1
|
Loan Nguyen TM, Guilloux JP, Defaix C, Mendez-David I, Etting I, Alvarez JC, McGowan JC, Highland JN, Zanos P, Lovett J, Moaddel R, Corruble E, David DJ, Gould TD, Denny CA, Gardier AM. Ketamine metabolism via hepatic CYP450 isoforms contributes to its sustained antidepressant actions. Neuropharmacology 2024; 258:110065. [PMID: 39004413 PMCID: PMC11492263 DOI: 10.1016/j.neuropharm.2024.110065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/03/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024]
Abstract
(R,S)-ketamine (ketamine) has rapid and sustained antidepressant (AD) efficacy at sub-anesthetic doses in depressed patients. A metabolite of ketamine, including (2R,6R)-hydroxynorketamine ((6)-HNKs) has been reported to exert antidepressant actions in rodent model of anxiety/depression. To further understand the specific role of ketamine's metabolism in the AD actions of the drug, we evaluated the effects of inhibiting hepatic cytochrome P450 enzymes on AD responses. We assessed whether pre-treatment with fluconazole (10 and 20 mg/kg, i. p.) 1 h prior to ketamine or HNKs (10 mg/kg, i. p.) administration would alter behavioral and neurochemical actions of the drugs in male BALB/cJ mice with a highly anxious phenotype. Extracellular microdialysate levels of glutamate and GABA (Gluext, GABAext) were also measured in the medial prefrontal cortex (mPFC). Pre-treatment with fluconazole altered the pharmacokinetic profile of ketamine, by increasing both plasma and brain levels of ketamine and (R,S)-norketamine, while robustly reducing those of (6)-HNKs. At 24 h post-injection (t24 h), fluconazole prevented the sustained AD-like response of ketamine responses in the forced swim test and splash test, as well as the enhanced cortical GABA levels produced by ketamine. A single (2R,6R)-HNK administration resulted in prevention of the effects of fluconazole on the antidepressant-like activity of ketamine in mice. Overall, these findings are consistent with an essential contribution of (6)-HNK to the sustained antidepressant-like effects of ketamine and suggest potential interactions between pharmacological CYPIs and ketamine during antidepressant treatment in patients.
Collapse
Affiliation(s)
- Thi Mai Loan Nguyen
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Jean-Philippe Guilloux
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Céline Defaix
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Indira Mendez-David
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Isabelle Etting
- Service de Pharmacologie-Toxicologie, Hôpital Raymond Poincaré, Groupe Hospitalier Universitaires AP-HP, Université Paris-Saclay, Inserm U-1018, CESP, MOODS Team, 92380 Garches, France
| | - Jean-Claude Alvarez
- Service de Pharmacologie-Toxicologie, Hôpital Raymond Poincaré, Groupe Hospitalier Universitaires AP-HP, Université Paris-Saclay, Inserm U-1018, CESP, MOODS Team, 92380 Garches, France
| | - Josephine C McGowan
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA; Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) /New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Jaclyn N Highland
- Departments of Psychiatry, Baltimore, MD, USA; Departments of Program in Toxicology, University of Maryland School of Medicine, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Panos Zanos
- Departments of Psychiatry, Baltimore, MD, USA; Departments of Pharmacology, Baltimore, MD, USA; Departments of Physiology, Baltimore, MD, USA; Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA; Department of Psychology, University of Cyprus, Nicosia, 2109, Cyprus
| | - Jacqueline Lovett
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Emmanuelle Corruble
- Université Paris-Saclay, Faculté de Médecine, Inserm UMR 1018, CESP, MOODS Team, 94270 Bicêtre Hospital, 94270 Le Kremin-Bicêtre, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, 94270 Le Kremlin Bicêtre, France
| | - Denis J David
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Todd D Gould
- Departments of Psychiatry, Baltimore, MD, USA; Departments of Pharmacology, Baltimore, MD, USA; Departments of Physiology, Baltimore, MD, USA; Departments of Neurobiology, Baltimore, MD, USA; Departments of Program in Toxicology, University of Maryland School of Medicine, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA; Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Christine A Denny
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA; Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) /New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Alain M Gardier
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France.
| |
Collapse
|
2
|
Rayan NA, Aow J, Lim MGL, Arcego DM, Ryan R, Nourbakhsh N, de Lima RMS, Craig K, Zhang TY, Goh YT, Sun AX, Tompkins T, Bronner S, Binda S, Diorio J, Parent C, Meaney MJ, Prabhakar S. Shared and unique transcriptomic signatures of antidepressant and probiotics action in the mammalian brain. Mol Psychiatry 2024; 29:3653-3668. [PMID: 38844534 DOI: 10.1038/s41380-024-02619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 11/08/2024]
Abstract
Understanding the shared and divergent mechanisms across antidepressant (AD) classes and probiotics is critical for improving treatment for mood disorders. Here we examine the transcriptomic effects of bupropion (NDRI), desipramine (SNRI), fluoxetine (SSRI) and a probiotic formulation (Lacidofil®) on 10 regions across the mammalian brain. These treatments massively alter gene expression (on average, 2211 differentially expressed genes (DEGs) per region-treatment combination), highlighting the biological complexity of AD and probiotic action. Intersection of DEG sets against neuropsychiatric GWAS loci, sex-specific transcriptomic portraits of major depressive disorder (MDD), and mouse models of stress and depression reveals significant similarities and differences across treatments. Interestingly, molecular responses in the infralimbic cortex, basolateral amygdala and locus coeruleus are region-specific and highly similar across treatments, whilst responses in the Raphe, medial preoptic area, cingulate cortex, prelimbic cortex and ventral dentate gyrus are predominantly treatment-specific. Mechanistically, ADs concordantly downregulate immune pathways in the amygdala and ventral dentate gyrus. In contrast, protein synthesis, metabolism and synaptic signaling pathways are axes of variability among treatments. We use spatial transcriptomics to further delineate layer-specific molecular pathways and DEGs within the prefrontal cortex. Our study reveals complex AD and probiotics action on the mammalian brain and identifies treatment-specific cellular processes and gene targets associated with mood disorders.
Collapse
Affiliation(s)
- Nirmala Arul Rayan
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Jonathan Aow
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Gek Liang Lim
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Danusa Mar Arcego
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Richard Ryan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Nooshin Nourbakhsh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | | | - Kelly Craig
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Tie Yuan Zhang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Alfred Xuyang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857, Singapore
| | - Thomas Tompkins
- Lallemand Bio-Ingredients, 1620 Rue Prefontaine, Montréal, QC, H1W 2N8, Canada
| | - Stéphane Bronner
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Sylvie Binda
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Josie Diorio
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Carine Parent
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Michael J Meaney
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada.
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.
- Brain-Body Initiative, Institute for Cell & Molecular Biology, A*STAR, Singapore, Singapore.
| | - Shyam Prabhakar
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
3
|
Ben-Azu B, Oritsemuelebi B, Oghorodi AM, Adebesin A, Isibor H, Eduviere AT, Otuacha OS, Akudo M, Ekereya S, Maidoh IF, Iyayi JO, Uzochukwu-Godfrey FC. Psychopharmacological interaction of alcohol and posttraumatic stress disorder: Effective action of naringin. Eur J Pharmacol 2024; 978:176791. [PMID: 38944175 DOI: 10.1016/j.ejphar.2024.176791] [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: 05/01/2024] [Revised: 06/09/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Posttraumatic stress disorder (PTSD) and alcohol use disorder (AUD) are prevalently co-occurring, important risk factors for a broad array of neuropsychiatric diseases. To date, how these two contrastive concomitant pairs increase the risk of neuropsychiatric states, notably exacerbating PTSD-related symptoms, remains unknown. Moreover, pharmacological interventions with agents that could reverse PTSD-AUD comorbidity, however, remained limited. Hence, we investigated the neuroprotective actions of naringin in mice comorbidly exposed to PTSD followed by repeated ethanol (EtOH)-induced AUD. Following a 7-day single-prolong-stress (SPS)-induced PTSD in mice, binge/heavy drinking, notably related to AUD, was induced in the PTSD mice with every-other-day ethanol (2 g/kg, p.o.) administration, followed by daily treatments with naringin (25 and 50 mg/kg) or fluoxetine (10 mg/kg), from days 8-21. PTSD-AUD-related behavioral changes, alcohol preference, hypothalamic-pituitary-adrenal (HPA)-axis dysfunction-induced neurochemical alterations, oxidative/nitrergic stress, and inflammation were examined in the prefrontal-cortex, striatum, and hippocampus. PTSD-AUD mice showed aggravated anxiety, spatial-cognitive, social impairments and EtOH intake, which were abated by naringin, similar to fluoxetine. Our assays on the HPA-axis showed exacerbated increased corticosterone release and adrenal hypertrophy, accompanied by marked dopamine and serotonin increase, with depleted glutamic acid decarboxylase enzyme in the three brain regions, which naringin, however, reversed, respectively. PTSD-AUD mice also showed increased TNF-α, IL-6, malondialdehyde and nitrite levels, with decreased antioxidant elements in the prefrontal-cortex, striatum, and hippocampus compared to SPS-EtOH-mice, mainly exacerbating catalase and glutathione decrease in the hippocampus relative SPS-mice. These findings suggest that AUD exacerbates PTSD pathologies in different brain regions, notably comprising neurochemical dysregulations, oxidative/nitrergic and cytokine-mediated inflammation, with HPA dysfunction, which were, however, revocable by naringin.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria.
| | - Benjamin Oritsemuelebi
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Akpobo M Oghorodi
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria; Department of Biomedical Engineering, Faculty of Technology, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Adaeze Adebesin
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria
| | - Happy Isibor
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Anthony T Eduviere
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Oghenemine S Otuacha
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Moses Akudo
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Surhirime Ekereya
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Isioma F Maidoh
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Joy O Iyayi
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Faith C Uzochukwu-Godfrey
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| |
Collapse
|
4
|
Floris G, Godar SC, Braccagni G, Piras IS, Ravens A, Zanda MT, Huentelman MJ, Bortolato M. The sinking platform test: a novel paradigm to measure persistence in animal models. Neuropsychopharmacology 2024; 49:1373-1382. [PMID: 38396257 PMCID: PMC11251282 DOI: 10.1038/s41386-024-01827-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024]
Abstract
Persistence is the propensity to maintain goal-directed actions despite adversities. While this temperamental trait is crucial to mitigate depression risk, its neurobiological foundations remain elusive. Developing behavioral tasks to capture persistence in animal models is crucial for understanding its molecular underpinnings. Here, we introduce the Sinking Platform Test (SPT), a novel high-throughput paradigm to measure persistence. Mice were trained to exit a water-filled tank by ascending onto a platform above water level. Throughout the training, mice were also occasionally exposed to "failure trials," during which an operator would submerge a platform right after the mouse climbed onto it, requiring the mouse to reach and ascend a newly introduced platform. Following training, mice were subjected to a 5-min test exclusively consisting of failure trials. Male and female mice exhibited comparable persistence, measured by the number of climbed platforms during the test. Furthermore, this index was increased by chronic administration of fluoxetine or imipramine; conversely, it was reduced by acute and chronic haloperidol. Notably, six weeks of social isolation reduced SPT performance, and this effect was rescued by imipramine treatment over the last two weeks. A 4-week regimen of voluntary wheel running also improved persistence in socially isolated mice. Finally, comparing transcriptomic profiles of the prefrontal cortex of mice with high and low SPT performance revealed significant enrichment of immediate-early genes known to shape susceptibility for chronic stress. These findings highlight the potential of SPT as a promising method to uncover the biological mechanisms of persistence and evaluate novel interventions to enhance this response.
Collapse
Affiliation(s)
- Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Sean C Godar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Giulia Braccagni
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Alicia Ravens
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Mary T Zanda
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
5
|
Aouci R, Fontaine A, Vion A, Belz L, Levi G, Narboux-Nême N. The Antidepressant Action of Fluoxetine Involves the Inhibition of Dlx5/6 in Cortical GABAergic Neurons through a TrkB-Dependent Pathway. Cells 2024; 13:1262. [PMID: 39120293 PMCID: PMC11311550 DOI: 10.3390/cells13151262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Major depressive disorder (MDD) is a complex and devastating illness that affects people of all ages. Despite the large use of antidepressants in current medical practice, neither their mechanisms of action nor the aetiology of MDD are completely understood. Experimental evidence supports the involvement of Parvalbumin-positive GABAergic neurons (PV-neurons) in the pathogenesis of MDD. DLX5 and DLX6 (DLX5/6) encode two homeodomain transcription factors involved in cortical GABAergic differentiation and function. In the mouse, the level of expression of these genes is correlated with the cortical density of PV-neurons and with anxiety-like behaviours. The same genomic region generates the lncRNA DLX6-AS1, which, in humans, participates in the GABAergic regulatory module downregulated in schizophrenia and ASD. Here, we show that the expression levels of Dlx5/6 in the adult mouse brain are correlated with the immobility time in the forced swim test, which is used to measure depressive-like behaviours. We show that the administration of the antidepressant fluoxetine (Flx) to normal mice induces, within 24 h, a rapid and stable reduction in Dlx5, Dlx6 and Dlx6-AS1 expression in the cerebral cortex through the activation of the TrkB-CREB pathway. Experimental Dlx5 overexpression counteracts the antidepressant effects induced by Flx treatment. Our findings show that one of the short-term effects of Flx administration is the reduction in Dlx5/6 expression in GABAergic neurons, which, in turn, has direct consequences on PV expression and on behavioural profiles. Variants in the DLX5/6 regulatory network could be implicated in the predisposition to depression and in the variability of patients' response to antidepressant treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicolas Narboux-Nême
- Molecular Physiology and Adaption, UMR7221 CNRS, Museum National d’Histoire Naturelle, 75005 Paris, France; (R.A.); (A.F.); (L.B.); (G.L.)
| |
Collapse
|
6
|
Filipović D, Inderhees J, Korda A, Tadić P, Schwaninger M, Inta D, Borgwardt S. Serum Metabolites as Potential Markers and Predictors of Depression-like Behavior and Effective Fluoxetine Treatment in Chronically Socially Isolated Rats. Metabolites 2024; 14:405. [PMID: 39195501 DOI: 10.3390/metabo14080405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Metabolic perturbation has been associated with depression. An untargeted metabolomics approach using liquid chromatography-high resolution mass spectrometry was employed to detect and measure the rat serum metabolic changes following chronic social isolation (CSIS), an animal model of depression, and effective antidepressant fluoxetine (Flx) treatment. Univariate and multivariate statistics were used for metabolic data analysis and differentially expressed metabolites (DEMs) determination. Potential markers and predictive metabolites of CSIS-induced depressive-like behavior and Flx efficacy in CSIS were evaluated by the receiver operating characteristic (ROC) curve, and machine learning (ML) algorithms, such as support vector machine with linear kernel (SVM-LK) and random forest (RF). Upregulated choline following CSIS may represent a potential marker of depressive-like behavior. Succinate, stachydrine, guanidinoacetate, kynurenic acid, and 7-methylguanine were revealed as potential markers of effective Flx treatment in CSIS rats. RF yielded better accuracy than SVM-LK (98.50% vs. 85.70%, respectively) in predicting Flx efficacy in CSIS vs. CSIS, however, it performed almost identically in classifying CSIS vs. control (75.83% and 75%, respectively). Obtained DEMs combined with ROC curve and ML algorithms provide a research strategy for assessing potential markers or predictive metabolites for the designation or classification of stress-induced depressive phenotype and mode of drug action.
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
- Bioanalytic Core Facility, 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
- Bioanalytic Core Facility, Center of Brain Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Dragoš Inta
- Department for Community Health, Faculty of Natural Sciences, Medicine, University of Fribourg, 1700 Fribourg, Switzerland
- Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| | - Stefan Borgwardt
- Department of Psychiatry and Psychotherapy, Center of Brain Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
7
|
Chaisanam R, Wattanathorn J, Thukham-mee W, Piyavhatkul N, Paholpak P. Anxiolytic, Antidepression, and Memory-Enhancing Effects of the Novel Instant Soup RJ6601 in the Middle-Aged of Female Rats. Foods 2024; 13:2170. [PMID: 39063255 PMCID: PMC11276534 DOI: 10.3390/foods13142170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Due to the health benefits of polyphenols and dietary fiber in combating mental disorders, we hypothesized that a polyphenol- and dietary fiber-enriched soup (RJ6601) would improve mental wellness in a rat model of middle-aged women. To test this hypothesis, female Wistar rats aged 18 months (350-450 g) were orally administered RJ6601 at doses of 200 and 400 mg/kg BW for 28 days. The anxiolytic, antidepression, and memory-enhancing effects were assessed every 7 days throughout the study period. The neuron density and levels of activities of AChE, total MAO, MAO-A, MAO-B, MDA, SOD, CAT, GSH-Px, IL-1β, IL-6, and BDNF in the prefrontal cortex at the end of study were also investigated. Furthermore, the amounts of Lactobacillus spp. and Bifidobacterium spp. in their feces were also determined. The results revealed that the developed soup shows anxiolytic, antidepression, and memory-enhancing effects. An increased neuron density; reductions in AChE, total MAO, MAO-A, MAO-B, and MDA; and an elevation of serum BDNF, together with increased amounts of both bacterial species in feces, were also observed. Our results suggest that RJ6601 is a potential mental wellness promotion supplement that enhances BDNF levels, brain plasticity, neurotransmitter balance, and oxidative stress and inflammation status, along with improving microbiota.
Collapse
Affiliation(s)
- Rujikan Chaisanam
- Department of Physiology and Graduate School (Neuroscience Program), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Jintanaporn Wattanathorn
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Research Institute for High Human Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wipawee Thukham-mee
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Research Institute for High Human Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nawanant Piyavhatkul
- Department Psychiatry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (N.P.); (P.P.)
| | - Pongsatorn Paholpak
- Department Psychiatry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (N.P.); (P.P.)
| |
Collapse
|
8
|
Chan D, Baker KD, Richardson R. The impact of chronic fluoxetine treatment in adolescence or adulthood on context fear memory and perineuronal nets. Dev Psychobiol 2024; 66:e22501. [PMID: 38807259 DOI: 10.1002/dev.22501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024]
Abstract
Selective serotonin reuptake inhibitors, such as fluoxetine (Prozac), are commonly prescribed pharmacotherapies for anxiety. Fluoxetine may be a useful adjunct because it can reduce the expression of learned fear in adult rodents. This effect is associated with altered expression of perineuronal nets (PNNs) in the amygdala and hippocampus, two brain regions that regulate fear. However, it is unknown whether fluoxetine has similar effects in adolescents. Here, we investigated the effect of fluoxetine exposure during adolescence or adulthood on context fear memory and PNNs in the basolateral amygdala (BLA), the CA1 subregion of the hippocampus, and the medial prefrontal cortex in rats. Fluoxetine impaired context fear memory in adults but not in adolescents. Further, fluoxetine increased the number of parvalbumin (PV)-expressing neurons surrounded by a PNN in the BLA and CA1, but not in the medial prefrontal cortex, at both ages. Contrary to previous reports, fluoxetine did not shift the percentage of PNNs toward non-PV cells in either the BLA or CA1 in the adults, or adolescents. These findings demonstrate that fluoxetine differentially affects fear memory in adolescent and adult rats but does not appear to have age-specific effects on PNNs.
Collapse
Affiliation(s)
- Diana Chan
- School of Psychology, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kathryn D Baker
- School of Psychology, UNSW Sydney, Sydney, New South Wales, Australia
| | - Rick Richardson
- School of Psychology, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
9
|
Ben-Azu B, Adebesin A, Moke GE, Ojiokor VO, Olusegun A, Jarikre TA, Akinluyi ET, Olukemi OA, Omeiza NA, Nkenchor P, Niemogha AR, Ewere ED, Igwoku C, Omamogho F. Alcohol exacerbates psychosocial stress-induced neuropsychiatric symptoms: Attenuation by geraniol. Neurochem Int 2024; 177:105748. [PMID: 38703789 DOI: 10.1016/j.neuint.2024.105748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Adaptation to psychosocial stress is psychologically distressing, initiating/promoting comorbidity with alcohol use disorders. Emerging evidence moreover showed that ethanol (EtOH) exacerbates social-defeat stress (SDS)-induced behavioral impairments, neurobiological sequelae, and poor therapeutic outcomes. Hence, this study investigated the effects of geraniol, an isoprenoid monoterpenoid alcohol with neuroprotective functions on EtOH escalated SDS-induced behavioral impairments, and neurobiological sequelae in mice. Male mice chronically exposed to SDS for 14 days were repeatedly fed with EtOH (2 g/kg, p. o.) from days 8-14. From days 1-14, SDS-EtOH co-exposed mice were concurrently treated with geraniol (25 and 50 mg/kg) or fluoxetine (10 mg/kg) orally. After SDS-EtOH translational interactions, arrays of behavioral tasks were examined, followed by investigations of oxido-inflammatory, neurochemicals levels, monoamine oxidase-B and acetylcholinesterase activities in the striatum, prefrontal-cortex, and hippocampus. The glial fibrillary acid protein (GFAP) expression was also quantified in the prefrontal-cortex immunohistochemically. Adrenal weights, serum glucose and corticosterone concentrations were measured. EtOH exacerbated SDS-induced low-stress resilience, social impairment characterized by anxiety, depression, and memory deficits were attenuated by geraniol (50 and 100 mg/kg) and fluoxetine. In line with this, geraniol increased the levels of dopamine, serotonin, and glutamic-acid decarboxylase enzyme, accompanied by reduced monoamine oxidase-B and acetylcholinesterase activities in the prefrontal-cortex, hippocampus, and striatum. Geraniol inhibited SDS-EtOH-induced adrenal hypertrophy, corticosterone, TNF-α, IL-6 release, malondialdehyde and nitrite levels, with increased antioxidant activities. Immunohistochemical analyses revealed that geraniol enhanced GFAP immunoreactivity in the prefrontal-cortex relative to SDS-EtOH group. We concluded that geraniol ameliorates SDS-EtOH interaction-induced behavioral changes via normalization of neuroimmune-endocrine and neurochemical dysregulations in mice brains.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria.
| | - Adaeze Adebesin
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Abafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Segamu Campus, Ogun State, Nigeria
| | - Goodes E Moke
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Vivian O Ojiokor
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Medicine, Enugu State University of Science and Technology (ESUT), Enugu, Enugu State, Nigeria
| | - Adebayo Olusegun
- Neurophysiology Unit, Department of Physiology, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port-Harcourt, River State, Nigeria
| | - Thiophilus A Jarikre
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Elizabeth T Akinluyi
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado- Ekiti, Nigeria
| | - Opajobi A Olukemi
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Noah A Omeiza
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria; Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Paul Nkenchor
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Avwenayeri R Niemogha
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Ejaita D Ewere
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Chioma Igwoku
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Favour Omamogho
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| |
Collapse
|
10
|
Kong CH, Lee JW, Jeon M, Kang WC, Kim MS, Park K, Bae HJ, Park SJ, Jung SY, Kim SN, Kleinfelter B, Kim JW, Ryu JH. D-Pinitol mitigates post-traumatic stress disorder-like behaviors induced by single prolonged stress in mice through mineralocorticoid receptor antagonism. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110990. [PMID: 38467326 DOI: 10.1016/j.pnpbp.2024.110990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/24/2024] [Accepted: 03/06/2024] [Indexed: 03/13/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a mental illness that can occur in individuals who have experienced trauma. Current treatments for PTSD, typically serotonin reuptake inhibitors, have limited effectiveness for patients and often cause serious adverse effects. Therefore, a novel class of treatment with better pharmacological profile is necessary. D-Pinitol has been reported to be effective for depression and anxiety disorders, but there are no reports associated with PTSD. In the present study, we investigated the effects of D-pinitol in a mouse model of PTSD induced by a single prolonged stress (SPS) protocol. We examined the therapeutic effects of D-pinitol on emotional and cognitive impairments in the SPS mouse model. We also investigated the effects of D-pinitol on fear memory formation. Mineralocorticoid receptor transactivation assay, Western blot, and quantitative PCR were employed to investigate how D-pinitol exerts its pharmacological activities. D-Pinitol ameliorated PTSD-like behaviors in a SPS mouse model. D-Pinitol also normalized the increased mRNA expression levels and protein levels of the mineralocorticoid receptor in the amygdala. A mineralocorticoid receptor agonist reversed the effects of D-pinitol on fear extinction and recall, and the antagonistic property of D-pinitol against the mineralocorticoid receptor was confirmed in vitro. Our findings suggest that D-pinitol could serve as a potential therapeutic agent for PTSD due to its antagonistic effect on the mineralocorticoid receptor.
Collapse
Affiliation(s)
- Chang Hyeon Kong
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin Woo Lee
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung-si 25451, Republic of Korea
| | - Mijin Jeon
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woo Chang Kang
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min Seo Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Keontae Park
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ho Jung Bae
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seo Yun Jung
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Su-Nam Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung-si 25451, Republic of Korea
| | - Benjamin Kleinfelter
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN 37240, United States of America
| | - Ji-Woon Kim
- Department of Pharmacy, College of Pharmacy, Kyung Hee Univeristy, Seoul 02447, Republic of Korea.
| | - Jong Hoon Ryu
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
11
|
Mena S, Cruikshank A, Best J, Nijhout HF, Reed MC, Hashemi P. Modulation of serotonin transporter expression by escitalopram under inflammation. Commun Biol 2024; 7:710. [PMID: 38851804 PMCID: PMC11162477 DOI: 10.1038/s42003-024-06240-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/24/2024] [Indexed: 06/10/2024] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are widely used for depression based on the monoamine deficiency hypothesis. However, the clinical use of these agents is controversial, in part because of their variable clinical efficacy and in part because of their delayed onset of action. Because of the complexities involved in replicating human disease and clinical dosing in animal models, the scientific community has not reached a consensus on the reasons for these phenomena. In this work, we create a theoretical hippocampal model incorporating escitalopram's pharmacokinetics, pharmacodynamics (competitive and non-competitive inhibition, and serotonin transporter (SERT) internalization), inflammation, and receptor dynamics. With this model, we simulate chronic oral escitalopram in mice showing that days to weeks are needed for serotonin levels to reach steady-state. We show escitalopram's chemical efficacy is diminished under inflammation. Our model thus offers mechanisms for how chronic escitalopram affects brain serotonin, emphasizing the importance of optimized dose and time for future antidepressant discoveries.
Collapse
Affiliation(s)
- Sergio Mena
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | | | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| | - H F Nijhout
- Department of Biology, Duke University, Durham, NC, USA
| | - Michael C Reed
- Department of Mathematics, Duke University, Durham, NC, USA
| | - Parastoo Hashemi
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
12
|
Piantadosi SC, Manning EE, Chamberlain BL, Hyde J, LaPalombara Z, Bannon NM, Pierson JL, K Namboodiri VM, Ahmari SE. Hyperactivity of indirect pathway-projecting spiny projection neurons promotes compulsive behavior. Nat Commun 2024; 15:4434. [PMID: 38789416 PMCID: PMC11126597 DOI: 10.1038/s41467-024-48331-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Compulsive behaviors are a hallmark symptom of obsessive compulsive disorder (OCD). Striatal hyperactivity has been linked to compulsive behavior generation in correlative studies in humans and causal studies in rodents. However, the contribution of the two distinct striatal output populations to the generation and treatment of compulsive behavior is unknown. These populations of direct and indirect pathway-projecting spiny projection neurons (SPNs) have classically been thought to promote or suppress actions, respectively, leading to a long-held hypothesis that increased output of direct relative to indirect pathway promotes compulsive behavior. Contrary to this hypothesis, here we find that indirect pathway hyperactivity is associated with compulsive grooming in the Sapap3-knockout mouse model of OCD-relevant behavior. Furthermore, we show that suppression of indirect pathway activity using optogenetics or treatment with the first-line OCD pharmacotherapy fluoxetine is associated with reduced grooming in Sapap3-knockouts. Together, these findings highlight the striatal indirect pathway as a potential treatment target for compulsive behavior.
Collapse
Affiliation(s)
- Sean C Piantadosi
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Elizabeth E Manning
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Brittany L Chamberlain
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Hyde
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biology, Southern Arkansas University, Magnolia, AK, USA
| | - Zoe LaPalombara
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas M Bannon
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jamie L Pierson
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Susanne E Ahmari
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Niwa M, Lockhart S, Wood DJ, Yang K, Francis-Oliveira J, Kin K, Ahmed A, Wand GS, Kano SI, Payne JL, Sawa A. Prolonged HPA axis dysregulation in postpartum depression associated with adverse early life experiences: A cross-species translational study. NATURE. MENTAL HEALTH 2024; 2:593-604. [PMID: 38736646 PMCID: PMC11087073 DOI: 10.1038/s44220-024-00217-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/21/2024] [Indexed: 05/14/2024]
Abstract
Childhood and adolescent stress increase the risk of postpartum depression (PPD), often providing an increased probability of treatment refractoriness. Nevertheless, the mechanisms linking childhood/adolescent stress to PPD remain unclear. Our study investigated the longitudinal effects of adolescent stress on the hypothalamic-pituitary-adrenal (HPA) axis and postpartum behaviors in mice and humans. Adolescent social isolation prolonged glucocorticoid elevation, leading to long-lasting postpartum behavioral changes in female mice. These changes were unresponsive to current PPD treatments but improved with post-delivery glucocorticoid receptor antagonist treatment. Childhood/adolescent stress significantly impacted HPA axis dysregulation and PPD in human females. Repurposing glucocorticoid receptor antagonists for some cases of treatment-resistant PPD may be considered.
Collapse
Affiliation(s)
- Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham School of Engineering, Birmingham, AL, USA
| | - Sedona Lockhart
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel J. Wood
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kun Yang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jose Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Kyohei Kin
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Adeel Ahmed
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Gary S. Wand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shin-ichi Kano
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Jennifer L. Payne
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlotte, VA, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
14
|
Zheng Z, Zhou H, Yang L, Zhang L, Guo M. Selective disruption of mTORC1 and mTORC2 in VTA astrocytes induces depression and anxiety-like behaviors in mice. Behav Brain Res 2024; 463:114888. [PMID: 38307148 DOI: 10.1016/j.bbr.2024.114888] [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: 12/11/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Dysfunction of the mechanistic target of rapamycin (mTOR) signaling pathway is implicated in neuropsychiatric disorders including depression and anxiety. Most studies have been focusing on neurons, and the function of mTOR signaling pathway in astrocytes is less investigated. mTOR forms two distinct complexes, mTORC1 and mTORC2, with key scaffolding protein Raptor and Rictor, respectively. The ventral tegmental area (VTA), a vital component of the brain reward system, is enrolled in regulating both depression and anxiety. In the present study, we aimed to examine the regulation effect of VTA astrocytic mTOR signaling pathway on depression and anxiety. We specifically deleted Raptor or Rictor in VTA astrocytes in mice and performed a series of behavioral tests for depression and anxiety. Deletion of Raptor and Rictor both decreased the immobility time in the tail suspension test and the latency to eat in the novelty suppressed feeding test, and increased the horizontal activity and the movement time in locomotor activity. Deletion of Rictor decreased the number of total arm entries in the elevated plus-maze test and the vertical activity in locomotor activity. These data suggest that VTA astrocytic mTORC1 plays a role in regulating depression-related behaviors and mTORC2 is involved in both depression and anxiety-related behaviors. Our results indicate that VTA astrocytic mTOR signaling pathway might be new targets for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Ziteng Zheng
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Han Zhou
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Lu Yang
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Lanlan Zhang
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Ming Guo
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China.
| |
Collapse
|
15
|
Ben-Azu B, Moke EG, Chris-Ozoko LE, Jaiyeoba-Ojigho EJ, Adebayo OG, Ajayi AM, Oyovwi MO, Odjugo G, Omozojie VI, Ejomafuwe G, Onike N, Eneni AEO, Ichipi-Ifukor CP, Achuba IF. Diosgenin alleviates alcohol-mediated escalation of social defeat stress and the neurobiological sequalae. Psychopharmacology (Berl) 2024; 241:785-803. [PMID: 38311692 DOI: 10.1007/s00213-023-06509-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 11/15/2023] [Indexed: 02/06/2024]
Abstract
RATIONALE Emerging evidence indicates that persistent alcohol consumption escalates psychosocial trauma achieved by social defeat stress (SDS)-induced neurobiological changes and behavioral outcomes. Treatment with compounds with neuroprotective functions is believed to reverse ethanol (EtOH)-aggravated SDS-induced behavioral impairments. OBJECTIVES We investigated the outcomes of diosgenin treatment, a phytosteroidal sapogenin in mice co-exposed to repeated SDS and EtOH administration. METHODS During a period of 14 days, SDS male mice were repeatedly administered EtOH (20%, 10 mL/kg) orally from days 8-14 (n = 9). Within days 1-14, SDS mice fed with EtOH were simultaneously treated with diosgenin (25 and 50 mg/kg) or fluoxetine (10 mg/kg) by oral gavage. Locomotor, cognitive-, depressive-, and anxiety-like behaviors were assessed. Adrenal weight, serum glucose, and corticosterone levels were assayed. Brain markers of oxido-inflammatory, neurochemical levels, monoamine oxidase-B, and acetylcholinesterase activities were measured in the striatum, prefrontal cortex, and hippocampus. RESULTS The anxiety-like behavior, depression, low stress resilience, social, and spatial/non-spatial memory decline exhibited by SDS mice exposed to repeated EtOH administration were alleviated by diosgenin (25 and 50 mg/kg) and fluoxetine, illustrated by increased dopamine and serotonin concentrations and reduced monoamine oxidase-B and acetylcholinesterase activities in the prefrontal cortex, hippocampus, and striatum. Diosgenin attenuated SDS + EtOH interaction induced corticosterone release and adrenal hypertrophy, accompanied by reduced TNF-α, IL-6, malondialdehyde, and nitrite levels in the striatum, prefrontal cortex, and hippocampus. Diosgenin increased glutathione, superoxide dismutase, and catalase levels in SDS + EtOH-exposed mice. CONCLUSIONS Our data suggest that diosgenin reverses SDS + EtOH interaction-induced behavioral changes via normalization of hypothalamic-pituitary-adrenal axis, neurochemical neurotransmissions, and inhibition of oxidative and inflammatory mediators in mice brains.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria.
| | - Emuesiri Goodies Moke
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Lilian E Chris-Ozoko
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Efe J Jaiyeoba-Ojigho
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Olusegun G Adebayo
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, Neurophysiology Unit, PAMO University of Medical Sciences, River State, Port-Harcourt, Nigeria
| | - Abayomi Mayowa Ajayi
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Mega O Oyovwi
- Department of Basic Medical Science, Achievers University, Owo, Ondo State, Nigeria
| | - Gideon Odjugo
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Vincent I Omozojie
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Goddey Ejomafuwe
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Nzubechukwu Onike
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Aya-Ebi O Eneni
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | | | - Ifeakachuku F Achuba
- Department of Biochemistry, Faculty of Sciences, Delta State University, Abraka, Delta State, Nigeria
| |
Collapse
|
16
|
Uzungil V, Luza S, Opazo CM, Mees I, Li S, Ang CS, Williamson NA, Bush AI, Hannan AJ, Renoir T. Phosphoproteomics implicates glutamatergic and dopaminergic signalling in the antidepressant-like properties of the iron chelator deferiprone. Neuropharmacology 2024; 246:109837. [PMID: 38184274 DOI: 10.1016/j.neuropharm.2024.109837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
BACKGROUND Current antidepressants have limitations due to insufficient efficacy and delay before improvement in symptoms. Polymorphisms of the serotonin transporter (5-HTT) gene have been linked to depression (when combined with stressful life events) and altered response to selective serotonergic reuptake inhibitors. We have previously revealed the antidepressant-like properties of the iron chelator deferiprone in the 5-HTT knock-out (KO) mouse model of depression. Furthermore, deferiprone was found to alter neural activity in the prefrontal cortex of both wild-type (WT) and 5-HTT KO mice. METHODS In the current study, we examined the molecular effects of acute deferiprone treatment in the prefrontal cortex of both genotypes via phosphoproteomics analysis. RESULTS In WT mice treated with deferiprone, there were 22 differentially expressed phosphosites, with gene ontology analysis implicating cytoskeletal proteins. In 5-HTT KO mice treated with deferiprone, we found 33 differentially expressed phosphosites. Gene ontology analyses revealed phosphoproteins that were predominantly involved in synaptic and glutamatergic signalling. In a drug-naïve cohort (without deferiprone administration), the analysis revealed 21 differentially expressed phosphosites in 5-HTT KO compared to WT mice. We confirmed the deferiprone-induced increase in tyrosine hydroxylase serine 40 residue phosphorylation (pTH-Ser40) (initially revealed in our phosphoproteomics study) by Western blot analysis, with deferiprone increasing pTH-Ser40 expression in WT and 5-HTT KO mice. CONCLUSION As glutamatergic and synaptic signalling are dysfunctional in 5-HTT KO mice (and are the target of fast-acting antidepressant drugs such as ketamine), these molecular effects may underpin deferiprone's antidepressant-like properties. Furthermore, dopaminergic signalling may also be involved in deferiprone's antidepressant-like properties.
Collapse
Affiliation(s)
- Volkan Uzungil
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Isaline Mees
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Nicholas A Williamson
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
17
|
Asim M, Wang H, Chen X, He J. Potentiated GABAergic neuronal activities in the basolateral amygdala alleviate stress-induced depressive behaviors. CNS Neurosci Ther 2024; 30:e14422. [PMID: 37715582 PMCID: PMC10915993 DOI: 10.1111/cns.14422] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/22/2023] [Accepted: 08/14/2023] [Indexed: 09/17/2023] Open
Abstract
AIMS Major depressive disorder is a severe psychiatric disorder that afflicts ~17% of the world population. Neuroimaging investigations of depressed patients have consistently reported the dysfunction of the basolateral amygdala in the pathophysiology of depression. However, how the BLA and related circuits are implicated in the pathogenesis of depression is poorly understood. METHODS Here, we combined fiber photometry, immediate early gene expression (c-fos), optogenetics, chemogenetics, behavioral analysis, and viral tracing techniques to provide multiple lines of evidence of how the BLA neurons mediate depressive-like behavior. RESULTS We demonstrated that the aversive stimuli elevated the neuronal activity of the excitatory BLA neurons (BLACAMKII neurons). Optogenetic activation of CAMKII neurons facilitates the induction of depressive-like behavior while inhibition of these neurons alleviates the depressive-like behavior. Next, we found that the chemogenetic inhibition of GABAergic neurons in the BLA (BLAGABA ) increased the firing frequency of CAMKII neurons and mediates the depressive-like phenotypes. Finally, through fiber photometry recording and chemogenetic manipulation, we proved that the activation of BLAGABA neurons inhibits BLACAMKII neuronal activity and alleviates depressive-like behavior in the mice. CONCLUSION Thus, through evaluating BLAGABA and BLACAMKII neurons by distinct interaction, the BLA regulates depressive-like behavior.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- Department of Biomedical ScienceCity University of Hong KongKowloon TongPeople's Republic of China
| | - Huajie Wang
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- Department of Biomedical ScienceCity University of Hong KongKowloon TongPeople's Republic of China
| | - Xi Chen
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- City University of Hong Kong Shenzhen Research InstituteShenzhenPeople's Republic of China
| | - Jufang He
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- City University of Hong Kong Shenzhen Research InstituteShenzhenPeople's Republic of China
| |
Collapse
|
18
|
Araki R, Kita A, Ago Y, Yabe T. Chronic social defeat stress induces anxiety-like behaviors via downregulation of serotonin transporter in the prefrontal serotonergic system in mice. Neurochem Int 2024; 174:105682. [PMID: 38301899 DOI: 10.1016/j.neuint.2024.105682] [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: 10/20/2023] [Revised: 01/14/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
The serotonergic (5-HTergic) system is closely involved in the pathophysiology of mood and anxiety disorders and the responsibility of this system may differ for each symptom. In this study, we examined the relationship between the dysfunction of the 5-HTergic system and abnormal behaviors in the social defeat stress model, an animal model of mood and anxiety disorders and in mice with knockdown of Slc6a4, the gene encoding SERT. Monoamine content, serotonin (5-HT) release, 5-HT uptake, 5-HT transporter (SERT) protein levels, and behaviors were investigated in mice subjected to chronic social defeat stress and in mice with knockdown of Slc6a4, in 5-HTergic neurons projecting to the prefrontal cortex (PFC). Furthermore, DNA methylation of Slc6a4 was examined in mice subjected to chronic social defeat stress. Increased turnover, increased extracellular basal levels, decreased release and decreased uptake of 5-HT, and decreased SERT protein levels were observed in the PFC of the stressed mice. The decreased 5-HT uptake correlated with anxiety-like behavior characterized by decreased time spent in the open arms of the elevated plus maze. DNA methylation was increased in the CpG island of Slc6a4 in 5-HTergic neurons projecting to the PFC of the stressed mice. Similar to the stressed mice, mice with Slc6a4 knockdown in 5-HTergic neurons projecting to the PFC also showed decreased release and uptake of 5-HT in the PFC and increased anxiety-like behavior. Chronic stress may induce anxiety due to dysfunction in the prefrontal 5-HTergic system via decreased SERT expression in the PFC.
Collapse
Affiliation(s)
- Ryota Araki
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan.
| | - Ayami Kita
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takeshi Yabe
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan.
| |
Collapse
|
19
|
Deyama S, Li X, Duman RS. Neuron-specific deletion of VEGF or its receptor Flk-1 occludes the antidepressant-like effects of desipramine and fluoxetine in mice. Neuropsychopharmacol Rep 2024; 44:246-249. [PMID: 37960997 PMCID: PMC10932798 DOI: 10.1002/npr2.12393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) signaling is known to be involved in the antidepressant-like effects of conventional antidepressants, such as desipramine (DMI), a tricyclic antidepressant, and fluoxetine (FLX), a selective serotonin reuptake inhibitor; however, the precise role of neuronal VEGF signaling in mediating these effects remains unclear. Using mice with excitatory neuron-specific deletion of VEGF and its receptor, fetal liver kinase 1 (Flk-1) in the forebrain, we examined the effects of forebrain excitatory neuron-specific deletion of VEGF or Flk-1 on the antidepressant-like effects of repeated DMI and chronic FLX administration in the forced swim test (FST). Repeated intraperitoneal (i.p.) injections of DMI (10, 10, and 20 mg/kg at 24, 4, and 1 h before the FST, respectively) significantly decreased immobility in control mice; however, this effect was completely blocked in mice with neuron-specific VEGF or Flk-1 deletion. Although chronic treatment with FLX (18 mg/kg/day, i.p.) did not impact immobility in control mice 1 day after the 22nd injection, immobility was significantly reduced 1 day after the preswim and the 23rd FLX injection. However, in mice with neuron-specific Flk-1 deletion, chronic FLX treatment significantly increased immobility in the preswim and failed to produce antidepressant-like effects. Collectively, these findings indicate that neuronal VEGF-Flk-1 signaling contributes to the antidepressant-like actions of conventional antidepressants.
Collapse
Affiliation(s)
- Satoshi Deyama
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | - Xiao‐Yuan Li
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Ronald S. Duman
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
20
|
Lin S, Chen Z, Wu Z, Fei F, Xu Z, Tong Y, Sun W, Wang P. Involvement of PI3K/AKT Pathway in the Rapid Antidepressant Effects of Crocetin in Mice with Depression-Like Phenotypes. Neurochem Res 2024; 49:477-491. [PMID: 37935859 DOI: 10.1007/s11064-023-04051-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/22/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023]
Abstract
The current first-line antidepressants have the drawback of slow onset, which greatly affects the treatment of depression. Crocetin, one of the main active ingredients in saffron (Crocus sativus L.), has been demonstrated to have antidepressant activities, but whether it has a rapid antidepressant effect remains unclear. This study aimed to investigate the onset, duration, and mechanisms of the rapid antidepressant activity of crocetin (20, 40 and 80 mg/kg, intraperitoneal injection) in male mice subjected to chronic restraint stress (CRS). The results of behavioral tests showed that crocetin exerted rapid antidepressant-like effect in mice with depression-like phenotypes, including rapid normalization of depressive-like behaviors within 3 h, and the effects could be maintained for 2 days. Hematoxylin-eosin (HE) and Nissl staining showed that crocetin ameliorated hippocampal neuroinflammation and nerve injuries in mice with depression-like phenotypes. The levels of inflammatory factors, corticosterone and pro brain-derived neurotrophic factor in crocetin-administrated mice serum were significantly reduced compared with those in the CRS group, as well as the levels of inflammatory factors in hippocampus. What's more, Western blot analyses showed that, compared to CRS-induced mice, the relative levels of mitogen-activated kinase phosphatase 1 and toll-like receptor 4 were significantly reduced after the administration of crocetin, and the relative expressions of extracellular signal-regulated kinase 1/2 (ERK1/2), cAMP-response element binding protein, phosphorylated phosphoinositide 3 kinase (p-PI3K)/PI3K, phosphorylated protein kinase B (p-AKT)/AKT, phosphorylated glycogen synthase kinase 3β (p-GSK3β)/GSK3β, phosphorylated mammalian target of rapamycin (p-mTOR)/mTOR were markedly upregulated. In conclusion, crocetin exerted rapid antidepressant effects via suppressing the expression of inflammatory cytokines and the apoptosis of neuronal cells through PI3K/AKT signaling pathways. The rapid antidepressant effect of crocetin (40 mg/kg) could be maintained for at least 2 days after single treatment.
Collapse
Affiliation(s)
- Susu Lin
- The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, People's Republic of China
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
| | - Ziwei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
| | - Zhaoruncheng Wu
- School of Biomedical engineering, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Fei Fei
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
| | - Zijin Xu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
- College of Pharmacy, Jiangxi Medical College, Shangrao, 334000, Jiangxi, People's Republic of China
| | - Yingpeng Tong
- Institute of Natural Medicine and Health Product, School of Advanced Study, Taizhou University, Taizhou, 318000, People's Republic of China
| | - Wenyu Sun
- The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, People's Republic of China.
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China.
| |
Collapse
|
21
|
Lamoureux L, Beverley JA, Marinelli M, Steiner H. Fluoxetine potentiates methylphenidate-induced behavioral responses: Enhanced locomotion or stereotypies and facilitated acquisition of cocaine self-administration. ADDICTION NEUROSCIENCE 2023; 9:100131. [PMID: 38222942 PMCID: PMC10785378 DOI: 10.1016/j.addicn.2023.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The medical psychostimulant methylphenidate (MP) is used to treat attention-deficit hyperactivity disorder and recreationally as a "cognitive enhancer". MP is a dopamine reuptake inhibitor, but does not affect serotonin. Serotonin contributes to addiction-related gene regulation and behavior. Previously, we showed that enhancing serotonin action by adding a selective serotonin reuptake inhibitor, fluoxetine (FLX), to MP potentiates MP-induced gene regulation in striatum and nucleus accumbens, mimicking cocaine effects. Here, we investigated the behavioral consequences of MP+FLX treatment. Young adult male rats received MP (5 mg/kg, i.p.) or MP+FLX (5 mg/kg each) daily for 6-8 days. Behavioral effects were assessed in an open-field test during the repeated treatment. Two weeks later the motor response to a cocaine challenge (25 mg/kg) and the rate of acquisition of cocaine self-administration behavior were determined. Our results demonstrate that FLX potentiates effects of MP on open-field behavior. However, we found differential behavioral responses to MP+FLX treatment, as approximately half of the rats developed high rates of focal stereotypies (termed "MP+FLX/high reactivity" group), whereas the other half did not, and only showed increased locomotion ("MP+FLX/low reactivity" group). Two weeks later, cocaine-induced locomotion and stereotypies were positively correlated with MP+FLX-induced behavior seen at the end of the repeated MP+FLX treatment. Moreover, the MP+FLX/high reactivity group, but not the low reactivity group, showed facilitated acquisition of cocaine self-administration. These results demonstrate that repeated MP+FLX treatment can facilitate subsequent cocaine taking behavior in a subpopulation of rats. These findings suggest that MP+FLX exposure in some individuals may increase the risk for psychostimulant use later in life.
Collapse
Affiliation(s)
- Lorissa Lamoureux
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Present address: Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Joel A. Beverley
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Michela Marinelli
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Present address: Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Heinz Steiner
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| |
Collapse
|
22
|
Ben-Azu B, Adebayo OG, Moke EG, Omogbiya AI, Oritsemuelebi B, Chidebe EO, Umukoro E, Nwangwa EK, Etijoro E, Umukoro E, Mamudu EJ, Chukwuma C. Geraniol attenuates behavioral and neurochemical impairments by inhibitions of HPA-axis and oxido-inflammatory perturbations in mice exposed to post-traumatic stress disorder. J Psychiatr Res 2023; 168:165-175. [PMID: 37913743 DOI: 10.1016/j.jpsychires.2023.10.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/23/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
Geraniol is an acyclic isoprenoid monoterpenoid analogue that has been shown to elicit neuroprotective functions, primarily through its ability to stimulate antioxidant and anti-inflammatory systems. An increase in inflammatory cytokines and oxidative stress exacerbate activation hypothalamic-pituitary-adrenal axis (HPA), leading to neurochemical dysfunction, which has important roles in the pathogenesis of post-traumatic disorder (PTSD), a mental health disorder characterized of post-trauma-induced intense fear. The aim of this study was to evaluate the anti-PTSD-like effects and underlying mechanisms of geraniol against single-prolonged-stress (SPS)-induced PTSD in mice. Following concomitant exposure to SPS (triple-paradigm traumatic events) and isolation for 7 days, mice (n = 9) were treated with geraniol (50 and 100 mg/kg, p.o.) or fluoxetine (10 mg/kg, p.o.) from days 8-21. Mice were assessed for behavioral changes. Neurochemical changes, inflammatory, oxido-nitrergic markers, adrenal weight, serum glucose and corticosterone concentrations were assayed. Geraniol inhibits SPS-induced anxiety- and depressive-like features as well as behavioral despair in the depression paradigms. SPS-induced locomotor and memory impairments were also abated by geraniol treatment similarly to fluoxetine. SPS-induced adrenal hypertrophy and increased blood glucose and corticosterone concentrations, were attenuated by the geraniol treatment. Elevated levels of TNF-α and IL-6, and malondialdehyde, nitrite, acetylcholinesterase enzyme were reduced by geraniol. Geraniol also increased glutathione, superoxide-dismutase, and catalase levels as well as dopamine, serotonin concentrations and GABAergic glutamic acid decarboxylase enzyme activity in the striatum, prefrontal cortex and hippocampus in the PTSD-mice relative to SPS control. In conclusion, geraniol attenuates behavioral impairments and neurochemical dysregulations by inhibitions of HPA-axis and oxido-inflammatory perturbations in mice exposed to PTSD.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria.
| | - Olusegun G Adebayo
- Neurophysiology Unit, Department of Physiology, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port-Harcourt, River State, Nigeria
| | - Emuesiri G Moke
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Adrian I Omogbiya
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Benjamin Oritsemuelebi
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emmanuel O Chidebe
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emuesiri Umukoro
- Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, College of Medicine Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Eze K Nwangwa
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emmanuel Etijoro
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emmanuel Umukoro
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Elizabeth J Mamudu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Chineye Chukwuma
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| |
Collapse
|
23
|
Wood CP, Avalos B, Alvarez C, DiPatrizio NV. A Sexually Dimorphic Role for Intestinal Cannabinoid Receptor Subtype-1 in the Behavioral Expression of Anxiety. Cannabis Cannabinoid Res 2023; 8:1045-1059. [PMID: 37862126 PMCID: PMC10771877 DOI: 10.1089/can.2023.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Background: Increasing evidence suggests that the endocannabinoid system (ECS) in the brain controls anxiety and may be a therapeutic target for the treatment of anxiety disorders. For example, both pharmacological and genetic disruption of cannabinoid receptor subtype-1 (CB1R) signaling in the central nervous system is associated with increased anxiety-like behaviors in rodents, while activating the system is anxiolytic. Sex is also a critical factor that controls the behavioral expression of anxiety; however, roles for the ECS in the gut in these processes and possible differences between sexes are largely unknown. Objective: In this study, we aimed to determine if CB1Rs in the intestinal epithelium exert control over anxiety-like behaviors in a sex-dependent manner. Methods: We subjected male and female mice with conditional deletion of CB1Rs in the intestinal epithelium (intCB1-/-) and controls (intCB1+/+) to the elevated plus maze (EPM), light/dark box, and open field test. Corticosterone (CORT) levels in plasma were measured at baseline and immediately after EPM exposure. Results: When compared with intCB1+/+ male mice, intCB1-/- male mice exhibited reduced levels of anxiety-like behaviors in the EPM and light/dark box. In contrast to male mice, no differences were found between female intCB1+/+ and intCB1-/- mice. Circulating CORT was higher in female versus male mice for both genotype groups at baseline and after EPM exposure; however, there was no effect of genotype on CORT levels. Conclusions: Collectively, these results indicate that genetic deletion of CB1Rs in the intestinal epithelium is associated with an anxiolytic phenotype in a sex-dependent manner.
Collapse
Affiliation(s)
- Courtney P. Wood
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Bryant Avalos
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Camila Alvarez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Nicholas V. DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
- University of California Riverside Center for Cannabinoid Research, Riverside, California, USA
| |
Collapse
|
24
|
Fricke HP, Krajco CJ, Perry MJ, Desorcy‐Scherer KM, Wake LA, Charles JF, Hernandez LL. Developmental fluoxetine exposure affects adolescent and adult bone depending on the dose and period of exposure in mice. Physiol Rep 2023; 11:e15881. [PMID: 38031314 PMCID: PMC10687345 DOI: 10.14814/phy2.15881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
At the end of gestation, fetal skeleton rapidly accumulates calcium, and bone development continues in offspring postnatally. To accommodate, maternal skeletal physiology is modulated in a serotonin-dependent manner. Selective serotonin reuptake inhibitors (SSRIs) are generally considered safe for treatment of major depressive disorder, postpartum depression, and other psychiatric illnesses during the peripartum period, but because serotonin affects bone remodeling, SSRIs are associated with decreased bone mass across all ages and sexes, and the impact of SSRIs during fetal and postnatal development has not been fully investigated. In the present study, our aim was to examine developmental fluoxetine exposure on offspring skeleton and to assess varying degrees of impact depending on dose and window of exposure in short-term and long-term contexts. We established that a low dose of lactational fluoxetine exposure caused a greater degree of insult to offspring bone than either a low dose during fetal and postpartum development or a high dose during lactation only in mice. We further discovered lasting impacts of developmental fluoxetine exposure, especially during lactation only, on adult bone and body composition. Herein, we provide evidence fluoxetine exposure during early development may have detrimental effects on the skeleton of offspring at weaning and into adulthood.
Collapse
Affiliation(s)
- Hannah P. Fricke
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chandler J. Krajco
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Molly J. Perry
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Katelyn M. Desorcy‐Scherer
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- School of NursingUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Lella A. Wake
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Julia F. Charles
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Laura L. Hernandez
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
25
|
Payet JM, Stevens L, Russo AM, Jaehne EJ, van den Buuse M, Kent S, Lowry CA, Baratta MV, Hale MW. The Role of Dorsal Raphe Nucleus Serotonergic Systems in Emotional Learning and Memory in Male BALB/c Mice. Neuroscience 2023; 534:1-15. [PMID: 37852412 DOI: 10.1016/j.neuroscience.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the first-line pharmacological treatment for a variety of anxiety-, trauma- and stressor-related disorders. Although they are efficacious, therapeutic improvements require several weeks of treatment and are often associated with an initial exacerbation of symptoms. The dorsal raphe nucleus (DR) has been proposed as an important target for the modulation of emotional responses and the therapeutic effects of SSRIs. Using a fear-conditioning paradigm we aimed to understand how SSRIs affect emotional learning and memory, and their effects on serotonergic circuitry. Adult male BALB/c mice were treated with vehicle (n = 16) or the SSRI fluoxetine (18 mg/kg/d) acutely (n = 16), or chronically (21d, n = 16), prior to fear conditioning. Treatment was stopped, and half of the mice (n = 8/treatment group) were exposed to cued fear memory recall 72 h later. Activation of DR serotonergic neurons during fear conditioning (Experiment 1) or fear memory recall (Experiment 2), was measured using dual-label immunohistochemistry for Tph2 and c-Fos. Acute and chronic fluoxetine treatment reduced associative fear learning without affecting memory recall and had opposite effects on anxiety-like behaviour. Acute fluoxetine decreased serotonergic activity in the DR, while chronic treatment led to serotonergic activity that was indistinguishable from that of control levels in DRD and DRV subpopulations. Chronic fluoxetine facilitated fear extinction, which was associated with rostral DRD inhibition. These findings provide further evidence that SSRIs can alter aspects of learning and memory processes and are consistent with a role for discrete populations of DR serotonergic neurons in regulating fear- and anxiety-related behaviours.
Collapse
Affiliation(s)
- Jennyfer M Payet
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Laura Stevens
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Adrian M Russo
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Emily J Jaehne
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Maarten van den Buuse
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Stephen Kent
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Christopher A Lowry
- Department of Integrative Physiology and Centre for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Matthew W Hale
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
26
|
Fricke HP, Krajco CJ, Perry MJ, Brettingen LJ, Wake LA, Charles JF, Hernandez LL. Fluoxetine treatment during the postpartal period may have short-term impacts on murine maternal skeletal physiology. Front Pharmacol 2023; 14:1244580. [PMID: 38074149 PMCID: PMC10701399 DOI: 10.3389/fphar.2023.1244580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023] Open
Abstract
Postpartum depression affects many individuals after parturition, and selective serotonin reuptake inhibitors (SSRIs) are often used as the first-line treatment; however, both SSRIs and lactation are independently associated with bone loss due to the role of serotonin in bone remodeling. Previously, we have established that administration of the SSRI fluoxetine during the peripartal period results in alterations in long-term skeletal characteristics. In the present study, we treated mice with either a low or high dose of fluoxetine during lactation to determine the consequences of the perturbation of serotonin signaling during this time period on the dam skeleton. We found that lactational fluoxetine exposure affected both cortical and trabecular parameters, altered gene expression and circulating markers of bone turnover, and affected mammary gland characteristics, and that these effects were more pronounced in the dams that were exposed to the low dose of fluoxetine in comparison to the high dose. Fluoxetine treatment during the postpartum period in rodents had short term effects on bone that were largely resolved 3 months post-weaning. Despite the overall lack of long-term insult to bone, the alterations in serotonin-driven lactational bone remodeling raises the question of whether fluoxetine is a safe option for the treatment of postpartum depression.
Collapse
Affiliation(s)
- Hannah P. Fricke
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Chandler J. Krajco
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Molly J. Perry
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Lauren J. Brettingen
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Lella A. Wake
- Departments of Orthopedics and Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Julia F. Charles
- Departments of Orthopedics and Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Laura L. Hernandez
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
27
|
Borozdenko DA, Gonchar DI, Bogorodova VI, Tarasenko DV, Kramarova EP, Khovanova SS, Golubev YV, Kiseleva NM, Shmigol TA, Ezdoglian AA, Sobyanin KA, Negrebetsky VV, Baukov YI. The Antidepressant Activity of a Taurine-Containing Derivative of 4-Phenylpyrrolidone-2 in a Model of Chronic Unpredictable Mild Stress. Int J Mol Sci 2023; 24:16564. [PMID: 38068887 PMCID: PMC10705968 DOI: 10.3390/ijms242316564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
This study investigates the therapeutic potential of a new compound, potassium 2-[2-(2-oxo-4-phenylpyrrolidin-1-yl) acetamido]ethanesulfonate (Compound I), in depression. Willner's chronic unpredictable mild stress model of male Wistar rats was used as a depression model. The rats were randomized into four groups, including an intact group, a Compound I group, a Fluoxetine group, and a control group with saline. Behavioral tests, such as the Porsolt forced swim test, hole-board test, elevated plus maze test, and light-dark box, were used to assess the animals' conditions. Our results demonstrated that Compound I effectively reduced the immobilization time of rats in the forced swim test, increased orientation and exploratory behavior, and decreased the latency period of going into the dark compartment compared to the control group. Hippocampal and striatal serotonin concentrations were increased in the Compound I group, and the compound also reduced the level of corticosterone in the blood plasma of rats compared to the intact animals. These results suggest that Compound I has reliable antidepressant activity, comparable to that of the reference antidepressant Fluoxetine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yuri I. Baukov
- Institute of Pharmacy and Medicinal Chemistry, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (D.I.G.); (V.I.B.); (D.V.T.); (E.P.K.); (S.S.K.); (Y.V.G.); (N.M.K.); (T.A.S.); (A.A.E.); (K.A.S.); (V.V.N.)
| |
Collapse
|
28
|
Fricke HP, Krajco CJ, Perry MJ, Reisner MA, Brettingen LJ, Wake LA, Charles JF, Hernandez LL. In utero, lactational, or peripartal fluoxetine administration has differential implications on the murine maternal skeleton. Physiol Rep 2023; 11:e15837. [PMID: 37813559 PMCID: PMC10562136 DOI: 10.14814/phy2.15837] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/04/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023] Open
Abstract
The peripartal period is marked by alterations in calcium metabolism to accommodate for embryonic skeletal mineralization and support bone development of offspring in early life, and serotonin plays a critical role in modulating peripartal bone remodeling. Selective serotonin reuptake inhibitors (SSRIs) are commonly used as first-line treatment for psychiatric illness during pregnancy and the postpartum period and considered safe for maternal use during this time frame. In order to evaluate the effect of peripartal alterations of the serotonergic system on maternal skeletal physiology, we treated dams with the SSRI fluoxetine during gestation only, lactation only, or during the entire peripartal period. Overall, we found a low dose of fluoxetine during gestation only had minimal impacts on maternal bone at weaning, but there were implications on maternal skeleton at weaning when dams were exposed during lactation only or during the entire peripartal period. We found that these effects were differential between female mice dosed lactationally or peripartally, and there were also impacts on maternal mammary gland at weaning in both of these groups. Though SSRIs are largely considered safe maternally during the peripartal period, this study raises the question whether safety of SSRIs, specifically fluoxetine, during the peripartal period should be reevaluated.
Collapse
Affiliation(s)
- Hannah P. Fricke
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chandler J. Krajco
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Molly J. Perry
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Maggie A. Reisner
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - Lella A. Wake
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Julia F. Charles
- Departments of Orthopedics and MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Laura L. Hernandez
- Endocrinology and Reproductive Physiology ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
29
|
Lundgren KA, Løvlie H. Increased dietary 5-hydroxytryptophan reduces fearfulness in red junglefowl hens ( Gallus gallus). Front Physiol 2023; 14:1213986. [PMID: 37766752 PMCID: PMC10520959 DOI: 10.3389/fphys.2023.1213986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023] Open
Abstract
Our production animals typically suffer poor welfare, which can be revealed by measuring the affective state these animals are in. Negative affective state is linked to poorer welfare, and can be measured as fearfulness. While continuing to research how to improve animal welfare, a compliment to reduce negative affective state could therefore be to reduce individuals' fearfulness, similar to how negative affective states are medicated in humans. A proposed mechanism for this is via the monoaminergic systems. This is based on previous studies across species that have linked the serotonergic system and fear-related behaviour. We here aimed to experimentally manipulate the serotonergic system in red junglefowl hens (Gallus gallus), the main ancestor of all domesticated chickens. We measured fearfulness as latency remaining immobile in a tonic immobility test, and did so both before and after our experimental manipulation. We set out to experimentally manipulate the serotonergic system via sub-chronic dietary treatment of 5-hydroxytryptophan (the precursor to serotonin). Our dietary manipulation of 5-hydroxytryptophan significantly reduced measured fearfulness in the manipulated hens, while latency in tonic immobility did not significantly change in our unmanipulated, control hens. This finding is promising since it indicates that increased tryptophan levels can be used to reduce fearfulness. Additionally, our result suggests that this can be done non-invasively via food (instead of injections), thus presenting a potentially feasible manipulation also for larger settings. Nevertheless, the serotonergic system is complex and its role in modulating behaviour in the fowl should be explored further to evaluate our findings, and more directly explored also in a production setting.
Collapse
Affiliation(s)
| | - Hanne Løvlie
- IFM Biology, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
30
|
Jin H, Xu G, Lu Y, Niu C, Zhang X, Kan T, Cao J, Yang X, Cheng Q, Zhang J, Dong J. Fluoxetine partially alleviates inflammation in the kidney of socially stressed male C57 BL/6 mice. FEBS Open Bio 2023; 13:1723-1736. [PMID: 37400956 PMCID: PMC10476569 DOI: 10.1002/2211-5463.13670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/19/2023] [Accepted: 07/01/2023] [Indexed: 07/05/2023] Open
Abstract
Stress-related illnesses are linked to the onset and progression of renal diseases and depressive disorders. To investigate stress-induced changes in the renal transcriptome associated with the development of depressive behaviors, we generated here a chronic social defeat stress (CSDS) model of C57 BL/6 male mice and then performed RNA sequencing of the kidneys to obtain an inflammation-related transcriptome. Administration of the antidepressant drug fluoxetine (10 mg·kg-1 ·day-1 ) during CSDS induction could partially alleviate renal inflammation and reverse CSDS-induced depression-like behaviors. Moreover, fluoxetine also modulated gene expression of stress-related hormone receptors, including prolactin and melanin-concentrating hormone. These results suggest that CSDS can induce gene expression changes associated with inflammation in the kidney of C57 BL/6 male mice, and this inflammation can be treated effectively by fluoxetine.
Collapse
Affiliation(s)
- Hailong Jin
- The Third CenterPLA General HospitalBeijingChina
| | - Guanglei Xu
- Beijing Institute of Basic Medical SciencesChina
| | - Yuchen Lu
- Beijing Institute of Basic Medical SciencesChina
| | - Chunxiao Niu
- Beijing Institute of Basic Medical SciencesChina
| | | | - Tongtong Kan
- Beijing Institute of Basic Medical SciencesChina
| | - Junxia Cao
- Beijing Institute of Basic Medical SciencesChina
| | - Xiqin Yang
- Beijing Institute of Basic Medical SciencesChina
| | | | - Jiyan Zhang
- Beijing Institute of Basic Medical SciencesChina
| | - Jie Dong
- Beijing Institute of Basic Medical SciencesChina
| |
Collapse
|
31
|
Alyoshina NM, Tkachenko MD, Nikishina YO, Nikishin DA. Serotonin Transporter Activity in Mouse Oocytes Is a Positive Indicator of Follicular Growth and Oocyte Maturity. Int J Mol Sci 2023; 24:11247. [PMID: 37511007 PMCID: PMC10379015 DOI: 10.3390/ijms241411247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is known to be a regulator of oocyte maturation in a large number of animal species. In maturing mammalian oocytes, the accumulation of exogenous, maternal serotonin occurs due to the activity of the membrane transporter SERT. In this work, we investigated how SERT activity in oocytes correlates with indicators of follicular selection and oocyte maturity. An immunohistochemical study showed that the difference in the 5-HT intake activity in oocytes does not correlate with the marker of apoptosis in follicular cells, but positively correlates with markers of follicular growth, such as granulosa proliferation and follicle size. Functional analysis of oocytes at different stages of maturation showed that the expression and activity of SERT increases with oocyte maturation. An in vivo experiment on administration of the selective serotonin reuptake inhibitor fluoxetine (20 mg/kg) for 7 days showed a significant decrease in the content of serotonin in both growing GV-oocytes and ovulated mature MII-oocytes. The data obtained clearly indicate that the mechanism of specific membrane transport of serotonin normally ensures the accumulation of serotonin in maturing oocytes, and can be considered as a promising positive marker of their mature status.
Collapse
Affiliation(s)
- Nina M Alyoshina
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street, 26, 119334 Moscow, Russia
| | - Maria D Tkachenko
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street, 26, 119334 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119991 Moscow, Russia
| | - Yulia O Nikishina
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street, 26, 119334 Moscow, Russia
| | - Denis A Nikishin
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street, 26, 119334 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119991 Moscow, Russia
| |
Collapse
|
32
|
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: 2.0] [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
|
33
|
Musaelyan K, Horowitz MA, McHugh S, Szele FG. Fluoxetine Can Cause Epileptogenesis and Aberrant Neurogenesis in Male Wild-Type Mice. Dev Neurosci 2023; 46:158-166. [PMID: 37302394 DOI: 10.1159/000531478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Antidepressants in general, and fluoxetine in particular, increase adult hippocampal neurogenesis (AHN) in mice. Here we asked how the antidepressant fluoxetine affects behavior and AHN in a corticosterone model of depression. In three groups of adult male C57BL/6j mice, we administered either vehicle (VEH), corticosterone (CORT) treatment to induce a depression-like state, or corticosterone plus a standard dose of fluoxetine (CORT+FLX). Following treatment, mice performed the open field test, the novelty suppressed feeding (NSF) test, and the splash test. Neurogenesis was assessed by means of immunohistochemistry using BrdU and neuronal maturation markers. Unexpectedly, 42% of the CORT+FLX-treated mice exhibited severe weight loss, seizures, and sudden death. As expected, the CORT-treated group had altered behaviors compared to the VEH group, but the CORT+FLX mice that survived did not show any behavioral improvement compared to the CORT group. Antidepressants generally increase neurogenesis and here we also found that compared to CORT mice, CORT+FLX mice that survived had a significantly greater density of BrdU+, BrdU+DCX+, and BrdU+NeuN+ cells, suggesting increased neurogenesis. Moreover, the density of BrdU+NeuN+ cells was increased in an aberrant location, the hilus, of CORT+FLX mice, similar to previous studies describing aberrant neurogenesis following seizures. In conclusion, fluoxetine could induce considerable adverse effects in wild-type mice, including seizure-like activity. Fluoxetine-induced neurogenesis increases could be related to this activity; therefore, proneurogenic effects of fluoxetine and other antidepressants, especially in the absence of any behavioral therapeutic effects, should be interpreted with caution.
Collapse
Affiliation(s)
- Ksenia Musaelyan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Mark A Horowitz
- Research and Development Department, North East London NHS Foundation Trust, Ilford, UK
- Department of Psychiatry, University College London, London, UK
| | - Stephen McHugh
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Filipović D, Novak B, Xiao J, Yan Y, Bernardi RE, Turck CW. Chronic fluoxetine treatment in socially-isolated rats modulates the prefrontal cortex synaptoproteome. J Proteomics 2023; 282:104925. [PMID: 37164273 DOI: 10.1016/j.jprot.2023.104925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Exposure to chronic social isolation (CSIS) and synapse dysfunction have been implicated in the etiology of major depressive disorder (MDD). Fluoxetine (Flx) has been widely used to treat MDD, but its mechanisms of action remain elusive. We employed comparative synaptoproteomics to investigate the changes in the levels of proteins and molecular signaling pathways in prefrontal cortical samples of adult male Wistar rats exposed to CSIS, a rat model of depression, and CSIS rats treated with chronic Flx and controls, using liquid chromatography coupled to tandem mass spectrometry. Flx-treated control rats showed a decreased level of proteins involved in vesicle-mediated transport, and a predominantly increased level of exocytosis-associated proteins. CSIS significantly reduced the level of proteins involved in the ATP metabolic process, clathrin-dependent endocytosis, and proteolysis. Flx treatment in CSIS rats stimulated synaptic vesicle trafficking by increasing the regulation of exo/endocytosis-associated proteins, proteins involved in synaptic plasticity including neurogenesis, Cox5a, mitochondria-associated proteins involved in oxidative phosphorylation, and ion transport proteins (Slc8a2, Atp1b2). Flx treatment resulted in an increased synaptic vesicle dynamic, plasticity and mitochondrial functionality, and a suppression of CSIS-induced impairment of these processes. BIOLOGICAL SIGNIFICANCE: Identifying biomarkers of MDD and treatment response is the goal of many studies. Contemporary studies have shown that many molecular alterations associated with the pathophysiology of MDD reside within the synapse. As part of this research, a growing importance is the use of proteomics, as monitoring the changes in protein levels enables the identification of (possible) biochemical pathways and processes of importance for the development of depressive-like behavior and the efficacy of antidepressant treatments. We profiled proteomic changes representative of the development of CSIS-induced depressive-like behavior and the antidepressant effects of Flx. Our study has identified synaptosomal proteins and altered molecular pathways that may be potential markers of prefrontal cortical synaptic dysfunction associated with depressive-like behavior, and further clarified the mechanisms of depressive-like behavior and mode of action of Flx. Our findings indicate potential PFC synaptic targets for antidepressant treatment.
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, Belgrade, Serbia.
| | - Božidar Novak
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jinqiu Xiao
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Yu Yan
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph W Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
35
|
Fricke HP, Hernandez LL. The Serotonergic System and Bone Metabolism During Pregnancy and Lactation and the Implications of SSRI Use on the Maternal-Offspring Dyad. J Mammary Gland Biol Neoplasia 2023; 28:7. [PMID: 37086330 PMCID: PMC10122632 DOI: 10.1007/s10911-023-09535-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Lactation is a physiological adaptation of the class Mammalia and is a product of over 200 million years of evolution. During lactation, the mammary gland orchestrates bone metabolism via serotonin signaling in order to provide sufficient calcium for the offspring in milk. The role of serotonin in bone remodeling was first discovered over two decades ago, and the interplay between serotonin, lactation, and bone metabolism has been explored in the years following. It is estimated that postpartum depression affects 10-15% of the population, and selective serotonin reuptake inhibitors (SSRI) are often used as the first-line treatment. Studies conducted in humans, nonhuman primates, sheep, and rodents have provided evidence that there are consequences on both parent and offspring when serotonin signaling is disrupted during the peripartal period; however, the long-term consequences of disruption of serotonin signaling via SSRIs during the peripartal period on the maternal and offspring skeleton are not fully known. This review will focus on the relationship between the mammary gland, serotonin, and bone remodeling during the peripartal period and the skeletal consequences of the dysregulation of the serotonergic system in both human and animal studies.
Collapse
Affiliation(s)
- Hannah P Fricke
- Animal and Dairy Sciences Department, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura L Hernandez
- Animal and Dairy Sciences Department, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
36
|
Inaba H, Li H, Kawatake-Kuno A, Dewa KI, Nagai J, Oishi N, Murai T, Uchida S. GPCR-mediated calcium and cAMP signaling determines psychosocial stress susceptibility and resiliency. SCIENCE ADVANCES 2023; 9:eade5397. [PMID: 37018397 PMCID: PMC10075968 DOI: 10.1126/sciadv.ade5397] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/02/2023] [Indexed: 06/19/2023]
Abstract
Chronic stress increases the risk of developing psychiatric disorders, including mood and anxiety disorders. Although behavioral responses to repeated stress vary across individuals, the underlying mechanisms remain unclear. Here, we perform a genome-wide transcriptome analysis of an animal model of depression and patients with clinical depression and report that dysfunction of the Fos-mediated transcription network in the anterior cingulate cortex (ACC) confers a stress-induced social interaction deficit. Critically, CRISPR-Cas9-mediated ACC Fos knockdown causes social interaction deficits under stressful situation. Moreover, two classical second messenger pathways, calcium and cyclic AMP, in the ACC during stress differentially modulate Fos expression and regulate stress-induced changes in social behaviors. Our findings highlight a behaviorally relevant mechanism for the regulation of calcium- and cAMP-mediated Fos expression that has potential as a therapeutic target for psychiatric disorders related to stressful environments.
Collapse
Affiliation(s)
- Hiromichi Inaba
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Haiyan Li
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ayako Kawatake-Kuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ken-ichi Dewa
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Jun Nagai
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoya Oishi
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshiya Murai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shusaku Uchida
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
37
|
Xhakaza NK, Nkomozepi P, Mbajiorgu EF. Boophone disticha attenuates five day repeated forced swim-induced stress and adult hippocampal neurogenesis impairment in male Balb/c mice. Anat Cell Biol 2023; 56:69-85. [PMID: 36267006 PMCID: PMC9989792 DOI: 10.5115/acb.22.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/27/2022] Open
Abstract
Depression is one of the most common neuropsychiatric disorders and is associated with dysfunction of the neuroendocrine system and alterations in specific brain proteins. Boophone disticha (BD) is an indigenous psychoactive bulb that belongs to the Amaryllidacae family, which is widely used in Southern Africa to treat depression, with scientific evidence of potent antidepressant-like effects. The present study examined the antidepressant effects of BD and its mechanisms of action by measuring some behavioural parameters in the elevated plus maze, brain content of corticosterone, brain derived neurotropic factor (BDNF), and neuroblast differentiation in the hippocampus of Balb/c mice exposed to the five day repeated forced swim stress (5d-RFSS). Male Balb/c mice were subjected to the 5d-RFSS protocol to induce depressive-like behaviour (decreased swimming, increased floating, decreased open arm entry, decreased time spent in the open arms and decreased head dips in the elevated plus maze test) and treated with distilled water, fluoxetine and BD. BD treatment (10 mg/kg/p.o for 3 weeks) significantly attenuated the 5d-RFSS-induced behavioural abnormalities and the elevated serum corticosterone levels observed in stressed mice. Additionally, 5d-RFSS exposure significantly decreased the number of neuroblasts in the hippocampus and BDNF levels in the brain of Balb/c mice, while fluoxetine and BD treatment attenuated these changes. The antidepressant effects of BD were comparable to those of fluoxetine, but unlike fluoxetine, BD did not show any anxiogenic effects, suggesting better pharmacological functions. In conclusion, our study shows that BD exerted antidepressant-like effects in 5d-RFSS mice, mediated in part by normalizing brain corticosterone and BDNF levels.
Collapse
Affiliation(s)
- Nkosiphendule Khuthazelani Xhakaza
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Anatomy, School of Medicine, Sefako Magkatho Health Sciences University, Pretoria, South Africa
| | - Pilani Nkomozepi
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Ejekemi Felix Mbajiorgu
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
38
|
Menon N, Wang C, Carr JA. Sub-chronic administration of fluoxetine does not alter prey-capture or predator avoidance behaviors in adult South African clawed frogs (Xenopus laevis). Behav Brain Res 2023; 442:114317. [PMID: 36709047 DOI: 10.1016/j.bbr.2023.114317] [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: 07/28/2022] [Revised: 12/23/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Animals will halt foraging efforts and engage defensive behaviors in response to predator cues. Some researchers have proposed that the switch from appetitive to avoidance behavior resembles anxiety, but most work on this has been performed in a limited number of animal models, primarily zebrafish and rodents. We used adult South African clawed frogs (Xenopus laevis) to determine if the canonical anxiolytic fluoxetine alters predator-induced changes in appetitive and avoidance behavior in a laboratory-based trade-off task that mimics foraging/predator avoidance tradeoffs in the wild. We hypothesized that sub-chronic fluoxetine treatment (20 d) would not affect baseline behavior but would reverse predator-induced changes in food intake, appetitive and avoidance behavior, and the abundance of anxiety related gene transcripts in the optic tectum, a brain area central to ecological decision making in frogs. We found that fluoxetine significantly reduced baseline locomotion compared to vehicle-treated animals. Fluoxetine had no effect on appetitive and avoidance behaviors that were sensitive to predator cues in this assay and did not alter any of the anxiety-related transcripts in the tectum. We conclude that while peripheral sub-chronic administration of fluoxetine significantly reduces locomotion, it does not modify predator-induced changes in approach and avoidance behaviors in this assay. Our findings are not consistent with visual predator cues causing state anxiety in adult frogs.
Collapse
Affiliation(s)
- Nikhil Menon
- Texas Tech University, Department of Biological Sciences, 2901 Main St, Lubbock, TX 79409, USA
| | - Caoyuanhui Wang
- Texas Tech University, Department of Biological Sciences, 2901 Main St, Lubbock, TX 79409, USA
| | - James A Carr
- Texas Tech University, Department of Biological Sciences, 2901 Main St, Lubbock, TX 79409, USA.
| |
Collapse
|
39
|
Erdem AS, Şimşek Kaya G, Kaya M, Altunay B, Alkan TY, Toru HS. Comparison of the effects of fluoxetine and venlafaxine on bone healing in a rat calvarial defect model. Injury 2023:S0020-1383(23)00290-5. [PMID: 37003871 DOI: 10.1016/j.injury.2023.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/02/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023]
Abstract
OBJECTIVE The purpose of this study was to compare the effects of the selective serotonin reuptake inhibitor (SSRI) fluoxetine and the serotonin-norepinephrine reuptake inhibitor (SNRI) venlafaxine on bone defect healing. MATERIALS AND METHODS Wistar rats were randomly divided into three groups of eight animals each. The first received 0.1 ml/kg sterile saline solution, the second 5 mg/kg fluoxetine, and the third 5 mg/kg venlafaxine, daily by gastric gavage over 7 weeks. At week 3 of drug therapy, 5-mm diameter calvarial defects were created in the parietal bone of all of the animals. All rats were euthanized four weeks after surgery, micro-CT analysis and histomorphometric analysis were carried out to evaluate the following parameters: Bone volume fraction (BV/TV), bone surface (BS), bone surface density (BS/BV; bone surface/bone volume, 1/mm), trabecular number (Tb. N), trabecular thickness (Tb. Th), areas of new bone structure (positive areas), areas of mature bone structure (negative areas). RESULTS Micro-CT analysis showed the presence of similar levels of bone formation within the defect site in all three groups (p>0.05). Histomorphometric analysis revealed the presence of bone-forming cells at the defect periphery, with less activity indicating bone formation at the center. No statistically significant difference was observed between the groups (p>0.05). CONCLUSION Based on the findings of this study, it can be said that the use of both antidepressants hasn't any effect on bone defect healing.
Collapse
Affiliation(s)
- Arif Sermed Erdem
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Akdeniz University, Antalya, Turkey
| | - Göksel Şimşek Kaya
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Akdeniz University, Antalya, Turkey.
| | - Mahir Kaya
- Department of Medical Imaging Techniques, Vocational School of Health Services, Akdeniz University, Antalya, Turkey
| | - Büşra Altunay
- Department of Pathology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - Taha Yiğit Alkan
- Department of Computer Engineering, Institute of Natural and Applied Sciences, Akdeniz University, Antalya, Turkey
| | - Havva Serap Toru
- Department of Pathology, School of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
40
|
Nguyen TML, Defaix C, Mendez-David I, Tritschler L, Etting I, Alvarez JC, Choucha W, Colle R, Corruble E, David DJ, Gardier AM. Intranasal (R, S)-ketamine delivery induces sustained antidepressant effects associated with changes in cortical balance of excitatory/inhibitory synaptic activity. Neuropharmacology 2023; 225:109357. [PMID: 36462636 DOI: 10.1016/j.neuropharm.2022.109357] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
In 2019, an intranasal (IN) spray of esketamine SPRAVATO® was approved as a fast-acting antidepressant by drug Agencies US FDA and European EMA. At sub-anesthetic doses, (±)-ketamine, a non-competitive glutamate N-methyl-d-aspartate (NMDA) receptor antagonist, increases the overall excitability of the medial prefrontal cortex (mPFC), an effect being essential for its rapid antidepressant activity. We wondered if this effect of ketamine could come from changes in the balance between neuronal excitation and inhibition (E/I balance) in the mPFC. Here, we performed a preclinical approach to study neurochemical and behavioral responses to a single IN ketamine dose in BALB/cJ mice, a strain more sensitive to stress. By using in vivo microdialysis, we measured cortical E/I balance as the ratio between glutamate to GABA extracellular levels 24 h post-ketamine. We found, for the first time, that E/I balance was shifted in favor of excitation rather than inhibition in the mPFC but more robustly with IN KET than with a single intraperitoneal (IP) dose. Increases in plasma and brain ketamine, norketamine and HNKs levels suggest different metabolic profiles of IP and IN ketamine 30 min post-dose. A significantly larger proportion of ketamine and HNKs in the brain are derived from the IN route 30 min post-dose. It may be linked to the greater magnitude in E/I ratio following IN delivery relative to IP at t24 h. This study suggests that both IP and IN are effective brain delivery methods inducing similar sustained antidepressant efficacy of KET, but the way they induced neurotransmitter changes is slightly different.
Collapse
Affiliation(s)
- Thi Mai Loan Nguyen
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Céline Defaix
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Indira Mendez-David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Laurent Tritschler
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Isabelle Etting
- Lab. Pharmacologie-Toxicologie, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | - Jean-Claude Alvarez
- Lab. Pharmacologie-Toxicologie, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | - Walid Choucha
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Romain Colle
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Denis J David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Alain M Gardier
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France.
| |
Collapse
|
41
|
Olson KM, Hillhouse TM, Burgess GE, West JL, Hallahan JE, Dripps IJ, Ladetto AG, Rice KC, Jutkiewicz EM, Traynor JR. Delta Opioid Receptor-Mediated Antidepressant-Like Effects of Diprenorphine in Mice. J Pharmacol Exp Ther 2023; 384:343-352. [PMID: 36456196 PMCID: PMC9976798 DOI: 10.1124/jpet.122.001182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Major depressive disorder is a highly common disorder, with a lifetime prevalence in the United States of approximately 21%. Traditional antidepressant treatments are limited by a delayed onset of action and minimal efficacy in some patients. Ketamine is effective and fast-acting, but there are concerns over its abuse liability. Thus, there is a need for safe, fast-acting antidepressant drugs. The opioid buprenorphine shows promise but also has abuse liability due to its mu-agonist component. Preclinical evidence indicates that the delta-opioid system contributes to mood disorders, and delta-opioid agonists are effective in preclinical models of depression- and anxiety-like states. In this study, we test the hypothesis that the mu-opioid antagonist diprenorphine by virtue of its partial delta opioid agonist activity may offer a beneficial profile for an antidepressant medication without abuse liability. Diprenorphine was confirmed to bind with high affinity to all three opioid receptors, and functional experiments for G protein activation verified diprenorphine to be a partial agonist at delta- and kappa-opioid receptors and a mu-antagonist. Studies in C57BL/6 mice demonstrated that an acute dose of diprenorphine produced antidepressant-like effects in the tail suspension test and the novelty-induced hypophagia test that were inhibited in the presence of the delta-selective antagonist, naltrindole. Diprenorphine did not produce convulsions, a side effect of many delta agonists but rather inhibited convulsions caused by the full delta agonist SNC80; however, diprenorphine did potentiate pentylenetetrazole-induced convulsions. Diprenorphine, and compounds with a similar pharmacological profile, may provide efficient and safe rapidly acting antidepressants. SIGNIFICANCE STATEMENT: The management of major depressive disorder, particularly treatment-resistant depression, is a significant unmet medical need. Here we show that the opioid diprenorphine, a compound with mu-opioid receptor antagonist activity and delta- and kappa-opioid receptor partial agonist activities, has rapid onset antidepressant-like activity in animal models. Diprenorphine and compounds with a similar pharmacological profile to diprenorphine should be explored as novel antidepressant drugs.
Collapse
MESH Headings
- Animals
- Mice
- Analgesics, Opioid/pharmacology
- Antidepressive Agents/pharmacology
- Depressive Disorder, Major
- Diprenorphine/pharmacology
- Mice, Inbred C57BL
- Receptors, Opioid
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/metabolism
- Seizures/chemically induced
Collapse
Affiliation(s)
- Keith M Olson
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Todd M Hillhouse
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Gwendolyn E Burgess
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Joshua L West
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - James E Hallahan
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Isaac J Dripps
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Allison G Ladetto
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Kenner C Rice
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - Emily M Jutkiewicz
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| | - John R Traynor
- Department of Pharmacology and Edward F Domino Research Center (K.M.O., T.M.H., G.E.B., J.L.W., J.E.H., I.J.D., A.G.L., E.M.J., J.R.T.) and Department of Medicinal Chemistry (J.R.T.), University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H.); and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (K.C.R.)
| |
Collapse
|
42
|
Adongo DW, Mante PK, Kukuia KKE, Benneh CK, Biney RP, Boakye-Gyasi E, Amekyeh H, Harley BK, Tandoh A, Okyere PD, Woode E. Fast-onset effects of Pseudospondias microcarpa (A. Rich) Engl. (Anacardiaceae) hydroethanolic leaf extract on behavioral alterations induced by chronic mild stress in mice. PLoS One 2023; 18:e0278231. [PMID: 36730151 PMCID: PMC9894402 DOI: 10.1371/journal.pone.0278231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 11/10/2022] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Pseudospondias microcarpa (Anacardiaceae) is a plant widely used traditionally for treating various central nervous system disorders. A previous study in our laboratory confirmed that the hydroethanolic leaf extract (PME) of the plant produces an antidepressant-like effect in rodent models of behavioral despair. However, its effect on depressive-like behavior induced by chronic mild stress (CMS) and its time course of action are still unknown. In this context, the long-term effects of PME on cognitive function and depressive- and anxiety-like behavior caused by CMS were assessed. METHODS Male ICR mice were exposed to CMS for nine weeks and anhedonia was evaluated by monitoring sucrose intake (SIT) weekly. PME (30, 100, or 300 mg kg-1) or fluoxetine (FLX) (3, 10, or 30 mg kg-1) was administered to the mice during the last six weeks of CMS. Behavioral tests-coat state, splash test, forced swimming test (FST), tail suspension test (TST), elevated plus maze (EPM), open field test (OFT), novelty suppressed feeding (NSF), EPM transfer latency, and Morris water maze (MWM)-were performed after the nine-week CMS period. RESULTS When the mice were exposed to CMS, their SIT and grooming behavior reduced (splash test), their coat status was poor, they became more immobile (FST and TST), more anxious (OFT, EPM, and NSF), and their cognitive function was compromised (EPM transfer latency and MWM tests). Chronic PME treatment, however, was able to counteract these effects. Additionally, following two (2) weeks of treatment, PME significantly boosted SIT in stressed mice (30 mg kg-1, P<0.05; 100 mg kg-1, P<0.05; and 300 mg kg-1, P<0.001), as compared to four (4) weeks of treatment with FLX. CONCLUSION The present findings demonstrate that PME produces a rapid and sustained antidepressant-like action and reverses behavioral changes induced by chronic exposure to mild stressors.
Collapse
Affiliation(s)
- Donatus Wewura Adongo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
- * E-mail:
| | - Priscilla Kolibea Mante
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Kennedy Kwami Edem Kukuia
- Department of Medical Pharmacology, University of Ghana Medical School, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana
| | - Charles Kwaku Benneh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Robert Peter Biney
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Eric Boakye-Gyasi
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Hilda Amekyeh
- Department of Pharmaceutics, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Benjamin Kingsley Harley
- Department of Pharmacognosy and Herbal Medicine, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Augustine Tandoh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Prince Dagadu Okyere
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Eric Woode
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| |
Collapse
|
43
|
Neuronal nitric oxide synthase inhibition accelerated the removal of fluoxetine's anxiogenic activity in an animal model of PTSD. Behav Brain Res 2023; 437:114128. [PMID: 36174841 DOI: 10.1016/j.bbr.2022.114128] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/06/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022]
Abstract
While SSRIs are the current first-line pharmacotherapies against post-traumatic stress disorder (PTSD), they suffer from delayed onset of efficacy and low remission rates. One solution is to combine SSRIs with other treatments. Neuronal nitric oxide synthase (nNOS) has been shown to play a role in serotonergic signaling, and there is evidence of synergism between nNOS modulation and SSRIs in models of other psychiatric conditions. Therefore, in this study, we combined subchronic fluoxetine (Flx) with 7-nitroindazole (NI), a selective nNOS inhibitor, and evaluated their efficacy against anxiety-related behavior in an animal model of PTSD. We used the underwater trauma model to induce PTSD in rats. Animals underwent the open field (OFT) and elevated plus maze tests on days 14 (baseline) and 21 (post-treatment) after PTSD induction to assess anxiety-related behaviors. Between the two tests, the rats received daily intraperitoneal injections of 10 mg/kg Flx or saline, and were injected intraperitoneally before the second test with either 15 mg/kg NI or saline. The change in behaviors between the two tests was compared between treatment groups. Individual treatment with both Flx and NI had anxiogenic effects in the OFT. These effects were associated with modest increases in cFOS expression in the hippocampus. Combination therapy with Flx + NI did not show any anxiogenic effects, while causing even higher expression levels of cFOS. In conclusion, addition of NI treatment to subchronic Flx therapy accelerated the abrogation of Flx's anxiogenic properties. Furthermore, hippocampal activity, as evidenced by cFOS expression, was biphasically related to anxiety-related behavior.
Collapse
|
44
|
Funayama Y, Li H, Ishimori E, Kawatake-Kuno A, Inaba H, Yamagata H, Seki T, Nakagawa S, Watanabe Y, Murai T, Oishi N, Uchida S. Antidepressant Response and Stress Resilience Are Promoted by CART Peptides in GABAergic Neurons of the Anterior Cingulate Cortex. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:87-98. [PMID: 36712563 PMCID: PMC9874166 DOI: 10.1016/j.bpsgos.2021.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/09/2021] [Accepted: 12/27/2021] [Indexed: 02/01/2023] Open
Abstract
Background A key challenge in the understanding and treatment of depression is identifying cell types and molecular mechanisms that mediate behavioral responses to antidepressant drugs. Because treatment responses in clinical depression are heterogeneous, it is crucial to examine treatment responders and nonresponders in preclinical studies. Methods We used the large variance in behavioral responses to long-term treatment with multiple classes of antidepressant drugs in different inbred mouse strains and classified the mice into responders and nonresponders based on their response in the forced swim test. Medial prefrontal cortex tissues were subjected to RNA sequencing to identify molecules that are consistently associated across antidepressant responders. We developed and used virus-mediated gene transfer to induce the gene of interest in specific cell types and performed forced swim, sucrose preference, social interaction, and open field tests to investigate antidepressant-like and anxiety-like behaviors. Results Cartpt expression was consistently upregulated in responders to four types of antidepressants but not in nonresponders in different mice strains. Responder mice given a single dose of ketamine, a fast-acting non-monoamine-based antidepressant, exhibited high CART peptide expression. CART peptide overexpression in the GABAergic (gamma-aminobutyric acidergic) neurons of the anterior cingulate cortex led to antidepressant-like behavior and drove chronic stress resiliency independently of mouse genetic background. Conclusions These data demonstrate that activation of CART peptide signaling in GABAergic neurons of the anterior cingulate cortex is a common molecular mechanism across antidepressant responders and that this pathway also drives stress resilience.
Collapse
Affiliation(s)
- Yuki Funayama
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haiyan Li
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Erina Ishimori
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ayako Kawatake-Kuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiromichi Inaba
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirotaka Yamagata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tomoe Seki
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shin Nakagawa
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yoshifumi Watanabe
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Toshiya Murai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoya Oishi
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shusaku Uchida
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
45
|
Huang J, Xie XM, Lyu N, Fu BB, Zhao Q, Zhang L, Wang G. Agomelatine in the treatment of anhedonia, somatic symptoms, and sexual dysfunction in major depressive disorder. Front Psychiatry 2023; 14:1115008. [PMID: 37151978 PMCID: PMC10157485 DOI: 10.3389/fpsyt.2023.1115008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/27/2023] [Indexed: 05/09/2023] Open
Abstract
Objective This study evaluated the treatment outcomes of agomelatine on anhedonic state, anxiety/somatic symptoms, and sexual function in Chinese patients with major depressive disorder (MDD). Method In total, 93 adult patients with MDD were enrolled, and 68 of them were included in a prospective, open-label, multicenter clinical study. All patients received agomelatine monotherapy during a 9-week treatment phase. The effectiveness of the treatment was reflected by the improvement of anhedonia and somatic symptoms based on the 17-item Hamilton Depression Rating Scale (HAMD-17). In addition, the Arizona Sexual Dysfunction Scale (ASEX), Sheehan Disability Scale (SDS), and Short Form of Quality-of-Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q-SF) were administered to all participants at baseline and at the 3-, 6-, and 9-week follow-ups. Results After 9 weeks of treatment with agomelatine, the response and remission rates were 73.5% and 39.7%, respectively. Somatic symptoms significantly improved at week 9 (p < 0.001), and significant effects were also observed on the HAMD anhedonia items (p < 0.001). The patients exhibited lower levels of disease severity (the SDS score dropped from 15.52 ± 4.7 to 7.09 ± 5.62 at week 9; the ASEX score dropped from 21.89 ± 4.06 to 16.19 ± 4.79, p < 0.001) and higher levels of QOL (the Q-LES-Q-SF score dropped from 41.02 ± 5.99 to 50.49 ± 8.57, p < 0.001) during the follow-up. Furthermore, treatment with agomelatine improved depressive symptoms without causing serious adverse events. Conclusion These analyses indicate that agomelatine is a treatment option for improving anhedonic status, anxiety/somatic symptoms, and sexual dysfunction in MDD patients.
Collapse
Affiliation(s)
- Juan Huang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xiao-Meng Xie
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Nan Lyu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Bing-Bing Fu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qian Zhao
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ling Zhang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- *Correspondence: Ling Zhang
| | - Gang Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Gang Wang
| |
Collapse
|
46
|
Salvan P, Fonseca M, Winkler AM, Beauchamp A, Lerch JP, Johansen-Berg H. Serotonin regulation of behavior via large-scale neuromodulation of serotonin receptor networks. Nat Neurosci 2023; 26:53-63. [PMID: 36522497 PMCID: PMC9829536 DOI: 10.1038/s41593-022-01213-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/24/2022] [Indexed: 12/23/2022]
Abstract
Although we understand how serotonin receptors function at the single-cell level, what role different serotonin receptors play in regulating brain-wide activity and, in turn, human behavior, remains unknown. Here, we developed transcriptomic-neuroimaging mapping to characterize brain-wide functional signatures associated with specific serotonin receptors: serotonin receptor networks (SRNs). Probing SRNs with optogenetics-functional magnetic resonance imaging (MRI) and pharmacology in mice, we show that activation of dorsal raphe serotonin neurons differentially modulates the amplitude and functional connectivity of different SRNs, showing that receptors' spatial distributions can confer specificity not only at the local, but also at the brain-wide, network level. In humans, using resting-state functional MRI, SRNs replicate established divisions of serotonin effects on impulsivity and negative biases. These results provide compelling evidence that heterogeneous brain-wide distributions of different serotonin receptor types may underpin behaviorally distinct modes of serotonin regulation. This suggests that serotonin neurons may regulate multiple aspects of human behavior via modulation of large-scale receptor networks.
Collapse
Affiliation(s)
- Piergiorgio Salvan
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| | - Madalena Fonseca
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Anderson M Winkler
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Antoine Beauchamp
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jason P Lerch
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Heidi Johansen-Berg
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
47
|
Kyi-Tha-Thu C, Fujitani Y, Hirano S, Win-Shwe TT. Early-Life Exposure to Traffic-Related Air Pollutants Induced Anxiety-like Behaviors in Rats via Neurotransmitters and Neurotrophic Factors. Int J Mol Sci 2022; 24:ijms24010586. [PMID: 36614034 PMCID: PMC9820394 DOI: 10.3390/ijms24010586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Recent epidemiological studies have reported significantly increasing hospital admission rates for mental disorders such as anxiety and depression, not only in adults but also in children and adolescents, indicating more research is needed for evaluation of the etiology and possible reduction and prevention of these disorders. The aim of the present study was to examine the associations between perinatal exposure to traffic-related air pollutants and anxiety-like behaviors and alterations in neurological and immunological markers in adulthood using a rat model. Sprague Dawley pregnant rats were exposed to clean air (control), diesel exhaust (DE) 101 ± 9 μg/m3 or diesel exhaust origin secondary organic aerosol (DE-SOA) 118 ± 23 μg/m3 from gestational day 14 to postnatal day 21. Anxiety-related behavioral tests including open field tests, elevated plus maze, light/dark transition tests and novelty-induced hypophagia were performed on 10-week-old rats. The hippocampal expression of neurotransmitters, neurotrophic factors, and inflammatory molecular markers was examined by real-time RT-PCR. Anxiety-like behaviors were observed in both male and female rat offspring exposed to DE or DE-SOA. Moreover, serotonin receptor (5HT1A), dopamine receptor (Drd2), brain-derived neurotrophic factor and vascular endothelial growth factor A mRNAs were significantly decreased, whereas interleukin-1β, cyclooxygenase-2, heme oxygenase-1 mRNAs and microglial activation were significantly increased in both male and female rats. These findings indicate that brain developmental period exposure to traffic-related air pollutants may induce anxiety-like behaviors via modulation of neurotransmitters, neurotrophic factors, and immunological molecular markers, triggering neuroinflammation and microglia activation in rats.
Collapse
Affiliation(s)
- Chaw Kyi-Tha-Thu
- Department of Immunology, School of Medicine, International University of Health and Welfare, 4-3, Kozunomori, Narita 286-8686, Chiba, Japan
| | - Yuji Fujitani
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Ibaraki, Japan
| | - Seishiro Hirano
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Ibaraki, Japan
| | - Tin-Tin Win-Shwe
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Ibaraki, Japan
- Correspondence: ; Tel.: +81-29-850-2542
| |
Collapse
|
48
|
Sarkar I, Snippe-Strauss M, Tenenhaus Zamir A, Benhos A, Richter-Levin G. Individual behavioral profiling as a translational approach to assess treatment efficacy in an animal model of post-traumatic stress disorder. Front Neurosci 2022; 16:1071482. [PMID: 36620437 PMCID: PMC9815535 DOI: 10.3389/fnins.2022.1071482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
A major challenge in treating post-traumatic stress disorder (PTSD) continues to be the large variability in responsiveness to pharmacotherapy. Only 20-30% of patients experience total remission to a specific treatment, while others demonstrate either partial remission or no response. However, this heterogeneity in response to pharmacotherapy has not been adequately addressed in animal models, since these analyze the averaged group effects, ignoring the individual variability to treatment response, which seriously compromises the translation power of such models. Here we examined the possibility of employing an "individual behavioral profiling" approach, originally developed to differentiate between "affected" and "exposed-unaffected" individuals in an animal model of PTSD, to also enable dissociating "responders" or "non-responders" after SSRI (fluoxetine) treatment. Importantly, this approach does not rely on a group averaged response to a single behavioral parameter, but considers a cluster of behavioral parameters, to individually characterize an animal as either "responder" or "non-responder" to the treatment. The main variable to assess drug efficacy thus being the proportion of "responders" following treatment. Alteration in excitatory/inhibitory (E/I) balance has been proposed as being associated with stress-related psychopathology. Toward a functional proof of concept for our behaviorally-based characterization approach, we examined the expression patterns of α1 and α2 subunits of GABAA receptor, and GluN1 and GluN2A subunits of the NMDAR receptor in the ventral hippocampus, as well as electrophysiologically local circuit activity in the dorsal dentate gyrus (DG). We demonstrate that with both parameters, treatment "responders" differed from treatment "non-responders," confirming the functional validity of the behavior-based categorization. The results suggest that the ability to respond to fluoxetine treatment may be linked to the ability to modulate excitation-inhibition balance in the hippocampus. We propose that employing the "individual behavioral profiling" approach, and the resultant novel variable of the proportion of "recovered" individuals following treatment, offers an effective translational tool to assess pharmacotherapy treatment efficacy in animal models of stress and trauma-related psychopathology.
Collapse
Affiliation(s)
- Ishita Sarkar
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | | | | | - Amir Benhos
- School of Psychological Sciences, University of Haifa, Haifa, Israel
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel,School of Psychological Sciences, University of Haifa, Haifa, Israel,The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel,*Correspondence: Gal Richter-Levin,
| |
Collapse
|
49
|
Interactions of Apigenin and Safranal with the 5HT1A and 5HT2A Receptors and Behavioral Effects in Depression and Anxiety: A Molecular Docking, Lipid-Mediated Molecular Dynamics, and In Vivo Analysis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248658. [PMID: 36557792 PMCID: PMC9783496 DOI: 10.3390/molecules27248658] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/24/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The current study utilizes in silico molecular docking/molecular dynamics to evaluate the binding affinity of apigenin and safranal with 5HT1AR/5HT2AR, followed by assessment of in vivo effects of these compounds on depressive and anxious behavior. METHODS The docking between apigenin and safranal and the 5HT1A and 5HT2A receptors was performed utilizing AutoDock Vina software, while MD and protein-lipid molecular dynamics simulations were executed by AMBER16 software. For in vivo analysis, healthy control (HC), disease control (DC), fluoxetine-, and apigenin-safranal-treated rats were tested for changes in depression and anxiety using the forced swim test (FST) and the elevated plus-maze test (EPMT), respectively. RESULTS The binding affinity estimations identified the superior interacting capacity of apigenin over safranal for 5HT1A/5HT2A receptors over 200 ns MD simulations. Both compounds exhibit oral bioavailability and absorbance. In the rodent model, there was a significant increase in the overall mobility time in the FST, while in the EPMT, there was a decrease in latency and an increase in the number of entries for the treated and HC rats compared with the DC rats, suggesting a reduction in depressive/anxiety symptoms after treatment. CONCLUSIONS Our analyses suggest apigenin and safranal as prospective medication options to treat depression and anxiety.
Collapse
|
50
|
Duarte B, Feijão E, Cruz de Carvalho R, Duarte IA, Marques AP, Maia M, Hertzog J, Matos AR, Cabrita MT, Caçador I, Figueiredo A, Silva MS, Cordeiro C, Fonseca VF. Untargeted Metabolomics Reveals Antidepressant Effects in a Marine Photosynthetic Organism: The Diatom Phaeodactylum tricornutum as a Case Study. BIOLOGY 2022; 11:1770. [PMID: 36552278 PMCID: PMC9775013 DOI: 10.3390/biology11121770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
The increased use of antidepressants, along with their increased occurrence in aquatic environments, is of concern for marine organisms. Although these pharmaceutical compounds have been shown to negatively affect marine diatoms, their mode of action in these non-target, single-cell phototrophic organisms is yet unknown. Using a Fourier-transform ion cyclotron-resonance mass spectrometer (FT-ICR-MS) we evaluated the effects of fluoxetine in the metabolomics of the model diatom Phaeodactylum tricornutum, as well as the potential use of the identified metabolites as exposure biomarkers. Diatom growth was severely impaired after fluoxetine exposure, particularly in the highest dose tested, along with a down-regulation of photosynthetic and carbohydrate metabolisms. Notably, several mechanisms that are normally down-regulated by fluoxetine in mammal organisms were also down-regulated in diatoms (e.g., glycerolipid metabolism, phosphatidylinositol signalling pathway, vitamin metabolism, terpenoid backbone biosynthesis and serotonin remobilization metabolism). Additionally, the present work also identified a set of potential biomarkers of fluoxetine exposure that were up-regulated with increasing fluoxetine exposure concentration and are of high metabolic significance following the disclosed mode of action, reinforcing the use of metabolomics approaches in ecotoxicology.
Collapse
Affiliation(s)
- Bernardo Duarte
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Eduardo Feijão
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Ricardo Cruz de Carvalho
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- cE3c, Centre for Ecology, Evolution and Environmental Changes, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, Edifício C2, Piso 5, 1749-016 Lisbon, Portugal
| | - Irina A. Duarte
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Ana Patrícia Marques
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Marisa Maia
- Université de Lorraine, LCP-A2MC, F-57000 Metz, France
| | | | - Ana Rita Matos
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- BioISI—Biosystems and Integrative Sciences Institute, Plant Functional Genomics Group, Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Maria Teresa Cabrita
- Centro de Estudos Geográficos (CEG), Instituto de Geografia e Ordenamento do Território (IGOT), Universidade de Lisboa, Rua Branca Edmée Marques, 1600-276 Lisbon, Portugal
- Associated Laboratory Terra, 1349-017 Lisbon, Portugal
| | - Isabel Caçador
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Andreia Figueiredo
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- BioISI—Biosystems and Integrative Sciences Institute, Plant Functional Genomics Group, Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Marta Sousa Silva
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Carlos Cordeiro
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Vanessa F. Fonseca
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Departamento de Biologia Animal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| |
Collapse
|