderhees J, Korda A, Tadić P, Schwaninger M, Inta D, Borgwardt S. Metabolic Fingerprints of Effective Fluoxetine Treatment in the Prefrontal Cortex of Chronically Socially Isolated Rats: Marker Candidates and Predictive Metabolites. Int J Mol Sci 2023; 24:10957. [PMID: 37446133 PMCID: PMC10341512 DOI: 10.3390/ijms241310957] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
The increasing prevalence of depression requires more effective therapy and the understanding of antidepressants' mode of action. We carried out untargeted metabolomics of the prefrontal cortex of rats exposed to chronic social isolation (CSIS), a rat model of depression, and/or fluoxetine treatment using liquid chromatography-high resolution mass spectrometry. The behavioral phenotype was assessed by the forced swim test. To analyze the metabolomics data, we employed univariate and multivariate analysis and biomarker capacity assessment using the receiver operating characteristic (ROC) curve. We also identified the most predictive biomarkers using a support vector machine with linear kernel (SVM-LK). Upregulated myo-inositol following CSIS may represent a potential marker of depressive phenotype. Effective fluoxetine treatment reversed depressive-like behavior and increased sedoheptulose 7-phosphate, hypotaurine, and acetyl-L-carnitine contents, which were identified as marker candidates for fluoxetine efficacy. ROC analysis revealed 4 significant marker candidates for CSIS group discrimination, and 10 for fluoxetine efficacy. SVM-LK with accuracies of 61.50% or 93.30% identified a panel of 7 or 25 predictive metabolites for depressive-like behavior or fluoxetine effectiveness, respectively. Overall, metabolic fingerprints combined with the ROC curve and SVM-LK may represent a new approach to identifying marker candidates or predictive metabolites for ongoing disease or disease risk and treatment outcome.
Collapse
Affiliation(s)
- Dragana Filipović
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Julica Inderhees
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany; (J.I.); (M.S.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg-Kiel-Lübeck, 20251 Hamburg, Germany
- Center of Brain Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Alexandra Korda
- Department of Psychiatry and Psychotherapy, Center of Brain Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany;
| | - Predrag Tadić
- School of Electrical Engineering, University of Belgrade, 11000 Belgrade, Serbia;
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany; (J.I.); (M.S.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg-Kiel-Lübeck, 20251 Hamburg, Germany
| | - Dragoš Inta
- Department for Community Health, Faculty of Natural Sciences, Medicine, University of Fribourg, 1700 Fribourg, Switzerland; (D.I.); (S.B.)
- Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| | - Stefan Borgwardt
- Department for Community Health, Faculty of Natural Sciences, Medicine, University of Fribourg, 1700 Fribourg, Switzerland; (D.I.); (S.B.)
| |
Collapse
|
7
|
Jetsonen E, Didio G, Winkel F, Llach Pou M, Boj C, Kuczynski-Noyau L, Võikar V, Guirado R, Taira T, Lauri SE, Castrén E, Umemori J. Activation of TrkB in Parvalbumin interneurons is required for the promotion of reversal learning in spatial and fear memory by antidepressants. Neuropsychopharmacology 2023; 48:1021-1030. [PMID: 36944718 PMCID: PMC10209093 DOI: 10.1038/s41386-023-01562-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/23/2023]
Abstract
Critical period-like plasticity (iPlasticity) can be reinstated in the adult brain by several interventions, including drugs and optogenetic modifications. We have demonstrated that a combination of iPlasticity with optimal training improves behaviors related to neuropsychiatric disorders. In this context, the activation of TrkB, a receptor for BDNF, in Parvalbumin-positive (PV+) interneurons has a pivotal role in cortical network changes. However, it is unknown if the activation of TrkB in PV+ interneurons is important for other plasticity-related behaviors, especially for learning and memory. Here, using mice with heterozygous conditional TrkB deletion in PV+ interneurons (PV-TrkB hCKO) in IntelliCage and fear erasure paradigms, we show that chronic treatment with fluoxetine, a widely prescribed antidepressant drug that is known to promote the activation of TrkB, enhances behavioral flexibility in spatial and fear memory, largely depending on the expression of the TrkB receptor in PV+ interneurons. In addition, hippocampal long-term potentiation was enhanced by chronic treatment with fluoxetine in wild-type mice, but not in PV-TrkB hCKO mice. Transcriptomic analysis of PV+ interneurons after fluoxetine treatment indicated intrinsic changes in synaptic formation and downregulation of enzymes involved in perineuronal net formation. Consistently, immunohistochemistry has shown that the fluoxetine treatment alters PV expression and reduces PNNs in PV+ interneurons, and here we show that TrkB expression in PV+ interneurons is required for these effects. Together, our results provide molecular and network mechanisms for the induction of critical period-like plasticity in adulthood.
Collapse
Affiliation(s)
- Elias Jetsonen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Giuliano Didio
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Frederike Winkel
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Maria Llach Pou
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Integrative Neurobiology of Cholinergic Systems, Neuroscience Department, Institut Pasteur, Paris, France
| | - Chloe Boj
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Laura Kuczynski-Noyau
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, F-59000, Lille, France
| | - Vootele Võikar
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ramon Guirado
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain
| | - Tomi Taira
- Department of Veterinary Biosciences and Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Sari E Lauri
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Juzoh Umemori
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland.
- Gene and Cell Technology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
8
|
Gerace E, Polenzani L, Magnani M, Zianni E, Stocca G, Gardoni F, Pellegrini-Giampietro DE, Corradetti R. Antidepressant-induced increase in GluA2 expression does not translate in changes of AMPA receptor-mediated synaptic transmission at CA3/CA1 synapses in rats. Neuropharmacology 2023; 223:109307. [PMID: 36334766 DOI: 10.1016/j.neuropharm.2022.109307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/14/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Chronic treatment with serotonin selective reuptake inhibitors or tryciclic antidepressant drugs in rodents has been shown to increase the expression of GluA1 and/or GluA2 AMPA receptor (AMPAR) subunits in several brain areas, including the hippocampus. These changes in AMPAR composition have been suggested to result in increased glutamatergic neurotransmission and possibly underlie enhanced hippocampal synaptic plasticity through the increased availability of calcium-permeable AMPARs, specifically at CA3/CA1 synapses. However, the possibility that chronic treatment with antidepressants actually results in strengthened glutamatergic neurotransmission in CA1 has poorly been investigated. Here, we studied whether chronic treatment with the multimodal antidepressant drug trazodone mimicked the effect of paroxetine on the expression of AMPAR subunits in male wistar rat hippocampus and whether these drugs produced a parallel facilitation of field excitatory postsynaptic potentials (fEPSP) responses evoked by activation of CA3/CA1 synapses in dorsal hippocampal slices. In addition, we investigated whether the quality of glutamatergic AMPARs involved in basal neurotransmission was changed by altered subunit expression, e.g. leading to appearance of calcium-permeable AMPARs. We found a significant increase in GluA2 subunit expression following treatment with trazodone or paroxetine for twenty-one days, but not after seven-days treatment. In contrast, we did not find any significant changes in fEPSP responses supporting either a facilitation of glutamatergic neurotransmission in basal conditions or the appearance of functional calcium-permeable AMPARs at CA3/CA1 pyramidal neuron synapses. Thus, neurochemically-detected increases in the expression of AMPAR subunits cannot directly be extrapolated in increased number of functioning receptors and/or facilitated basal neurotransmission.
Collapse
Affiliation(s)
- Elisabetta Gerace
- Department of NEUROFARBA, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy; Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| | | | | | - Elisa Zianni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Gabriella Stocca
- Department of NEUROFARBA, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | | | - Renato Corradetti
- Department of NEUROFARBA, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
9
|
Kouba BR, Torrá ACNC, Camargo A, Rodrigues ALS. The antidepressant-like effect elicited by vitamin D 3 is associated with BDNF/TrkB-related synaptic protein synthesis. Metab Brain Dis 2023; 38:601-611. [PMID: 36350480 DOI: 10.1007/s11011-022-01115-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
Vitamin D3 (cholecalciferol) has been shown to exert antidepressant-like responses, but the role BDNF/TrkB-related synaptic plasticity in this effect remains to be established. Thus, this study investigated the time-course antidepressant-like response of vitamin D3 in female and male mice and the possible role of BDNF/TrkB signaling in this response. The repeated (7 and 21 days), but not acute (60 min), administration of vitamin D3 (2.5 μg/kg, p.o.) exerted an antidepressant-like effect in female and male mice subjected to the tail suspension test, without altering the basal locomotor activity in the open-field test. Notably, vitamin D3 caused a similar time-dependent antidepressant-like effect in male and female mice, suggesting that this behavioral response in the tail suspension test might not be affected by sex differences. Vitamin D3 administration for 21 days, but not for 7 days or 1 h, augmented BDNF levels in the hippocampus and prefrontal cortex of mice. No effects on phospho-CREB/CREB levels were detected in the hippocampus and prefrontal cortex after chronic vitamin D3 administration. Additionally, vitamin D3 increased TrkB, GluA1, and PSD-95 levels in the prefrontal cortex, but not in the hippocampus. Furthermore, an upregulation of synapsin level was observed in both brain regions after vitamin D3 administration. These findings reinforce and extend the notion that vitamin D3 is effective to produce antidepressant-like responses in male and female mice and provide novel evidence that this effect could be associated with BDNF/TrkB-related synaptic protein synthesis. Finally, vitamin D3 could be a feasible nutritional strategy for the management of depression.
Collapse
Affiliation(s)
- Bruna R Kouba
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Ana Clara N C Torrá
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
10
|
Filipović D, Costina V, Findeisen P, Inta D. Fluoxetine Enhances Synaptic Vesicle Trafficking and Energy Metabolism in the Hippocampus of Socially Isolated Rats. Int J Mol Sci 2022; 23:ijms232315351. [PMID: 36499675 PMCID: PMC9735484 DOI: 10.3390/ijms232315351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic social isolation (CSIS)-induced alternation in synaptic and mitochondrial function of specific brain regions is associated with major depressive disorder (MDD). Despite the wide number of available medications, treating MDD remains an important challenge. Although fluoxetine (Flx) is the most frequently prescribed antidepressant, its mode of action is still unknown. To delineate affected molecular pathways of depressive-like behavior and identify potential targets upon Flx treatment, we performed a comparative proteomic analysis of hippocampal purified synaptic terminals (synaptosomes) of rats exposed to six weeks of CSIS, an animal model of depression, and/or followed by Flx treatment (lasting three weeks of six-week CSIS) to explore synaptic protein profile changes. Results showed that Flx in controls mainly induced decreased expression of proteins involved in energy metabolism and the redox system. CSIS led to increased expression of proteins that mainly participate in Ca2+/calmodulin-dependent protein kinase II (Camk2)-related neurotransmission, vesicle transport, and ubiquitination. Flx treatment of CSIS rats predominantly increased expression of proteins involved in synaptic vesicle trafficking (exocytosis and endocytosis), and energy metabolism (glycolytic and mitochondrial respiration). Overall, these Flx-regulated changes in synaptic and mitochondrial proteins of CSIS rats might be critical targets for new therapeutic development for the treatment of MDD.
Collapse
Affiliation(s)
- Dragana Filipović
- Department of Molecular Biology and Endocrinology, “VINČA”, Institute of Nuclear Sciences—National Institute of thе Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence: ; Tel./Fax: +381-(11)-6455-561
| | - Victor Costina
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, 68159 Mannhem, Germany
| | - Peter Findeisen
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, 68159 Mannhem, Germany
| | - Dragos Inta
- Department for Community Health Faculty of Natural Sciences, Medicine University of Fribourg, 1700 Fribourg, Switzerland
- Department of Biomedicine, University of Basel, 4052 Basel, Switzerland
| |
Collapse
|
11
|
Deng D, Cui Y, Gan S, Xie Z, Cui S, Cao K, Wang S, Shi G, Yang L, Bai S, Shi Y, Liu Z, Zhao J, Zhang R. Sinisan alleviates depression-like behaviors by regulating mitochondrial function and synaptic plasticity in maternal separation rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154395. [PMID: 36103769 DOI: 10.1016/j.phymed.2022.154395] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Sinisan (SNS) consists of four kinds of herbs, which is the core of antidepressant prescription widely used in traditional Chinese medicine clinic treatment for depression induced by early life stress. However, the role and precise mechanism of SNS antidepressant have not yet been elucidated. PURPOSE This study aimed to investigate the mechanism SNS on antidepressant of regulating mitochondrial function to improve hippocampal synaptic plasticity. METHODS 90 Sprague-Dawley (SD) rats male pups on Post-Natal Day (PND) 0 were randomly divided into Control group (ddH20), Model group (ddH20), Fluoxetine group (5.0 mg/kg fluoxetine), and SNS-L group (2.5 g/kg SNS), SNS-M group (5.0 g/kg SNS) and SNS-H group (10.0 g/kg SNS), 15 animals per group. Maternal separation (MS) from PND1 to PND21, drug intervention from PND60 to PND90, and behavior tests including sucrose preference test, open field test and forced swimming test from PND83 to PND90 were performed. Synaptic structure and mitochondrial structure were observed by TEM. The expression levels of PSD-95 and SYN were detected by immunohistochemistry and western blot test, the adenosine triphosphate (ATP) content in the hippocampus was detected by assay kits, and the expression levels of Mfn2, Drp1 and Fis1 protein were detected by western bolt test. RESULTS SNS can alleviate depression-like and anxiety-like behaviors in MS rats, improve the damage of synapses and mitochondria, reduce the decrease of ATP in hippocampus, and reverse the expression levels of PSD-95, SYN, Mfn2, Drp1, and Fis1 proteins. CONCLUSION SNS reduced the risk of early life stress induced depression disorder via regulating mitochondrial function and synaptic plasticity. Targeting mitochondrial may be a novel prospective therapeutic avenue for antidepressant.
Collapse
Affiliation(s)
- Di Deng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongfei Cui
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shu Gan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zedan Xie
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sainan Cui
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kerun Cao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shanshan Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoqi Shi
- School of Foreign Studies, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Yang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinlan Zhao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Rong Zhang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
12
|
Kim H, Kim D, Cho Y, Kim K, Roh JD, Kim Y, Yang E, Kim SS, Ahn S, Kim H, Kang H, Bae Y, Kim E. Early postnatal serotonin modulation prevents adult-stage deficits in Arid1b-deficient mice through synaptic transcriptional reprogramming. Nat Commun 2022; 13:5051. [PMID: 36030255 PMCID: PMC9420115 DOI: 10.1038/s41467-022-32748-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 08/12/2022] [Indexed: 11/19/2022] Open
Abstract
Autism spectrum disorder is characterized by early postnatal symptoms, although little is known about the mechanistic deviations that produce them and whether correcting them has long-lasting preventive effects on adult-stage deficits. ARID1B, a chromatin remodeler implicated in neurodevelopmental disorders, including autism spectrum disorder, exhibits strong embryonic- and early postnatal-stage expression. We report here that Arid1b-happloinsufficient (Arid1b+/-) mice display autistic-like behaviors at juvenile and adult stages accompanied by persistent decreases in excitatory synaptic density and transmission. Chronic treatment of Arid1b+/- mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first three postnatal weeks prevents synaptic and behavioral deficits in adults. Mechanistically, these rescues accompany transcriptomic changes, including upregulation of FMRP targets and normalization of HDAC4/MEF2A-related transcriptional regulation of the synaptic proteins, SynGAP1 and Arc. These results suggest that chronic modulation of serotonergic receptors during critical early postnatal periods prevents synaptic and behavioral deficits in adult Arid1b+/- mice through transcriptional reprogramming.
Collapse
Affiliation(s)
- Hyosang Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yisul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Kyungdeok Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yangsik Kim
- Graduate School of Biomedical Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Esther Yang
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Seong Soon Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Sunjoo Ahn
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Korea
| | - Yongchul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea.
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea.
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea.
| |
Collapse
|
13
|
Liang X, Tang J, Qi YQ, Luo YM, Yang CM, Dou XY, Jiang L, Xiao Q, Zhang L, Chao FL, Zhou CN, Tang Y. Exercise more efficiently regulates the maturation of newborn neurons and synaptic plasticity than fluoxetine in a CUS-induced depression mouse model. Exp Neurol 2022; 354:114103. [PMID: 35525307 DOI: 10.1016/j.expneurol.2022.114103] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 11/24/2022]
Abstract
Depression, a common and important cause of morbidity and mortality worldwide, is commonly treated with antidepressants, electric shock and psychotherapy. Recently, increasing evidence has shown that exercise can effectively alleviate depression. To determine the difference in efficacy between exercise and the classic antidepressant fluoxetine in treating depression, we established four groups: the Control, chronic unpredictable stress (CUS/STD), running (CUS/RUN) and fluoxetine (CUS/FLX) groups. The sucrose preference test (SPT), the forced swimming test (FST), the tail suspension test (TST), immunohistochemistry, immunofluorescence and stereological analyses were used to clarify the difference in therapeutic efficacy and mechanism between exercise and fluoxetine in the treatment of depression. In the seventh week, the sucrose preference of the CUS/RUN group was significantly higher than that of the CUS/STD group, while the sucrose preference of the CUS/FLX group did not differ from that of the CUS/STD group until the eighth week. Exercise reduced the immobility time in the FST and TST, while fluoxetine only reduced immobility time in the TST. Hippocampal structure analysis showed that the CUS/STD group exhibited an increase in immature neurons and a decrease in mature neurons. Exercise reduced the number of immature neurons and increased the number of mature neurons, but no increase in the number of mature neurons was observed after fluoxetine treatment. In addition, both running and fluoxetine reversed the decrease in the number of MAP2+ dendrites in depressed mice. Exercise increased the number of spinophilin-positive (Sp+) dendritic spines in the hippocampal CA1, CA3, and dentate gyrus (DG) regions, whereas fluoxetine only increased the number of SP+ spines in the DG. In summary, exercise promoted newborn neuron maturation in the DG and regulated neuronal plasticity in three hippocampal subregions, which might explain why running exerts earlier and more comprehensive antidepressant effects than fluoxetine.
Collapse
Affiliation(s)
- Xin Liang
- Department of Pathophysiology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Ying-Qiang Qi
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan-Min Luo
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Physiology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Chun-Mao Yang
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Xiao-Yun Dou
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Qian Xiao
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Radioactive Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Lei Zhang
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Feng-Lei Chao
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Chun-Ni Zhou
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yong Tang
- Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
14
|
Electroacupuncture Ameliorates Depression-Like Behaviour in Rats by Enhancing Synaptic Plasticity via the GluN2B/CaMKII/CREB Signalling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2146001. [PMID: 34777532 PMCID: PMC8580672 DOI: 10.1155/2021/2146001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/11/2021] [Indexed: 12/27/2022]
Abstract
Background Hippocampal synaptic plasticity during the pathological process of depression has received increasing attention. Hippocampal neuron atrophy and the reduction in synaptic density induced by chronic stress are important pathological mechanisms of depression. Electroacupuncture (EA) exerts beneficial effects on depression, but the mechanism is unclear. This study explored the effect of EA on synaptic plasticity and the potential mechanism. Methods Forty-eight SD rats were randomly divided into the control, chronic unpredictable mild stress (CUMS), EA, and fluoxetine (FLX) groups, and each group consisted of 12 rats. The sucrose preference test, open field test, and forced swimming test were used for the evaluation of depression-like behaviour, and Golgi and Nissl staining were used for the assessment of synaptic plasticity. Western blotting and immunofluorescence were conducted to detect proteins related to synaptic plasticity and to determine their effects on signalling pathways. Results We found that CUMS led to depression-like behaviours, including a reduced preference for sucrose, a prolonged immobility time, and reduced exploration activity. The dendritic spine densities and neuron numbers and the protein levels of MAP-2, PSD-95, and SYN were decreased in the hippocampi of rats with CUMS-induced depression, and these trends were reversed by EA. The molecular mechanism regulating this plasticity may involve the GluN2B/CaMKII/CREB signalling pathway. Conclusion These results suggest that EA can improve depression-like behaviour and hippocampal plasticity induced by CUMS, and the mechanism may be related to the GluN2B/CaMKII/CREB pathway.
Collapse
|
15
|
Yin YY, Wang YH, Liu WG, Yao JQ, Yuan J, Li ZH, Ran YH, Zhang LM, Li YF. The role of the excitation:inhibition functional balance in the mPFC in the onset of antidepressants. Neuropharmacology 2021; 191:108573. [PMID: 33945826 DOI: 10.1016/j.neuropharm.2021.108573] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Currently available antidepressants, such as selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs), generally require weeks to months to produce a therapeutic response, but the mechanism of action underlying the delayed onset of antidepressant-like action remains to be elucidated. The balance between excitatory glutamatergic pyramidal neurons and inhibitory γ-aminobutyric acid (GABA) interneurons, i.e., the excitation:inhibition functional (E:I) balance, in the medial prefrontal cortex (mPFC) is critical in regulating several behaviors and might play an important mediating role in the mechanism of rapid antidepressant-like action reported by several studies. In the present study, the multichannel electrophysiological technique was used to record the firing activities of pyramidal neurons and interneurons and investigate the effects of a single dose of fluoxetine and ketamine (both 10 mg/kg, i.p.) on the E:I functional balance in the rat mPFC after 90 min or 24 h, and the forced swimming test (FST) was used to evaluate the antidepressant-like effects of fluoxetine and ketamine. The present study also explored the effects of chronic treatment with fluoxetine (10 mg/kg, i.g.) for 7 d or 21 d on the E:I functional balance in the mPFC. The present results suggested that a single dose of ketamine could both significantly increase the firing activities of pyramidal neurons and significantly decrease the firing activities of interneurons in the mPFC and exerted significant antidepressant-like action on the FST after 90 min and 24 h, but fluoxetine had no such effects under the same conditions. However, chronic treatment with fluoxetine for 21 d (but not 7 d) could significantly affect the firing activities of pyramidal neurons and interneurons in the mPFC. Taken together, the present results indicated that rapid regulation of the E:I functional balance in the mPFC might be an important common mechanism of rapid-acting antidepressants and the delayed onset of SSRIs might be partly attributed to their inability to rapidly regulate the E:I functional balance in the mPFC. The present study provided a new entry point to the development of rapid-acting antidepressants.
Collapse
Affiliation(s)
- Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yun-Hui Wang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | | | - Jun-Qi Yao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Jin Yuan
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Ze-Han Li
- Capital Normal University High School, Beijing, China
| | - Yu-Hua Ran
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China; Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|
16
|
Li CX, Zheng Y, Zhu H, Li CW, He Z, Wang C, Ding JH, Hu G, Lu M. β-arrestin 2 is essential for fluoxetine-mediated promotion of hippocampal neurogenesis in a mouse model of depression. Acta Pharmacol Sin 2021; 42:679-690. [PMID: 33526871 PMCID: PMC8115338 DOI: 10.1038/s41401-020-00576-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Over the last decade, the roles of β-arrestins in the treatment of neuropsychological diseases have become increasingly appreciated. Fluoxetine is the first selective serotonin reuptake inhibitor developed and is approved for the clinical treatment of depression. Emerging evidence suggests that fluoxetine can directly combine with the 5-HT receptor, which is a member of the G protein-coupled receptor (GPCR) family, in addition to suppressing the serotonin transporter. In this study, we prepared a chronic mild stress (CMS)-induced depression model with β-arrestin2-/- mice and cultured adult neural stem cells (ANSCs) to investigate the involvement of the 5-HT receptor-β-arrestin axis in the pathogenesis of depression and in the therapeutic effect of fluoxetine. We found that β-arrestin2 deletion abolished the fluoxetine-mediated improvement in depression-like behaviors and monoamine neurotransmitter levels, although β-arrestin2 knockout did not aggravate CMS-induced changes in mouse behaviors and neurotransmitters. Notably, the β-arrestin2-/- mice had a shortened dendritic length and reduced dendritic spine density, as well as decreased neural precursor cells, compared to the WT mice under both basal and CMS conditions. We further found that β-arrestin2 knockout decreased the number of proliferating cells in the hippocampal dentate gyrus and suppressed the proliferative capability of ANSCs in vitro. Moreover, β-arrestin2 knockout aggravated the impairment of cell proliferation induced by corticosterone and further blocked the fluoxetine-mediated promotion of mouse hippocampal neurogenesis. Mechanistically, we found that the 5-HT2BR-β-arrestin2-PI3K/Akt axis is essential to maintain the modulation of hippocampal neurogenesis in depressed mice. Our study may provide a promising target for the development of new antidepressant drugs.
Collapse
Affiliation(s)
- Chen-Xin Li
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying Zheng
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng-Wu Li
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhang He
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
- Neuroprotective Drug Discovery Key Laboratory, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
17
|
An Extracellular Perspective on CNS Maturation: Perineuronal Nets and the Control of Plasticity. Int J Mol Sci 2021; 22:ijms22052434. [PMID: 33670945 PMCID: PMC7957817 DOI: 10.3390/ijms22052434] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
During restricted time windows of postnatal life, called critical periods, neural circuits are highly plastic and are shaped by environmental stimuli. In several mammalian brain areas, from the cerebral cortex to the hippocampus and amygdala, the closure of the critical period is dependent on the formation of perineuronal nets. Perineuronal nets are a condensed form of an extracellular matrix, which surrounds the soma and proximal dendrites of subsets of neurons, enwrapping synaptic terminals. Experimentally disrupting perineuronal nets in adult animals induces the reactivation of critical period plasticity, pointing to a role of the perineuronal net as a molecular brake on plasticity as the critical period closes. Interestingly, in the adult brain, the expression of perineuronal nets is remarkably dynamic, changing its plasticity-associated conditions, including memory processes. In this review, we aimed to address how perineuronal nets contribute to the maturation of brain circuits and the regulation of adult brain plasticity and memory processes in physiological and pathological conditions.
Collapse
|
18
|
Casarotto PC, Girych M, Fred SM, Kovaleva V, Moliner R, Enkavi G, Biojone C, Cannarozzo C, Sahu MP, Kaurinkoski K, Brunello CA, Steinzeig A, Winkel F, Patil S, Vestring S, Serchov T, Diniz CRAF, Laukkanen L, Cardon I, Antila H, Rog T, Piepponen TP, Bramham CR, Normann C, Lauri SE, Saarma M, Vattulainen I, Castrén E. Antidepressant drugs act by directly binding to TRKB neurotrophin receptors. Cell 2021; 184:1299-1313.e19. [PMID: 33606976 PMCID: PMC7938888 DOI: 10.1016/j.cell.2021.01.034] [Citation(s) in RCA: 411] [Impact Index Per Article: 102.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 12/22/2020] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
It is unclear how binding of antidepressant drugs to their targets gives rise to the clinical antidepressant effect. We discovered that the transmembrane domain of tyrosine kinase receptor 2 (TRKB), the brain-derived neurotrophic factor (BDNF) receptor that promotes neuronal plasticity and antidepressant responses, has a cholesterol-sensing function that mediates synaptic effects of cholesterol. We then found that both typical and fast-acting antidepressants directly bind to TRKB, thereby facilitating synaptic localization of TRKB and its activation by BDNF. Extensive computational approaches including atomistic molecular dynamics simulations revealed a binding site at the transmembrane region of TRKB dimers. Mutation of the TRKB antidepressant-binding motif impaired cellular, behavioral, and plasticity-promoting responses to antidepressants in vitro and in vivo. We suggest that binding to TRKB and allosteric facilitation of BDNF signaling is the common mechanism for antidepressant action, which may explain why typical antidepressants act slowly and how molecular effects of antidepressants are translated into clinical mood recovery.
Collapse
Affiliation(s)
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Senem M Fred
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Vera Kovaleva
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | - Rafael Moliner
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Giray Enkavi
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Caroline Biojone
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Katja Kaurinkoski
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | | | - Anna Steinzeig
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Frederike Winkel
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Sudarshan Patil
- Department of Biomedicine and KG Jebsen Center for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Stefan Vestring
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tsvetan Serchov
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cassiano R A F Diniz
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paul, Brazil
| | - Liina Laukkanen
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Iseline Cardon
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Brain Master Program, Faculty of Science, Aix-Marseille Université, Marseille, France; Department of Psychiatry, University of Regensburg, Regenburg, Germany
| | - Hanna Antila
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tomasz Rog
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Timo Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Center for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Claus Normann
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Basics in Neuromodulation (NeuroModul Basics), University of Freiburg, Freiburg, Germany
| | - Sari E Lauri
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland; Computational Physics Laboratory, Tampere University, Tampere, Finland
| | - Eero Castrén
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
19
|
Yi LT, Zhu JX, Dong SQ, Chen M, Li CF. Berberine exerts antidepressant-like effects via regulating miR-34a-synaptotagmin1/Bcl-2 axis. CHINESE HERBAL MEDICINES 2021; 13:116-123. [PMID: 36117760 PMCID: PMC9476493 DOI: 10.1016/j.chmed.2020.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 10/13/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
|
20
|
Yoo DY, Jung HY, Kim W, Hahn KR, Kwon HJ, Nam SM, Chung JY, Yoon YS, Kim DW, Hwang IK. Entacapone Treatment Modulates Hippocampal Proteins Related to Synaptic Vehicle Trafficking. Cells 2020; 9:cells9122712. [PMID: 33352833 PMCID: PMC7765944 DOI: 10.3390/cells9122712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 11/29/2022] Open
Abstract
Entacapone, a reversible inhibitor of catechol-O-methyl transferase, is used for patients in Parkinson’s disease because it increases the bioavailability and effectiveness of levodopa. In the present study, we observed that entacapone increases novel object recognition and neuroblasts in the hippocampus. In the present study, two-dimensional electrophoresis (2-DE) and matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry were performed to compare the abundance profiles of proteins expressed in the hippocampus after entacapone treatment in mice. Results of 2-DE, MALDI-TOF mass spectrometry, and subsequent proteomic analysis revealed an altered protein expression profile in the hippocampus after entacapone treatment. Based on proteomic analysis, 556 spots were paired during the image analysis of 2-DE gels and 76 proteins were significantly changed more than two-fold among identified proteins. Proteomic analysis indicated that treatment with entacapone induced expressional changes in proteins involved in synaptic transmission, cellular processes, cellular signaling, the regulation of cytoskeletal structure, energy metabolism, and various subcellular enzymatic reactions. In particular, entacapone significantly increased proteins related to synaptic trafficking and plasticity, such as dynamin 1, synapsin I, and Munc18-1. Immunohistochemical staining showed the localization of the proteins, and western blot confirmed the significant increases in dynamin I (203.5% of control) in the hippocampus as well as synapsin I (254.0% of control) and Munc18-1 (167.1% of control) in the synaptic vesicle fraction of hippocampus after entacapone treatment. These results suggest that entacapone can enhance hippocampal synaptic trafficking and plasticity against various neurological diseases related to hippocampal dysfunction.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea;
| | - Sung Min Nam
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Iksan 54538, Korea;
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea;
- Correspondence: (D.W.K.); (I.K.H.)
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (D.Y.Y.); (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Correspondence: (D.W.K.); (I.K.H.)
| |
Collapse
|
21
|
Zeitler S, Schumacher F, Monti J, Anni D, Guhathakurta D, Kleuser B, Friedland K, Fejtová A, Kornhuber J, Rhein C. Acid Sphingomyelinase Impacts Canonical Transient Receptor Potential Channels 6 (TRPC6) Activity in Primary Neuronal Systems. Cells 2020; 9:E2502. [PMID: 33218173 PMCID: PMC7698877 DOI: 10.3390/cells9112502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 11/13/2020] [Indexed: 12/30/2022] Open
Abstract
: The acid sphingomyelinase (ASM)/ceramide system exhibits a crucial role in the pathology of major depressive disorder (MDD). ASM hydrolyzes the abundant membrane lipid sphingomyelin to ceramide that regulates the clustering of membrane proteins via microdomain and lipid raft organization. Several commonly used antidepressants, such as fluoxetine, rely on the functional inhibition of ASM in terms of their antidepressive pharmacological effects. Transient receptor potential canonical 6 (TRPC6) ion channels are located in the plasma membrane of neurons and serve as receptors for hyperforin, a phytochemical constituent of the antidepressive herbal remedy St. John's wort. TRPC6 channels are involved in the regulation of neuronal plasticity, which likely contributes to their antidepressant effect. In this work, we investigated the impact of reduced ASM activity on the TRPC6 function in neurons. A lipidomic analysis of cortical brain tissue of ASM deficient mice revealed a decrease in ceramide/sphingomyelin molar ratio and an increase in sphingosine. In neurons with ASM deletion, hyperforin-mediated Ca2+-influx via TRPC6 was decreased. Consequently, downstream activation of nuclear phospho-cAMP response element-binding protein (pCREB) was changed, a transcriptional factor involved in neuronal plasticity. Our study underlines the importance of balanced ASM activity, as well as sphingolipidome composition for optimal TRPC6 function. A better understanding of the interaction of the ASM/ceramide and TRPC6 systems could help to draw conclusions about the pathology of MDD.
Collapse
Affiliation(s)
- Stefanie Zeitler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Fabian Schumacher
- Department of Toxicology, University of Potsdam, 14558 Nuthetal, Germany;
- Department of Pharmacology & Toxicology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
- Institute of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Juliana Monti
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Daniela Anni
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Burkhard Kleuser
- Department of Pharmacology & Toxicology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
| | - Kristina Friedland
- Institute for Pharmacy and Biochemistry, Johannes-Gutenberg Universität Mainz, 55128 Mainz, Germany;
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Cosima Rhein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
- Department of Psychosomatic Medicine and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
22
|
Yi LT, Zhu JX, Dong SQ, Li CF, Zhang QP, Cheng J, Liu Q. miR-34a induces spine damages via inhibiting synaptotagmin-1 in depression. Neurobiol Stress 2020; 13:100243. [PMID: 33344699 PMCID: PMC7739037 DOI: 10.1016/j.ynstr.2020.100243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 11/24/2022] Open
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that participate in the pathophysiology of depression by targeting many functional genes. As shown in our previous study, chronic stress up-regulates miR-34a in the hippocampus. However, little is known about the mechanism by which miR-34a regulates the process of depression or its functions as an antidepressant by regulating its targets. In the present study, the dynamic alterations in miR-34a expression and the mechanism underlying miR-34a regulation were assessed after the administration of the antidepressant fluoxetine to mice exposed to chronic stress. In addition, the effects of miR-34a inhibition on mice were directly evaluated. Both lipopolysaccharide (LPS) and corticosterone treatment caused depression-like symptoms and increased miR-34a expression. Additionally, the expression of miR-34a, which was regulated by tropomyosin receptor kinase B (TrkB)/MEK1/ERK signaling, was consistent with the onset of action of fluoxetine. A luciferase reporter assay identified synaptotagmin-1 and Bcl-2 as the targets of miR-34a. Moreover, a miR-34a antagomir exerted antidepressant-like effects, activated TrkB/MEK1/ERK signaling and improved spine morphology in the hippocampus. In conclusion, hippocampal miR-34a overexpression was a typical feature in depression-like animals, and miR-34a downregulation exerts antidepressant-like effects by restoring the spine morphology through its target synaptotagmin-1.
Collapse
Affiliation(s)
- Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| | - Ji-Xiao Zhu
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi province, PR China
| | - Shu-Qi Dong
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| | - Cheng-Fu Li
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361009, Fujian province, PR China
| | - Qiu-Ping Zhang
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361009, Fujian province, PR China
| | - Jie Cheng
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| | - Qing Liu
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| |
Collapse
|
23
|
Hisaoka-Nakashima K, Yokoe T, Watanabe S, Nakamura Y, Kajitani N, Okada-Tsuchioka M, Takebayashi M, Nakata Y, Morioka N. Lysophosphatidic acid induces thrombospondin-1 production in primary cultured rat cortical astrocytes. J Neurochem 2020; 158:849-864. [PMID: 33118159 DOI: 10.1111/jnc.15227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/09/2020] [Accepted: 10/25/2020] [Indexed: 11/25/2022]
Abstract
Lysophosphatidic acid (LPA), a brain membrane-derived lipid mediator, plays important roles including neural development, function, and behavior. In the present study, the effects of LPA on astrocyte-derived synaptogenesis factor thrombospondins (TSPs) production were examined by real-time PCR and western blotting, and the mechanism underlying this event was examined by pharmacological approaches in primary cultured rat cortical astrocytes. Treatment of astrocytes with LPA increased TSP-1 mRNA, and TSP-2 mRNA, but not TSP-4 mRNA expression. TSP-1 protein expression and release were also increased by LPA. LPA-induced TSP-1 production were inhibited by AM966 a LPA1 receptor antagonist, and Ki16425, LPA1/3 receptors antagonist, but not by H2L5146303, LPA2 receptor antagonist. Pertussis toxin, Gi/o inhibitor, but not YM-254890, Gq inhibitor, and NF499, Gs inhibitor, inhibited LPA-induced TSP-1 production, indicating that LPA increases TSP-1 production through Gi/o-coupled LPA1 and LPA3 receptors. LPA treatment increased phosphorylation of extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (JNK). LPA-induced TSP-1 mRNA expression was inhibited by U0126, MAPK/ERK kinase (MEK) inhibitor, but not SB202190, p38 MAPK inhibitor, or SP600125, JNK inhibitor. However, LPA-induced TSP-1 protein expression was diminished with inhibition of all three MAPKs, indicating that these signaling molecules are involved in TSP-1 protein production. Treatment with antidepressants, which bind to astrocytic LPA1 receptors, increased TSP-1 mRNA and protein production. The current findings show that LPA/LPA1/3 receptors signaling increases TSP-1 production in astrocytes, which could be important in the pathogenesis of affective disorders and could potentially be a target for the treatment of affective disorders.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Toshiki Yokoe
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shintaro Watanabe
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Naoto Kajitani
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Mami Okada-Tsuchioka
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Minoru Takebayashi
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Yoshihiro Nakata
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
24
|
Zavvari F, Nahavandi A. Fluoxetine increases hippocampal neural survival by improving axonal transport in stress-induced model of depression male rats. Physiol Behav 2020; 227:113140. [PMID: 32828030 DOI: 10.1016/j.physbeh.2020.113140] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Axonal transport deficit is a key mechanism involved in neurodegenerative conditions. Fluoxetine, a commonly used antidepressant for treatment of depression, is known to regulate several important structural and neurochemical aspects of hippocampal functions. However, the mechanisms underlying these effects are still poorly understood. This study aimed to investigate the effects of chronic fluoxetine treatment on axonal transport in the hippocampus of rat stress-induced model of depression. METHODS We have analyzed the effects of chronic fluoxetine treatment (20 mg/kg/day, 24 days) on immobility behavior (forced swimming test), hippocampal iNOS (inflammatory factor) expression (RT-PCR) as well as hippocampal BDNF, kinesin and dynein expression (RT-PCR) and hippocampal neuronal survival (Nissl staining). RESULTS This study provided evidence that fluoxetine could effectively suppress iNOS expression following unpredictable chronic mild stress (P < 0.01), increase hippocampal BDNF (P < 0.01), kinesin (P < 0.05) and dynein (P < 0.01) gene expression, and control neuronal death in CA1 (P < 0.01) and CA3 regions (P < 0.01) of the hippocampus and thereby improve immobility behavior (P < 0.001). CONCLUSION Based on the findings of this study, we concluded the neuroprotective effect of fluoxetine may be due to its ability to improve axonal transmission, followed by increased energy supply and neurotrophin concentration and function.
Collapse
Affiliation(s)
- Fahime Zavvari
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Arezo Nahavandi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran; Neuroscience Research Center, Iran University of Medical Science, Tehran, Iran.
| |
Collapse
|
25
|
Protective Mechanism and Treatment of Neurogenesis in Cerebral Ischemia. Neurochem Res 2020; 45:2258-2277. [PMID: 32794152 DOI: 10.1007/s11064-020-03092-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Stroke is the fifth leading cause of death worldwide and is a main cause of disability in adults. Neither currently marketed drugs nor commonly used treatments can promote nerve repair and neurogenesis after stroke, and the repair of neurons damaged by ischemia has become a research focus. This article reviews several possible mechanisms of stroke and neurogenesis and introduces novel neurogenic agents (fibroblast growth factors, brain-derived neurotrophic factor, purine nucleosides, resveratrol, S-nitrosoglutathione, osteopontin, etc.) as well as other treatments that have shown neuroprotective or neurogenesis-promoting effects.
Collapse
|
26
|
Shen C, Cao K, Cui S, Cui Y, Mo H, Wen W, Dong Z, Lin H, Bai S, Yang L, Zhang R, Shi Y. SiNiSan ameliorates depression-like behavior in rats by enhancing synaptic plasticity via the CaSR-PKC-ERK signaling pathway. Biomed Pharmacother 2020; 124:109787. [DOI: 10.1016/j.biopha.2019.109787] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/25/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
|
27
|
Wu Z, Wang G, Wang H, Xiao L, Wei Y, Yang C. Fluoxetine exposure for more than 2 days decreases the neuronal plasticity mediated by CRMP2 in differentiated PC12 cells. Brain Res Bull 2020; 158:99-107. [PMID: 32070769 DOI: 10.1016/j.brainresbull.2020.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/22/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Recent studies indicate that antidepressants treatment restores neuronal plasticity. In contrast, some researchers claim that serotonergic antidepressants, including fluoxetine (FLU), may exacerbate neuronal plasticity, which is contradictory and rarely studied. Since almost those studies exposed cells with drugs for 1-2 days as treatment models of antidepressants, it is possible that FLU exposure for longer periods would have opposite effects on neuronal plasticity. RESULTS In the present study, we examined the effects of FLU exposure (up to 3 days) on the neuronal plasticity in differentiated PC12 cells. The cell viability shown a slight decrease at day 2 (93.5 ± 3.5 %), followed by a highly significant decrease at day 3(71.4 ± 4.4 %). As previously reported, neuronal plasticity was significantly upregulated by FLU exposure at day 1. However, the neurite length, activity-regulated cytoskeleton-associated protein (Arc) and c-Fos mRNA were inhibited with FLU exposure at day 3. Similarly, the expression of tubulin, which play important roles in the neuronal plasticity, was the same result. Furthermore, we found α-tubulin interacted with collapsing response mediator protein 2(CRMP2), which is related to neuronal plasticity, and the regulation of CRMP2 activity influenced the neurite length, Arc, c-Fos and tubulin expression. CONCLUSIONS The results demonstrated that neuronal plasticity was increased by FLU exposure at day 1, but exposure with FLU for more than 2 days had opposite effect on it. The reduction in neuronal plasticity with FLU exposure for more than 2 days might be involved in some aspects of the therapeutic effect of antidepressant on depression.
Collapse
Affiliation(s)
- Zuotian Wu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Yanyan Wei
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Can Yang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| |
Collapse
|
28
|
Ye Q, Zeng C, Luo C, Wu Y. Ferrostatin-1 mitigates cognitive impairment of epileptic rats by inhibiting P38 MAPK activation. Epilepsy Behav 2020; 103:106670. [PMID: 31864943 DOI: 10.1016/j.yebeh.2019.106670] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 12/27/2022]
Abstract
Evidence indicates that ferrostain-1 (Fer-1), a specific inhibitor of ferroptosis, could ameliorate cognitive dysfunction of rats with kainic acid (KA)-induced temporal lobe epilepsy (TLE) by suppressing ferroptosis processes. Recent studies suggest that P38 mitogen-activated protein kinase (MAPK) pathway could be mediated by ferroptosis processes. The activation of P38 MAPK results in cognitive impairment by suppressing the expression of synaptic plasticity-related proteins. However, it is unclear whether Fer-1 can mitigate cognitive impairment of rats with KA-induced TLE by inhibiting P38 MAPK activation. In the present study, treatment with Fer-1 blocked the activation of P38 MAPK, which resulted in an increased expression of synaptophysin (SYP) and postsynaptic density protein 95 (PSD-95) in the hippocampus of rats with KA-induced TLE, hence, ameliorating their cognitive impairment. Also, P38 MAPK activation in the hippocampus of the rats reduced the expression of both PSD-95 and SYP proteins. Treatment of the rats with SB203580, a P38 MAPK-specific inhibitor, prevented the activation of P38 MAPK, which resulted in an increase in SYP and PSD95 protein levels in the hippocampus. These results suggest that Fer-1 could mitigate the cognitive impairment by suppressing P38 MAPK activation thus restoring the expression of synaptic proteins. Ferroptosis processes might be involved in suppressing synaptic protein expression.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, 6th Shuangyong Road, Nanning, China; Department of Neurology, The First Affiliated Hospital of University of South China, 69th Chuanshan Road, Hengyang, China
| | - Chunmei Zeng
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, 6th Shuangyong Road, Nanning, China
| | - Chun Luo
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, 262th East Mingxiu Road, Nanning, Guangxi, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, 6th Shuangyong Road, Nanning, China.
| |
Collapse
|
29
|
Fluoxetine attenuates stress-induced depressive-like behavior through modulation of hippocampal GAP43 and neurogenesis in male rats. J Chem Neuroanat 2019; 103:101711. [PMID: 31705954 DOI: 10.1016/j.jchemneu.2019.101711] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/20/2019] [Accepted: 11/03/2019] [Indexed: 11/21/2022]
Abstract
Based on the monoaminergic theory, Serotonin-Selective Reuptake Inhibitors (SSRIs) are used for treating depression. Recent hypotheses suggest that antidepressants may influence neurogenesis and synaptic plasticity. However, the mechanisms underlying these effects are still poorly understood. The aim of the present study was to evaluate the effect of fluoxetine, a widely used SSRI antidepressant, on the neurogenesis and the expression of Growth-Associated Protein 43 (GAP43), a synaptic protein, in the rat hippocampus exposed to Unpredictable Chronic Mild Stress (UCMS; the model of depressive-like behavior). We have analyzed the effects of chronic fluoxetine treatment on immobility behavior (forced swimming test), plasma interleukin-6 and corticosterone (enzyme-linked immunosorbent assay), BrdU-positive cells in the dentate gyrus and GAP43 expression in the CA3 region (Immunohistochemistry) of the hippocampus. This study provides evidence that fluoxetine is a potent enhancer of GAP-43, a protein related to the neuronal plasticity, in the hippocampus of the rat model of depression. Interestingly, our results showed that although fluoxetine significantly is effective in increasing BrdU positive cells, it is more effective in increasing the neurite formation compared with neurogenesis. The results support the idea that antidepressants can promote neuronal plasticity. We concluded that the increase of GAP-43- induced neurite formation may be an important mechanism by which fluoxetine augments hippocampal neuroplasticity and play its pivotal antidepressant role.
Collapse
|
30
|
Van Dyke AM, Francis TC, Chen H, Bailey AM, Thompson SM. RETRACTED: Chronic fluoxetine treatment in vivo enhances excitatory synaptic transmission in the hippocampus. Neuropharmacology 2019; 150:38-45. [PMID: 30851310 PMCID: PMC6475886 DOI: 10.1016/j.neuropharm.2019.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/07/2019] [Accepted: 03/02/2019] [Indexed: 12/17/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Authors. After publication, Scott M. Thompson found significant concerns about the data and duly notified The University of Maryland. The University of Maryland conducted an internal investigation which confirmed that the article was compromised. Namely in Figure 2B, the Investigation Committee determined that the western blots used to create the figure were not the ones used for the quantification and concluded that the figure was falsified to fit the hypothesis. In Figure 2C and 2D, the Investigation Committee determined that the densitometry data (pCaMKII, pS831, CamKII and GluA1) used to create the histogram were falsified to fit the hypothesis.
Collapse
Affiliation(s)
- Adam M Van Dyke
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Training Programs in Neuroscience, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - T Chase Francis
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Training Programs in Neuroscience, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Haiwen Chen
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Training Programs in Neuroscience, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Medical Scientist Training Program, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Aileen M Bailey
- Department of Psychology, Saint Mary's College of Maryland, St. Mary's City, MD, USA
| | - Scott M Thompson
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
31
|
Abstract
Selective serotonin reuptake inhibitors are among the most prescribed antidepressants. Fluoxetine is the lead molecule which exerts its therapeutic effects, at least in part, by promoting neuroplasticity through increased brain-derived neurotrophic factor (BDNF)/tropomyosin-related receptor kinase B (TrkB) signalling. It is unclear however, to which extent the neuroplastic effects of fluoxetine are solely mediated by the inhibition of the serotonin transporter (5-HTT). To answer this question, the effects of fluoxetine on neuroplasticity were analysed in both wild type (WT) and 5-Htt knock-out (KO) mice. Using Western blotting and RT-qPCR approaches, we showed that fluoxetine 10 µM activated BDNF/TrkB signalling pathways in both CD1 and C57BL/6J mouse primary cortical neurons. Interestingly, effects on BDNF signalling were observed in primary cortical neurons from both 5-Htt WT and KO mice. In addition, a 3-week in vivo fluoxetine treatment (15 mg/kg/d; i.p.) increased the expression of plasticity genes in brains of both 5-Htt WT and KO mice, and tended to equally enhance hippocampal cell proliferation in both genotypes, without reaching significance. Our results further suggest that fluoxetine-induced neuroplasticity does not solely depend on 5-HTT blockade, but might rely, at least in part, on 5-HTT-independent direct activation of TrkB.
Collapse
|
32
|
Sanacora G, Katz R. Ketamine: A Review for Clinicians. FOCUS: JOURNAL OF LIFE LONG LEARNING IN PSYCHIATRY 2018; 16:243-250. [PMID: 31975918 DOI: 10.1176/appi.focus.20180012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A growing series of clinical trials and case series now suggest that ketamine-originally used as an anesthetic agent-potentially offers an exciting new treatment option for severe depression. Increasing numbers of studies show that ketamine can provide prompt relief for many depressed patients, including those with severe treatment-refractory depression. Although the effects of a single treatment are commonly short-lived, multiple infusion protocols may offer sustained relief. The uniquely rapid onset of antidepressant action raises the potential for ketamine use in a variety of clinical situations, including the prevention or shortening of hospital stays, the treatment of acute suicidal ideation, and the facilitation of medication crossovers. Ketamine, in combination with other multimodal treatment approaches, including psychotherapy, may further augment response effect and duration. Promises of efficacy have led to increasingly unbridled use to treat a variety of psychiatric disorders, with diverse approaches and treatment environments, despite inadequate data demonstrating the true clinical efficacy and safety of the various protocols or a thorough understanding of mechanisms of action. This article briefly reviews the history of ketamine's development as a potential antidepressant, current hypotheses related to its mechanisms of action, and existing evidence for its safety and efficacy with a focus on clinicians' interests.
Collapse
Affiliation(s)
- Gerard Sanacora
- Drs. Sanacora and Katz are both with the Yale Department of Psychiatry, Yale University, New Haven CT, and the Yale Interventional Psychiatry Service, Yale New Haven Psychiatric Hospital, New Haven, CT
| | - Rachel Katz
- Drs. Sanacora and Katz are both with the Yale Department of Psychiatry, Yale University, New Haven CT, and the Yale Interventional Psychiatry Service, Yale New Haven Psychiatric Hospital, New Haven, CT
| |
Collapse
|
33
|
Inhibition of Phosphodiesterase 4 by FCPR03 Alleviates Lipopolysaccharide-Induced Depressive-Like Behaviors in Mice: Involvement of p38 and JNK Signaling Pathways. Int J Mol Sci 2018; 19:ijms19020513. [PMID: 29419799 PMCID: PMC5855735 DOI: 10.3390/ijms19020513] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 01/06/2023] Open
Abstract
Inflammatory responses induced by peripheral administration of lipopolysaccharide (LPS) triggers depressive-like behavioral syndrome in rodents. Inhibition of phosphodiesterase 4 (PDE4) produces a robust anti-inflammatory effect in inflammatory cells. Unfortunately, archetypal PDE4 inhibitors cause intolerable gastrointestinal side-effects, such as vomiting and nausea. N-isopropyl-3-(cyclopropylmethoxy)-4-difluoromethoxy benzamide (FCPR03) is a novel, selective PDE4 inhibitor with little, or no, emetic potency. Our previous studies show that FCPR03 is effective in attenuating neuroinflammation in mice treated with LPS. However, whether FCPR03 could exert antidepressant-like effect induced by LPS is largely unknown. In the present study, mice injected intraperitoneally (i.p.) with LPS was established as an in vivo animal model of depression. The antidepressant-like activities of FCPR03 were evaluated using a tail suspension test, forced swimming test, and sucrose preference test. We demonstrated that administration of FCPR03 (1 mg/kg) produced antidepressant-like effects in mice challenged by LPS, as evidenced by decreases in the duration of immobility in the forced swim and tail suspension tests, while no significant changes in locomotor activity were observed. FCPR03 also increased sucrose preference in mice treated with LPS. In addition, treatment with FCPR03 abolished the downregulation of brain-derived neurotrophic factor induced by LPS and decreased the level of corticosterone in plasma. Meanwhile, periphery immune challenge by LPS induced enhanced phosphorylation of p38-mitogen activated protein kinase (p38) and c-Jun N-terminal kinase (JNK) in both the cerebral cortex and hippocampus in mice. Interestingly, treatment with FCPR03 significantly blocked the role of LPS and reduced the levels of phosphorylated p38 and JNK. Collectively, these results indicate that FCPR03 shows antidepressant-like effects in mice challenged by LPS, and the p38/JNK signaling pathway is possibly involved in this process. Our findings suggest that FCPR03 is a potential compound for the prevention or treatment of depression.
Collapse
|