1
|
Pe LS, Pe KCS, Panmanee J, Govitrapong P, Yang JL, Mukda S. Plausible therapeutic effects of melatonin and analogs in the dopamine-associated pathophysiology of bipolar disorder. J Psychiatr Res 2025; 182:13-20. [PMID: 39793267 DOI: 10.1016/j.jpsychires.2024.12.046] [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: 09/04/2024] [Revised: 10/29/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Bipolar disorder (BD) is a significant neuropsychiatric condition characterized by marked psychological mood disturbances. Despite extensive research on the symptomatology of BD, the mechanisms underlying its development and presentation remain unknown. Consequently, potential treatments are limited, and existing medications often cause significant side effects, leading to treatment discontinuation. Dopamine (DA) has been implicated in behavioral regulation, reward systems, and mood, highlighting the importance of the dopaminergic system in BD. Elevated levels of DA and tyrosine hydroxylase are associated with the onset of manic episodes, whereas reduced levels are linked to the depressive phase. Additionally, endogenous melatonin (MEL) levels are considerably lower in patients with BD. When administered as a treatment, exogenous MEL and MEL agonists improve behavioral characteristics and significantly modulate DA-related pathophysiological pathways in BD, with minimal adverse effects achieved through MEL receptor activation. Moreover, MEL and MEL agonists offer neuroprotection by promoting physiological homeostasis during disruption. The aim of this review is to investigate and propose MEL receptors as potential novel therapeutic targets for BD. This review seeks to analyze the role of MEL and its agonists in modulating dopamine-related pathophysiological pathways, improving behavioral outcomes, and providing neuroprotection with minimal side effects.
Collapse
Affiliation(s)
- Laurence S Pe
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, 73170, Thailand.
| | - Kristine Cate S Pe
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.
| | - Jiraporn Panmanee
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, 73170, Thailand.
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Laksi, Bangkok, Thailand.
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
2
|
Koca RO, Gormus ZIS, Solak H, Celik FS, Kurar E, Kutlu S. Are the promnestic effects of neurokinin 3 receptor mediated by hippocampal neurogenesis in a Aβ-induced rat model of Alzheimer's disease? Int J Dev Neurosci 2024; 84:688-703. [PMID: 39010691 DOI: 10.1002/jdn.10362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/27/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterised by cognitive dysfunction, memory loss and mood changes. Hippocampal neurogenesis has been suggested to play a role in learning and memory. Neurokinin 3 receptor (NK3R) has been shown to be prevalent in the hippocampus region. The aim of the project was to investigate the role of hippocampal neurogenesis in the promnestic effects of NK3R agonist administration in an amyloid beta-induced AD rat model. Wistar albino rats were divided into control, Alzheimer, NK3R agonist and Alzheimer + NK3R agonist groups. The open field (OF) test and Morris water maze (MWM) test were performed for locomotor activity and memory analysis. Peptide gene expression levels (Nestin, DCX, Neuritin, MASH1, Neun, BDNF) were analysed by quantitative reverse transcription polymerase chain reaction (RT-PCR). In the OF test, the group-time relationship was found to be statistically different in the parameters of distance travelled and percentage of movement (p < 0.05). In MWM, the time to reach the platform and the time spent in the target quadrant were statistically significant between the groups (p < 0.05). Statistically significant differences were observed in gene expression levels (Nestin, DCX, Neuritin, MASH1) in the hippocampal tissue of rats between the groups (p < 0.05). NK3 receptor agonism favourably affected hippocampal neurogenesis in AD model rats. It was concluded that NK3 receptor agonism in the hippocampus, which is the first affected region in the physiopathology of AD, may be effective in both the formation of neural precursor cells and the reduction of neuronal degeneration. The positive effect of NK3R on cognitive functions may be mediated by hippocampal neurogenesis.
Collapse
Affiliation(s)
- Raviye Ozen Koca
- Department of Physiology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Z Isık Solak Gormus
- Department of Physiology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hatice Solak
- Department of Physiology, Faculty of Medicine, Kütahya Health Sciences University, Kutahya, Turkey
| | - Fatma Secer Celik
- Department of Medical Biology, Faculty of Medicine, Ankara Medipol University, Ankara, Turkey
| | - Ercan Kurar
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Selim Kutlu
- Department of Physiology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
3
|
Kajero JA, Seedat S, Ohaeri JU, Akindele A, Aina O. The effects of cannabidiol on behavioural and oxidative stress parameters induced by prolonged haloperidol administration. Acta Neuropsychiatr 2024; 36:265-275. [PMID: 36328984 DOI: 10.1017/neu.2022.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVES We investigated the influence of oral cannabidiol (CBD) on vacuous chewing movements (VCM) and oxidative stress parameters induced by short- and long-term administration of haloperidol in a rat model of tardive dyskinesia (TD). METHODS Haloperidol was administered either sub-chronically via the intraperitoneal (IP) route or chronically via the intramuscular (IM) route to six experimental groups only or in combination with CBD. VCM and oxidative stress parameters were assessed at different time points after the last dose of medication. RESULTS Oral CBD (5 mg/kg) attenuated the VCM produced by sub-chronic administration of haloperidol (5 mg/kg) but had minimal effects on the VCM produced by chronic administration of haloperidol (50 mg/kg). In both sub-chronic and chronic haloperidol groups, there were significant changes in brain antioxidant parameters compared with CBD only and the control groups. The sub-chronic haloperidol-only group had lower glutathione activity compared with sub-chronic haloperidol before CBD and the control groups; also, superoxide dismutase, catalase, and 2,2-diphenyl-1-picrylhydrazyl activities were increased in the sub-chronic (IP) haloperidol only group compared with the CBD only and control groups. Nitric oxide activity was increased in sub-chronic haloperidol-only group compared to the other groups; however, the chronic haloperidol group had increased malondialdehyde activity compared to the other groups. CONCLUSIONS Our findings indicate that CBD ameliorated VCM in the sub-chronic haloperidol group before CBD, but marginally in the chronic haloperidol group before CBD. There was increased antioxidant activity in the sub-chronic group compared to the chronic group.
Collapse
Affiliation(s)
- Jaiyeola Abiola Kajero
- Federal Neuropsychiatric Hospital, Yaba, Lagos, Nigeria
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jude U Ohaeri
- Department of Psychological Medicine, College of Medicine, University of Nigeria Enugu Campus, Enugu, Nigeria
| | - Abidemi Akindele
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Oluwagbemiga Aina
- Department of Biochemistry and Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| |
Collapse
|
4
|
Chaves-Filho A, Eyres C, Blöbaum L, Landwehr A, Tremblay MÈ. The emerging neuroimmune hypothesis of bipolar disorder: An updated overview of neuroimmune and microglial findings. J Neurochem 2024; 168:1780-1816. [PMID: 38504593 DOI: 10.1111/jnc.16098] [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: 10/13/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/21/2024]
Abstract
Bipolar disorder (BD) is a severe and multifactorial disease, with onset usually in young adulthood, which follows a progressive course throughout life. Replicated epidemiological studies have suggested inflammatory mechanisms and neuroimmune risk factors as primary contributors to the onset and development of BD. While not all patients display overt markers of inflammation, significant evidence suggests that aberrant immune signaling contributes to all stages of the disease and seems to be mood phase dependent, likely explaining the heterogeneity of findings observed in this population. As the brain's immune cells, microglia orchestrate the brain's immune response and play a critical role in maintaining the brain's health across the lifespan. Microglia are also highly sensitive to environmental changes and respond to physiological and pathological events by adapting their functions, structure, and molecular expression. Recently, it has been highlighted that instead of a single population of cells, microglia comprise a heterogeneous community with specialized states adjusted according to the local molecular cues and intercellular interactions. Early evidence has highlighted the contribution of microglia to BD neuropathology, notably for severe outcomes, such as suicidality. However, the roles and diversity of microglial states in this disease are still largely undermined. This review brings an updated overview of current literature on the contribution of neuroimmune risk factors for the onset and progression of BD, the most prominent neuroimmune abnormalities (including biomarker, neuroimaging, ex vivo studies) and the most recent findings of microglial involvement in BD neuropathology. Combining these different shreds of evidence, we aim to propose a unifying hypothesis for BD pathophysiology centered on neuroimmune abnormalities and microglia. Also, we highlight the urgent need to apply novel multi-system biology approaches to characterize the diversity of microglial states and functions involved in this enigmatic disorder, which can open bright perspectives for novel biomarkers and therapeutic discoveries.
Collapse
Affiliation(s)
- Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Women Health Research Institute, Vancouver, British Columbia, Canada
- Brain Health Cluster at the Institute on Aging & Lifelong Health (IALH), Victoria, British Columbia, Canada
| | - Capri Eyres
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Leonie Blöbaum
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Antonia Landwehr
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Women Health Research Institute, Vancouver, British Columbia, Canada
- Brain Health Cluster at the Institute on Aging & Lifelong Health (IALH), Victoria, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Quebec, Canada
- Department of Molecular Medicine, Université Laval, Québec City, Quebec, Canada
| |
Collapse
|
5
|
Valvassori SS, Varela RB, Resende WR, Possamai-Della T, Borba LDA, Behenck JP, Réus GZ, Quevedo J. Antidepressant Effect of Sodium Butyrate is Accompanied by Brain Epigenetic Modulation in Rats Subjected to Early or Late Life Stress. Curr Neurovasc Res 2024; 20:586-598. [PMID: 38288841 DOI: 10.2174/0115672026277345240115101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/15/2023] [Accepted: 10/22/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Major depression has a complex and multifactorial etiology constituted by the interaction between genetic and environmental factors in its development. OBJECTIVE The aim of this study was to evaluate the effects of sodium butyrate (SD) on epigenetic enzyme alterations in rats subjected to animal models of depression induced by maternal deprivation (MD) or chronic mild stress (CMS). METHODS To induce MD, male Wistar rats were deprived of maternal care during the first 10 days of life. To induce CMS, rats were subjected to the CMS for 40 days. Adult rats were then treated with daily injections of SD for 7 days. Animals were subjected to the forced swimming test (FST), and then, histone deacetylase (HDAC), histone acetyltransferase (HAT), and DNA methyltransferase (DNMT) activities were evaluated in the brain. RESULTS MD and CMS increased immobility time in FST and increased HDAC and DNMT activity in the animal brains. SD reversed increased immobility induced by both animal models and the alterations in HDAC and DNMT activities. There was a positive correlation between enzyme activities and immobility time for both models. HDAC and DNMT activities also presented a positive correlation between themselves. CONCLUSION These results suggest that epigenetics can play an important role in major depression pathophysiology triggered by early or late life stress and its treatment.
Collapse
Affiliation(s)
- Samira Silva Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Roger Bitencourt Varela
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Wilson Rodrigues Resende
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Taise Possamai-Della
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laura de Araujo Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
- Faillace Department of Psychiatry and Behavioral Sciences, Center for Interventional Psychiatry, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| |
Collapse
|
6
|
Menegas S, Keller GS, Possamai-Della T, Aguiar-Geraldo JM, Quevedo J, Valvassori SS. Potential mechanisms of action of resveratrol in prevention and therapy for mental disorders. J Nutr Biochem 2023; 121:109435. [PMID: 37669710 DOI: 10.1016/j.jnutbio.2023.109435] [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: 03/14/2022] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023]
Abstract
There is a growing body of evidence about the potential of diet and nutrients to improve the population's mental health and the treatment of psychiatric disorders. Some studies have suggested that resveratrol has therapeutic properties in mental disorders, such as major depressive disorder, bipolar disorder, Alzheimer's disease, and autism. In addition, resveratrol is known to induce several benefits modulated by multiple synergistic pathways, which control oxidative stress, inflammation, and cell death. This review collects the currently available data from animal and human studies and discusses the potential mechanisms of action of resveratrol in prevention and therapy for psychiatric disorders.
Collapse
Affiliation(s)
- Samira Menegas
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Gabriela S Keller
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Taise Possamai-Della
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Jorge M Aguiar-Geraldo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA; Center for Interventional Psychiatry, Faillace Department of Psychiatry and Behavior Sciences, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, Texas, USA
| | - Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil.
| |
Collapse
|
7
|
Reinhardt PR, Theis CDC, Juckel G, Freund N. Rodent models for mood disorders - understanding molecular changes by investigating social behavior. Biol Chem 2023; 404:939-950. [PMID: 37632729 DOI: 10.1515/hsz-2023-0190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
Mood disorders, including depressive and bipolar disorders, are the group of psychiatric disorders with the highest prevalence and disease burden. However, their pathophysiology remains poorly understood. Animal models are an extremely useful tool for the investigation of molecular mechanisms underlying these disorders. For psychiatric symptom assessment in animals, a meaningful behavioral phenotype is needed. Social behaviors constitute naturally occurring complex behaviors in rodents and can therefore serve as such a phenotype, contributing to insights into disorder related molecular changes. In this narrative review, we give a fundamental overview of social behaviors in laboratory rodents, as well as their underlying neuronal mechanisms and their assessment. Relevant behavioral and molecular changes in models for mood disorders are presented and an outlook on promising future directions is given.
Collapse
Affiliation(s)
- Patrick R Reinhardt
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
- International Graduate School of Neuroscience, Ruhr-University Bochum, D-44801 Bochum, Germany
| | - Candy D C Theis
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
| | - Georg Juckel
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
| |
Collapse
|
8
|
Valvassori SS, Quevedo J, Scaini G. Did we finally find a good animal model for bipolar disorder? Mol Psychiatry 2023; 28:2622-2623. [PMID: 37365242 DOI: 10.1038/s41380-023-02151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Affiliation(s)
- Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Center for Interventional Psychiatry, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Giselli Scaini
- Center for Interventional Psychiatry, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
9
|
Vasudevan R, Kandasamy G, Aldahish A, Almanasef M, Khobrani M, Shorog E, Prabahar K, Mohammed Alsawaq E, Wahab S, Begum MY, Paulsamy P. Antidepressant and Anxiolytic Potentials of the Chewing Stick, Salvadora persica. J Trop Med 2023; 2023:9683240. [PMID: 37096220 PMCID: PMC10122599 DOI: 10.1155/2023/9683240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/10/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023] Open
Abstract
Materials and Methods Salvadora persica stem bark was extracted with two different solvents, i.e., ethyl acetate and water, and preliminary phytochemical screening was performed. Two behavioral models were used: an elevated plus maze test (EPM) and the light and dark model test for anxiolytic parameters, and a forced swim test (FST) for antidepressant effects. Healthy mice weighing 18-40 gms were treated orally in four groups (n = 6), i.e., negative control treated with normal saline and positive control with 1 mg/kg diazepam (EPM) and 30 mg/kg fluoxetine (FST), and the test groups were treated with 500 mg/kg of aqueous and ethyl acetate Sp extract. The number of entries and duration spent in the open arm for 5 minutes were the parameters for evaluating the anxiolytic activity (EPM). Duration of immobility was measured for 5 min in the FST model. Results In EPM, both the Sp extracts significantly (p < 0.005) increased the number of entries and the time spent in the open arms and was much similar to those of diazepam. Similarly, these extracts and fluoxetine significantly (p < 0.005) decreased the immobility time in FST. Conclusion The results suggest the therapeutic potential of Salvadora persica an alternative in the management of comorbid anxiety and depression.
Collapse
Affiliation(s)
- Rajalakshimi Vasudevan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Geetha Kandasamy
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Afaf Aldahish
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Mona Almanasef
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Moteb Khobrani
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Eman Shorog
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Kousalya Prabahar
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Pharmacy Practice, Faculty of Pharmacy, Dr. M.G.R Educational and Research Institute, Velappanchavadi, Chennai, Tamil Nadu, India
| | - Enas Mohammed Alsawaq
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Saudi Arabia
| | - M. Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | | |
Collapse
|
10
|
Rajkhowa B, Mehan S, Sethi P, Prajapati A, Suri M, Kumar S, Bhalla S, Narula AS, Alshammari A, Alharbi M, Alkahtani N, Alghamdi S, Kalfin R. Activating SIRT-1 Signalling with the Mitochondrial-CoQ10 Activator Solanesol Improves Neurobehavioral and Neurochemical Defects in Ouabain-Induced Experimental Model of Bipolar Disorder. Pharmaceuticals (Basel) 2022; 15:ph15080959. [PMID: 36015107 PMCID: PMC9415079 DOI: 10.3390/ph15080959] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder (BD) is a chronic mental illness characterized by mood fluctuations that range from depressive lows to manic highs. Several studies have linked the downregulation of SIRT-1 (silent mating type information regulation-2 homologs) signaling to the onset of BD and other neurological dysfunctions. This research aimed to look into the neuroprotective potential of Solanesol (SNL) in rats given ICV-Ouabain injections, focusing on its effect on SIRT-1 signaling activation in the brain. Ouabain, found in hypothalamic and medullary neurons, is an endogenous inhibitor of brain Na+/K+ ATPase. The inhibition of brain Na+/K+ ATPase by Ouabain may also result in changes in neurotransmission within the central nervous system. SNL is a Solanaceae family active phytoconstituent produced from the plant Nicotiana tabacum. SNL is used as a precursor for the production of CoQ10 (Coenzyme Q10), a powerful antioxidant and neuroprotective compound. In the current study, lithium (Li), an important mood stabilizer drug, was used as a control. This study looked at the neuroprotective potential of SNL at dosages of 40 and 80 mg/kg in ICV-OUA injections that caused BD-like neurobehavioral and neurochemical defects in Wistar rats. Wistar rats were placed into eight groups (n = 6) and administered 1 mM/0.5 µL ICV-OUA injections for three days. Neurochemical assessments were done in rat brain homogenates, CSF, and blood plasma samples at the end of the experiment protocol schedule. Long-term SNL and lithium administration have been shown to decrease the number of rearing and crossings and reduce time spent in the center, locomotor activities, and immobility time. Solansesol treatment gradually raises the amount of Na+/K+ ATPase, limiting the severity of behavioural symptoms. These findings also revealed that SNL increases the levels of SIRT-1 in CSF, blood plasma, and brain homogenate samples. Moreover, in rat brain homogenates and blood plasma samples, SNL modulates apoptotic markers such as Caspase-3, Bax (pro-apoptotic), and Bcl-2 (anti-apoptotic). Mitochondrial-ETC complex enzymes, including complex-I, II, IV, V, and CoQ10, were also restored following long-term SNL treatment. Furthermore, SNL lowered inflammatory cytokines (TNF-α, IL-1β) levels while restoring neurotransmitter levels (serotonin, dopamine, glutamate, and acetylcholine) and decreasing oxidative stress markers. Histological examinations also validated Solanesol’s protective effect. As a result, our findings suggest that SNL, as a SIRT-1 signalling activator, may be a promising therapeutic approach for BD-like neurological dysfunctions.
Collapse
Affiliation(s)
- Bidisha Rajkhowa
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
- Correspondence: ; Tel.: +91-8059889909
| | - Pranshul Sethi
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
| | - Aradhana Prajapati
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
| | - Manisha Suri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
| | - Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
| | - Sonalika Bhalla
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, India; (B.R.); (P.S.); (A.P.); (M.S.); (S.K.); (S.B.)
| | - Acharan S. Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA;
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (A.A.); (M.A.); (N.A.); (S.A.)
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (A.A.); (M.A.); (N.A.); (S.A.)
| | - Nora Alkahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (A.A.); (M.A.); (N.A.); (S.A.)
| | - Saeed Alghamdi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (A.A.); (M.A.); (N.A.); (S.A.)
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, 1113 Sofia, Bulgaria;
- Department of Healthcare, South-West University “Neofit Rilski”, Ivan Mihailov St. 66, 2700 Blagoevgrad, Bulgaria
| |
Collapse
|
11
|
Gbadamosi IT, Henneh IT, Aluko OM, Yawson EO, Fokoua AR, Koomson A, Torbi J, Olorunnado SE, Lewu FS, Yusha'u Y, Keji-Taofik ST, Biney RP, Tagoe TA. Depression in Sub-Saharan Africa. IBRO Neurosci Rep 2022; 12:309-322. [PMID: 35746974 PMCID: PMC9210463 DOI: 10.1016/j.ibneur.2022.03.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 03/14/2022] [Indexed: 12/23/2022] Open
Abstract
Mood disorders can be considered among the most common and debilitating mental disorders. Major depression, as an example of mood disorders, is known to severely reduce the quality of life as well as psychosocial functioning of those affected. Its impact on the burden of disease worldwide has been enormous, with the World Health Organisation projecting depression to be the leading cause of mental illness by 2030. Despite several studies on the subject, little has been done to contextualise the condition in Africa, coupled with the fact that there is still much to be understood on the subject. This review attempts to shed more light on the prevalence of depression in Sub-Saharan Africa (SSA), its pathophysiology, risk factors, diagnosis and the experimental models available to study depression within the sub-region. It also evaluates the contribution of the sub-region to the global research output of depression as well as bottlenecks associated with full exploitation of the sub region's resources to manage the disorder.
Collapse
Affiliation(s)
- Ismail Temitayo Gbadamosi
- Department of Anatomy, University of Ilorin, Nigeria
- Laboratory for Translational Research in Neuropsychiatric Disorders, BRAINCITY Nencki-EMBL Center of Excellence for Neural Plasticty and Brain Disorders, Warsaw, Poland
| | - Isaac Tabiri Henneh
- Department of Pharmacotherapeutics and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Ghana
| | - Oritoke Modupe Aluko
- Department of Physiology, School of Basic Medical Sciences, Federal University of Technology, Akure, Nigeria
| | | | | | - Awo Koomson
- Department of Pharmacology and Toxicology University of Ghana, Ghana
| | - Joseph Torbi
- Department of Pharmacology and Toxicology University of Ghana, Ghana
| | | | | | - Yusuf Yusha'u
- Department of Human Physiology Ahmadu Bello University, Zaria, Nigeria
| | | | - Robert Peter Biney
- Department of Pharmacotherapeutics and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Ghana
| | | |
Collapse
|
12
|
Cheiran Pereira G, Piton E, Moreira Dos Santos B, Ramanzini LG, Muniz Camargo LF, Menezes da Silva R, Bochi GV. Microglia and HPA axis in depression: An overview of participation and relationship. World J Biol Psychiatry 2022; 23:165-182. [PMID: 34100334 DOI: 10.1080/15622975.2021.1939154] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Objectives: This narrative review article provides an overview on the involvement of microglia and the hypothalamic-pituitary-adrenal (HPA) axis in the pathophysiology of depression, as well investigates the mutual relationship between these two entities: how microglial activation can contribute to the dysregulation of the HPA axis, and vice versa.Methods: Relevant studies and reviews already published in the Pubmed electronic database involving the themes microglia, HPA axis and depression were used to meet the objectives.Results: Exposition to stressful events is considered a common factor in the mechanisms proposed to explain the depressive disorder. Stress can activate microglial cells, important immune components of the central nervous system (CNS). Moreover, another system involved in the physiological response to stressors is the hypothalamic-pituitary-adrenal (HPA) axis, the main stress response system responsible for the production of the glucocorticoid hormone (GC). Also, mediators released after microglial activation can stimulate the HPA axis, inducing production of GC. Likewise, high levels of GCs are also capable of activating microglia, generating a vicious cycle.Conclusion: Immune and neuroendocrine systems seems to work in a coordinated manner and that their dysregulation may be involved in the pathophysiology of depression since neuroinflammation and hypercortisolism are often observed in this disorder.
Collapse
Affiliation(s)
- Gabriele Cheiran Pereira
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil.,Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Elisa Piton
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Brenda Moreira Dos Santos
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil.,Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Luis Guilherme Ramanzini
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Luis Fernando Muniz Camargo
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Rossano Menezes da Silva
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Guilherme Vargas Bochi
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil.,Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
13
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
14
|
Does Sertraline Affect Hypothalamic Food Intake Peptides in the Rat Experimental Model of Chronic Mild Stress-Induced Depression? Neurochem Res 2022; 47:1299-1316. [PMID: 35080689 DOI: 10.1007/s11064-022-03529-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/12/2022] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
Depression is a chronic, recurrent and life-threatening disease affecting approximately 15% of the world population. Depression is responsible for neuropathologies like decreased neurogenesis and increased dendritic atrophy. Antidepressant treatments increase hippocampal neurogenesis and neurotrophic factor expression. Based on this information, it was aimed to investigate effect of sertraline on depression in rats with chronic mild stress (CMS) model and to determine how it affects cell proliferation and hypothalamic peptide levels in hypothalamus. 56 adult male Wistar albino; control, depression(D), depression + sertraline, sertraline were divided into groups. Various stressors were applied to D for 30 days. Open field test (OFT) and forced swimming test (FST) were conducted to check whether the animals were depressed. On the 16th day osmotic minipump was placed subcutaneously and sertraline (10 mg/kg/day) was administered for 15 days. Behavior tests were done. Hypothalamic peptide gene expression levels were analyzed by quantitative RT-PCR. Statistical evaluations were made using ANOVA. It caused a decrease in the percentage of movement in the D and control groups in the OFT, an increase in the immobility time in the D group in the FST, and an increase in the swimming behavior in the DS group. Animals did not show any anxiological behavior based on the elevated plus maze test results. CMS caused a decrease in GLUT2 and NPY gene expression in the hypothalamus of animals, an increase in POMC and FGFR2, and an increase in IGFIR and GLUT2 gene expression in the DS group. Sertraline has been shown to ameliorate the effects of CMS-induced depression. Sertraline is thought to have a positive regulatory effect on both the formation of neural precursor cells and the survival of newly formed neurons in the hypothalamus. Newly formed neurons in the hypothalamus express food intake-related NPY, POMC, GLUT2 neurons, and thus hypothalamic tanycytes may play a key role in the control of energy metabolism.
Collapse
|
15
|
Recart VM, Spohr L, Soares MSP, Luduvico KP, Stefanello FM, Spanevello RM. Therapeutic approaches employing natural compounds and derivatives for treating bipolar disorder: emphasis on experimental models of the manic phase. Metab Brain Dis 2021; 36:1481-1499. [PMID: 34264451 DOI: 10.1007/s11011-021-00776-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/06/2021] [Indexed: 12/19/2022]
Abstract
Bipolar disorder (BD) is a complex psychiatric disease characterized by mood swings that include episodes of mania and depression. Given its cyclical nature, BD is especially hard to model; however, the standard practice has been to mimic manic episodes in animal models. Despite scientific advances, the pathophysiology of BD is not fully understood, and treatment remains limited. In the last years, natural products have emerged as potential neuroprotective agents for the treatment of psychiatric diseases. Thus, the aim of this review was to explore the therapeutic potential of natural compounds and derivatives against BD, taking into account preclinical and clinical studies. Reliable articles indexed in databases such as PubMed, Web of Science and Science Direct were used. In clinical studies, treatment with herbal plants extracts, omega-3, inositol, n-acetylcysteine and vitamin D has been associated with a clinical improvement in symptoms of mania and depression in BD patients. In animal models, it has been shown that red fruits extracts, curcumin, quercetin, gallic acid, alpha-lipoic acid and carvone can modulate many neurochemical pathways involved in the pathophysiology of manic episodes. Thus, this review appointed the advances in the consumption of natural compounds and derivatives as an important therapeutic strategy to mitigate the symptoms of BD.
Collapse
Affiliation(s)
- Vânia Machado Recart
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão Do Leão, Pelotas, RS, 96010-900, Brazil
| | - Luiza Spohr
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão Do Leão, Pelotas, RS, 96010-900, Brazil
| | - Mayara Sandrielly Pereira Soares
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão Do Leão, Pelotas, RS, 96010-900, Brazil
| | - Karina Pereira Luduvico
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão Do Leão, Pelotas, RS, 96010-900, Brazil.
| |
Collapse
|
16
|
Tchekalarova J, Ivanova N, Nenchovska Z, Tzoneva R, Stoyanova T, Uzunova V, Surcheva S, Tzonev A, T Angelova V, Andreeva-Gateva P. Evaluation of neurobiological and antioxidant effects of novel melatonin analogs in mice. Saudi Pharm J 2020; 28:1566-1579. [PMID: 33424250 PMCID: PMC7783092 DOI: 10.1016/j.jsps.2020.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022] Open
Abstract
Based on the pharmacophore model of melatonin (MT1) receptor, we recently synthesized a series of indole derivatives that showed anticonvulsant activity with low neurotoxicity and hepatotoxicity in rodents. In the present study, the three most potent C3-modified derivatives with hydrazine structure 3c, 3e, and 3f, with 2-chlorophenyl, 2-furyl, and 2-thienyl fragments, respectively, were selected, and their neurobiological activity was explored in mice. In Experiment #1, the dose-dependent anxiolytic effect of a single i.p. administration of the novel compounds at doses of 10, 30, and 60 mg/kg were studied in the open field (OF) test. In Experiment#2, the analgesic effect of 3c, 3e, and 3f (30–100 mg/kg) was tested in the hot plate test and formalin test. Experiment#3 was designed to assess the antidepressant-like activity of 3c, 3e, and 3f (10–60 mg/kg). The forced swimming test (FST) and tail suspension test (TST)-induced effect on markers of oxidative stress in the frontal cortex (FC), and the hippocampus was evaluated. Melatonin was used in the same doses as melatonin analogs in all three experiments as a positive control. Desipramine (10 mg/kg) was also applied as a control in the FST. The three melatonin analogs bearing hydrazide/hydrazone substitution at 3C of the indol scaffold demonstrated improved antidepressant-like activity compared to the melatonin. The tested substances are devoided of anxiolytic effects. The antioxidant activity of the melatonin analogs and analgesic potential is comparable to that of melatonin. The 3C substitution with hydrazide/hydrazone moiety substantially contributes to the antidepressant and antioxidant activity of the melatonin analogs.
Collapse
Affiliation(s)
- Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Natasha Ivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Zlatina Nenchovska
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Rumiana Tzoneva
- Institute of Biophysics and Biomedical Engineering, BAS, Sofia, Bulgaria
| | - Tzveta Stoyanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Veselina Uzunova
- Institute of Biophysics and Biomedical Engineering, BAS, Sofia, Bulgaria
| | - Slavina Surcheva
- Department of Pharmacology and Toxicology, Medical Faculty, Medical University of Sofia, Bulgaria
| | - Alex Tzonev
- Department of Pharmacology and Toxicology, Medical Faculty, Medical University of Sofia, Bulgaria
| | - Violina T Angelova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, Bulgaria
| | - Pavlina Andreeva-Gateva
- Department of Pharmacology and Toxicology, Medical Faculty, Medical University of Sofia, Bulgaria
| |
Collapse
|
17
|
Pereira GC, Piton E, dos Santos BM, da Silva RM, de Almeida AS, Dalenogare DP, Schiefelbein NS, Fialho MFP, Moresco RN, dos Santos GT, Marchesan S, Bochi GV. Apocynin as an antidepressant agent: in vivo behavior and oxidative parameters modulation. Behav Brain Res 2020; 388:112643. [DOI: 10.1016/j.bbr.2020.112643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022]
|
18
|
Duan J, Xie P. The potential for metabolomics in the study and treatment of major depressive disorder and related conditions. Expert Rev Proteomics 2020; 17:309-322. [PMID: 32516008 DOI: 10.1080/14789450.2020.1772059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/15/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Major depressive disorder (MDD) is a common mental disease, associated with a debilitating condition and high prevalence. Although the underlying mechanism of MDD remains to be elucidated, several factors, including social, biological, and psychological factors, have been associated with disease pathogenesis. Metabolomics can provide new insights into the prognosis, treatment response, and related biomarkers associated with MDD at the metabolic level. AREAS COVERED In this review, we investigated the metabolic changes identified in different bio-samples from animal models of depression and MDD patients. Moreover, we summarized the metabolites associated with antidepressant treatment responses. Keywords used for the literature searches were 'depression' [MeSH] and 'metabolomics' [MeSH], in PubMed. EXPERT OPINION Metabolomic evidence in humans has indicated that amino acid metabolism, energy metabolism, and lipid metabolism are the primary metabolic alterations that are observed in the etiology of MDD, and animal models serve as an important theoretical reference in this field. Metabolomics has shed new light on the pathogenic mechanisms and treatment responses during MDD; however, study results are not always consistent. The application of metabolomic results to clinical practice will require the integration of different biological samples and other omics studies, as well as the clinical validation of study findings.
Collapse
Affiliation(s)
- Jiajia Duan
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, Chongqing Medical University , Chongqing, China
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University , Chongqing, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, Chongqing Medical University , Chongqing, China
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University , Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| |
Collapse
|
19
|
Varela RB, Resende WR, Dal-Pont GC, Gava FF, Tye SJ, Quevedo J, Valvassori SS. HDAC inhibitors reverse mania-like behavior and modulate epigenetic regulatory enzymes in an animal model of mania induced by Ouabain. Pharmacol Biochem Behav 2020; 193:172917. [PMID: 32222371 DOI: 10.1016/j.pbb.2020.172917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The etiology of bipolar disorder (BD) is multifactorial, involving both environmental and genetic factors. Current pharmacological treatment is associated with several side effects, which are the main reason patients discontinue treatment. Epigenetic alterations have been studied for their role in the pathophysiology of BD, as they bridge the gap between gene and environment. OBJECTIVE Evaluate the effects of histone deacetylase inhibitors on behavior and epigenetic enzymes activity in a rat model of mania induced by ouabain. METHODS Adult male rats were subjected to a single intracerebroventricular injection of ouabain (10-3 M) followed by 7 days of valproate (200 mg/kg) or sodium butyrate (600 mg/kg) administration. Locomotor and exploratory activities were evaluated in the open-field test. Histone deacetylase, DNA methyltransferase, and histone acetyltransferase activity were assessed in the frontal cortex, hippocampus, and striatum. RESULTS Ouabain induced hyperactivity in rats, which was reversed by valproate and sodium butyrate treatment. Ouabain did not alter the activity of any of the enzymes evaluated. However, valproate and sodium butyrate decreased the activity of histone deacetylase and DNA methyltransferase. Moreover, there was a positive correlation between these two enzymes. CONCLUSION These results suggest that targeting epigenetic mechanisms may play an important role in mania-like behavior management.
Collapse
Affiliation(s)
- Roger B Varela
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia; Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC) -, Criciúma, SC, Brazil
| | - Wilson R Resende
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC) -, Criciúma, SC, Brazil
| | - Gustavo C Dal-Pont
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC) -, Criciúma, SC, Brazil
| | - Fernanda F Gava
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC) -, Criciúma, SC, Brazil
| | - Susannah J Tye
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC) -, Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth) -, Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth) -, Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences -, Houston, TX, USA
| | - Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC) -, Criciúma, SC, Brazil.
| |
Collapse
|
20
|
Demin KA, Lakstygal AM, Chernysh MV, Krotova NA, Taranov AS, Ilyin NP, Seredinskaya MV, Tagawa N, Savva AK, Mor MS, Vasyutina ML, Efimova EV, Kolesnikova TO, Gainetdinov RR, Strekalova T, Amstislavskaya TG, de Abreu MS, Kalueff AV. The zebrafish tail immobilization (ZTI) test as a new tool to assess stress-related behavior and a potential screen for drugs affecting despair-like states. J Neurosci Methods 2020; 337:108637. [PMID: 32081675 DOI: 10.1016/j.jneumeth.2020.108637] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Affective disorders, especially depression and anxiety, are highly prevalent, debilitating mental illnesses. Animal experimental models are a valuable tool in translational affective neuroscience research. A hallmark phenotype of clinical and experimental depression, the learned helplessness, has become a key target for 'behavioral despair'-based animal models of depression. The zebrafish (Danio rerio) has recently emerged as a promising novel organism for affective disease modeling and CNS drug screening. Despite being widely used to assess stress and anxiety-like behaviors, there are presently no clear-cut despair-like models in zebrafish. NEW METHOD Here, we introduce a novel behavioral paradigm, the zebrafish tail immobilization (ZTI) test, as a potential tool to assess zebrafish despair-like behavior. Conceptually similar to rodent 'despair' models, the ZTI protocol involves immobilizing the caudal half of the fish body for 5 min, leaving the cranial part to move freely, suspended vertically in a small beaker with water. RESULTS To validate this model, we used exposure to low-voltage electric shock, alarm pheromone, selected antidepressants (sertraline and amitriptyline) and an anxiolytic drug benzodiazepine (phenazepam), assessing the number of mobility episodes, time spent 'moving', total distance moved and other activity measures of the cranial part of the body, using video-tracking. Both electric shock and alarm pheromone decreased zebrafish activity in this test, antidepressants increased it, and phenazepam was inactive. Furthermore, a 5-min ZTI exposure increased serotonin turnover, elevating the 5-hydroxyindoleacetic acid/serotonin ratio in zebrafish brain, while electric shock prior to ZTI elevated both this and the 3,4-dihydroxyphenylacetic acid/dopamine ratios. In contrast, preexposure to antidepressants sertraline and amitriptyline lowered both ratios, compared to the ZTI test-exposed fish. COMPARISON WITH EXISTINGMETHOD(S) The ZTI test is the first despair-like experimental model in zebrafish. CONCLUSIONS Collectively, this study suggests the ZTI test as a potentially useful protocol to assess stress-/despair-related behaviors, potentially relevant to CNS drug screening and behavioral phenotyping of zebrafish.
Collapse
Affiliation(s)
- Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| | - Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Maria V Chernysh
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Natalia A Krotova
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Aleksandr S Taranov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Maria V Seredinskaya
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Natsuki Tagawa
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Anna K Savva
- Laboratory of Insect Biopharmacology and Immunology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russia
| | - Mikael S Mor
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Marina L Vasyutina
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Evgeniya V Efimova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatyana O Kolesnikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatyana Strekalova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Maastricht University, Maastricht, The Netherlands; Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | | | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
21
|
Glucocorticoid and brain-derived neurotrophic factor relationship: a brief investigation into the model of depression by chronic administration of corticosterone. Behav Pharmacol 2020; 31:407-412. [PMID: 32040016 DOI: 10.1097/fbp.0000000000000547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Depression is considered a common mental disorder that affects more than 300 million people worldwide. Despite this high incidence, its etiology is not completely elucidated instigating further studies. For this purpose, different animal models are used to study routes and molecular changes involved in depression, among them the chronic administration of corticosterone. However, the knowledge about neurochemical changes after this protocol is still controversial. In this work, we evaluated serum corticosterone levels, adrenal/body weight ratio, as well as glucocorticoid receptor and brain-derived neurotrophic factor protein expression and its receptor, tropomyosin-receptor kinase B. These analyzes were performed on prefrontal cortex, hippocampus, and striatum samples taken of mice after 21 days of administration of corticosterone. Exposure to corticosterone reduced the serum corticosterone levels and the adrenal/body weight ratio. Moreover, the glucocorticoid receptor and tyrosine-receptor kinase B expression were increased in the hippocampus while the brain-derived neurotrophic factor expression was reduced in the prefrontal cortex. We also found a positive correlation between the expression of glucocorticoid receptor and tyrosine-receptor kinase B and our results suggest a possible relationship between the glucocorticoid/glucocorticoid receptor and brain-derived neurotrophic factor/tropomyosin-receptor kinase B routes after chronic corticosterone administration. To our knowledge, this is the first study that evaluate these parameters concomitantly in important mood-related structures. In addition, these results may be useful to other research groups seeking to explore new pathways and substances with therapeutic potential to treat this silent epidemic.
Collapse
|
22
|
Menegas S, Dal-Pont GC, Cararo JH, Varela RB, Aguiar-Geraldo JM, Possamai-Della T, Andersen ML, Quevedo J, Valvassori SS. Efficacy of folic acid as an adjunct to lithium therapy on manic-like behaviors, oxidative stress and inflammatory parameters in an animal model of mania. Metab Brain Dis 2020; 35:413-425. [PMID: 31840201 DOI: 10.1007/s11011-019-00503-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022]
Abstract
Evaluate the efficacy of folic acid (FA) as a therapeutic adjunct to lithium (Li) on the manic-like behaviors as well as parameters of oxidative stress and inflammation in an animal model of mania induced by m-amphetamine (m-AMPH). Wistar rats first received m-AMPH or saline (NaCl 0.9%, Sal) for 14 days. Between the 8th and 14th day, rats were treated with water, Li, FA or a combination of thereof drugs (Li + FA). Manic-like behaviors were assessed in the open-field test. Oxidative stress and inflammation parameters were assessed in the frontal cortex, striatum, and hippocampus. Administration of m-AMPH in rats significantly enhanced the exploratory and locomotor behaviors, as well as the risk-taking and stereotypic behaviors. Li + FA reversed these behavioral alterations elicited by m-AMPH. Administration of this psychostimulant also increased oxidative damage to lipids and proteins, whereas Li + FA reversed these oxidative damages. m-AMPH also induced an increase in the glutathione peroxidase (GPx) activity and a decrease in the glutathione reductase (GR) activity. Li + FA reversed the alteration in GR activity, but not in GPx activity. In addition, m-AMPH increased the IL-1β and TNF-α levels in the rat brain; Li + FA combined therapy reversed the alterations on these inflammatory parameters. FA administration per se reduced the increased TNF-α content induced by m-AMPH. Present study provides evidence that FA is effective as an adjunct to Li standard therapy on manic-like behaviors, oxidative stress and inflammatory parameters in a model of mania induced by m-AMPH.
Collapse
Affiliation(s)
- Samira Menegas
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gustavo C Dal-Pont
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - José H Cararo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Roger B Varela
- Queensland Brain Institute, The Universty of Queensland, St Lucia, QLD, 4072, Australia
| | - Jorge M Aguiar-Geraldo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Taise Possamai-Della
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Monica L Andersen
- Department of Psychobiology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
23
|
Stukalin Y, Lan A, Einat H. Revisiting the validity of the mouse tail suspension test: Systematic review and meta-analysis of the effects of prototypic antidepressants. Neurosci Biobehav Rev 2020; 112:39-47. [PMID: 32006552 DOI: 10.1016/j.neubiorev.2020.01.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 12/26/2019] [Accepted: 01/28/2020] [Indexed: 01/18/2023]
Abstract
Animal models in neuropsychiatric research need validation. One way to address external validity is systematic reviews and meta-analyses. The present study presents a meta-analysis of the effects of antidepressants in the mouse tail suspension test (TST). A PubMed search identified studies that examined imipramine and fluoxetine effects in the TST. Inclusion criteria were testing in the light phase; trial duration was six minutes; immobility time scored 6 or (last) 4 min; adult mice; acute intraperitoneal (IP) administration. Effect sizes (ES) were estimated using Cohen's d, heterogeneity of ES with Cochran's Q test, correlations between dose and ES with Pearson's correlation and differences between strains with Analysis of variance. Results show that antidepressants decrease immobility time in the TST and a correlation between drug dose and ES but no effects of strain. We suggest that the TST is a valid tool to quantitatively, consistently and reproducibly capture the immobility-reducing aspects of fluoxetine and imipramine and that the lack of strain effects is due to small number of experiments in many of the strains.
Collapse
Affiliation(s)
- Yelena Stukalin
- School of Behavioral Sciences, Tel Aviv-Yaffo Academic College, Tel-Aviv, Israel
| | - Anat Lan
- School of Behavioral Sciences, Tel Aviv-Yaffo Academic College, Tel-Aviv, Israel; The open University, Israel
| | - Haim Einat
- School of Behavioral Sciences, Tel Aviv-Yaffo Academic College, Tel-Aviv, Israel.
| |
Collapse
|
24
|
Neville V, Nakagawa S, Zidar J, Paul ES, Lagisz M, Bateson M, Løvlie H, Mendl M. Pharmacological manipulations of judgement bias: A systematic review and meta-analysis. Neurosci Biobehav Rev 2020; 108:269-286. [PMID: 31747552 PMCID: PMC6966323 DOI: 10.1016/j.neubiorev.2019.11.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 01/11/2023]
Abstract
Validated measures of animal affect are crucial to research spanning numerous disciplines. Judgement bias, which assesses decision-making under ambiguity, is a promising measure of animal affect. One way of validating this measure is to administer drugs with affect-altering properties in humans to non-human animals and determine whether the predicted judgement biases are observed. We conducted a systematic review and meta-analysis using data from 20 published research articles that use this approach, from which 557 effect sizes were extracted. Pharmacological manipulations overall altered judgement bias at the probe cues as predicted. However, there were several moderating factors including the neurobiological target of the drug, whether the drug induced a relatively positive or negative affective state in humans, dosage, and the presented cue. This may partially reflect interference from adverse effects of the drug which should be considered when interpreting results. Thus, the overall pattern of change in animal judgement bias appears to reflect the affect-altering properties of drugs in humans, and hence may be a valuable measure of animal affective valence.
Collapse
Affiliation(s)
- Vikki Neville
- Centre for Behavioural Biology, Bristol Veterinary School, University of Bristol, Langford BS40 5DU, United Kingdom.
| | - Shinichi Nakagawa
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Sydney, NSW 2052, Australia
| | - Josefina Zidar
- The Department of Physics, Chemistry and Biology, IFM Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Elizabeth S Paul
- Centre for Behavioural Biology, Bristol Veterinary School, University of Bristol, Langford BS40 5DU, United Kingdom
| | - Malgorzata Lagisz
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Sydney, NSW 2052, Australia
| | - Melissa Bateson
- Institute of Neuroscience and Centre for Behaviour and Evolution, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Hanne Løvlie
- The Department of Physics, Chemistry and Biology, IFM Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Michael Mendl
- Centre for Behavioural Biology, Bristol Veterinary School, University of Bristol, Langford BS40 5DU, United Kingdom
| |
Collapse
|
25
|
Tran HQ, Shin EJ, Saito K, Tran TV, Phan DH, Sharma N, Kim DW, Choi SY, Jeong JH, Jang CG, Cheong JH, Nabeshima T, Kim HC. Indoleamine-2,3-dioxygenase-1 is a molecular target for the protective activity of mood stabilizers against mania-like behavior induced by d-amphetamine. Food Chem Toxicol 2019; 136:110986. [PMID: 31760073 DOI: 10.1016/j.fct.2019.110986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/07/2023]
Abstract
It is recognized that d-amphetamine (AMPH)-induced hyperactivity is thought to be a valid animal model of mania. In the present study, we investigated whether a proinflammatory oxidative gene indoleamine-2,3-dioxygenase (IDO) is involved in AMPH-induced mitochondrial burden, and whether mood stabilizers (i.e., lithium and valproate) modulate IDO to protect against AMPH-induced mania-like behaviors. AMPH-induced IDO-1 expression was significantly greater than IDO-2 expression in the prefrontal cortex of wild type mice. IDO-1 expression was more pronounced in the mitochondria than in the cytosol. AMPH treatment activated intra-mitochondrial Ca2+ accumulation and mitochondrial oxidative burden, while inhibited mitochondrial membrane potential and activity of the mitochondrial complex (I > II), mitochondrial glutathione peroxidase, and superoxide dismutase, indicating that mitochondrial burden might be contributable to mania-like behaviors induced by AMPH. The behaviors were significantly attenuated by lithium, valproate, or IDO-1 knockout, but not in IDO-2 knockout mice. Lithium, valproate administration, or IDO-1 knockout significantly attenuated mitochondrial burden. Neither lithium nor valproate produced additive effects above the protective effects observed in IDO-1 KO in mice. Collectively, our results suggest that mood stabilizers attenuate AMPH-induced mania-like behaviors via attenuation of IDO-1-dependent mitochondrial stress, highlighting IDO-1 as a novel molecular target for the protective potential of mood stabilizers.
Collapse
Affiliation(s)
- Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan.
| | - The-Vinh Tran
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Dieu-Hien Phan
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Dae-Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, 24252, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jae Hoon Cheong
- Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan; Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
26
|
Coadministration of lithium and celecoxib reverses manic-like behavior and decreases oxidative stress in a dopaminergic model of mania induced in rats. Transl Psychiatry 2019; 9:297. [PMID: 31723123 PMCID: PMC6853972 DOI: 10.1038/s41398-019-0637-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 06/10/2019] [Accepted: 06/20/2019] [Indexed: 12/24/2022] Open
Abstract
The present study intends to investigate the effect of lithium (Li) and celecoxib (Cel) coadministration on the behavioral status and oxidative stress parameters in a rat model of mania induced by dextroamphetamine (d-AMPH). Male Wistar rats were treated with d-AMPH or saline (Sal) for 14 days; on the 8th day of treatment, rats received lithium (Li), celecoxib (Cel), Li plus Cel, or water until day 14. Levels of oxidative stress parameters were evaluated in the serum, frontal cortex, and hippocampus. d-AMPH administration induced hyperlocomotion in rats, which was significantly reversed by Li and Cel coadministration. In addition, d-AMPH administration induced damage to proteins and lipids in the frontal cortex and hippocampus of rats. All these impairments were reversed by treatment with Li and/or Cel, in a way dependent on cerebral area and biochemical analysis. Li and Cel coadministration reversed the d-AMPH-induced decrease in catalase activity in cerebral structures. The activity of glutathione peroxidase was decreased in the frontal cortex of animals receiving d-AMPH, and treatment with Li, Cel, or a combination thereof reversed this alteration in this structure. Overall, data indicate hyperlocomotion and alteration in oxidative stress biomarkers in the cerebral structures of rats receiving d-AMPH. Li and Cel coadministration can mitigate these modifications, comprising a potential novel approach for BD therapy.
Collapse
|
27
|
Azechi H, Hakamada K, Yamamoto T. A new inbred strain of Fawn-Hooded rats demonstrates mania-like behavioural and monoaminergic abnormalities. IBRO Rep 2019; 7:98-106. [PMID: 31763490 PMCID: PMC6861655 DOI: 10.1016/j.ibror.2019.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 11/02/2019] [Indexed: 01/06/2023] Open
Abstract
The Fawn-Hooded (FH) rat carries a gene mutation that results in a dysfunctional serotoninergic system. However, previous studies have reported differing features between the FH/Wjd and FH/Har strains. We aimed to compare the behavioural and neurobiological features of FH/HamSlc rats with those of Fischer 344 rats. We performed the open field, elevated minus-maze, Y-maze spontaneous alternation, and forced swim tests to investigate behavioural alterations. We also assessed neurobiological characteristics by quantifying monoamines and their related compounds in the prefrontal cortex, hippocampus, and striatum using high-performance liquid chromatography with an electrochemical detection system. FH/HamSlc rats showed hyperactivity and a high impulsivity tendency in the open field and the elevated minus maze test, but no cognitive dysfunction. In addition, the hyperactivity was suppressed immediately after the forced swim test. FH/HamSlc rats showed low dopamine levels, but high dopamine turnover in the striatum. Serotonin and noradrenaline levels were low in the prefrontal cortex and the hippocampus of FH/HamSlc rats, but high serotonin turnover was observed in the prefrontal cortex, hippocampus, and striatum. FH/HamSlc rats show (1) mania-like behavioural characteristics that are different from those of other strains of FH rats; (2) stimulus dependent suppression of hyperactivity similar to the clinical findings that exercise alleviates the symptoms of bipolar disorder; and (3) monoaminergic dysregulation such as monoamine imbalance and hyperturnover that may be associated with mania-related behavioural characteristics. Thus, the FH/HamSlc rat is a new animal model for mania including bipolar disorder.
Collapse
Key Words
- 5-HIAA, 5-hydroxyindoleacetic acid
- 5-HT, serotonin
- ADHD, attention-deficit hyperactivity disorder
- Bipolar mania model
- DA, dopamine
- DOPAC, 3,4-dihydroxyphenylacetic acid
- FH, Fawn-Hooded
- Fawn-Hooded rat
- HPLC, high-performance liquid chromatography
- HVA, homovanillic acid
- Hyperactivity
- Impulsivity
- MAO-A, monoamine oxidase A
- MHPG, 3-methoxy-4-hydroxyphenylglycol
- Monoaminergic dysregulation
- NA, noradrenaline
- PCA, perchloric acid
- SEM, standard error of the mean
- Stimulus responsivity
- TPH2, tryptophan hydroxylase 2
Collapse
Affiliation(s)
- Hirotsugu Azechi
- Department of Psychology, Tezukayama University, Nara 631-8585, Japan
| | - Kōsuke Hakamada
- Department of Neurophysiology and Cognitive Neuroscience, Graduate School of Psychological Sciences, Tezukayama University, Nara 631-8585, Japan
| | - Takanobu Yamamoto
- Department of Psychology, Tezukayama University, Nara 631-8585, Japan.,Department of Neurophysiology and Cognitive Neuroscience, Graduate School of Psychological Sciences, Tezukayama University, Nara 631-8585, Japan
| |
Collapse
|
28
|
Valvassori SS, Dal-Pont GC, Resende WR, Varela RB, Lopes-Borges J, Cararo JH, Quevedo J. Validation of the animal model of bipolar disorder induced by Ouabain: face, construct and predictive perspectives. Transl Psychiatry 2019; 9:158. [PMID: 31164628 PMCID: PMC6548776 DOI: 10.1038/s41398-019-0494-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
A particular challenge in the development of a bipolar disorder (BD) model in animals is the complicated clinical course of the condition, characterized by manic, depressive and mixed mood episodes. Ouabain (OUA) is an inhibitor of Na+/K+-ATPase enzyme. Intracerebroventricular (ICV) injection of this drug in rats has been regarded a proper model to study BD by mimic specific manic symptoms, which are reversed by lithium (Li), an important mood stabilizer drug. However, further validation of this experimental approach is required to characterize it as an animal model of BD, including depressive-like behaviors. The present study aimed to assess manic- and depressive-like behaviors, potential alteration in the hypothalamic-pituitary-adrenal (HPA) system and oxidative stress parameters after a single OUA ICV administration in adult male Wistar rats. Moreover, we evaluated Li effects in this experimental setting. Data show that OUA ICV administration could constitute a suitable model for BD since the injection of the drug triggered manic- and depressive-like behaviors in the same animal. Additionally, the OUA model mimics significant physiological and neurochemical alterations detected in BD patients, including an increase in oxidative stress and change in HPA axis. Our findings suggest that decreased Na+/K+-ATPase activity detected in bipolar patients may be linked to increased secretion of glucocorticoid hormones and oxidative damage, leading to the marked behavioral swings. The Li administration mitigated these pathological changes in the rats. The proposed OUA model is regarded as suitable to simulate BD by complying with all validities required to a proper animal model of the psychiatric disorder.
Collapse
Affiliation(s)
- Samira S. Valvassori
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil
| | - Gustavo C. Dal-Pont
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil
| | - Wilson R. Resende
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil
| | - Roger B. Varela
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil
| | - Jéssica Lopes-Borges
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil
| | - José Henrique Cararo
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil
| | - João Quevedo
- 0000 0001 2150 7271grid.412287.aTranslational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC Brazil ,0000 0000 9206 2401grid.267308.8Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX USA ,0000 0000 9206 2401grid.267308.8Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX USA ,0000 0001 2291 4776grid.240145.6Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX USA
| |
Collapse
|
29
|
Petreanu M, Maia P, da Rocha Pittarello JL, Loch LC, Monache FD, Perez AL, Solano-Arias G, Filho VC, de Souza MM, Niero R. Antidepressant-like effect and toxicological parameters of extract and withanolides isolated from aerial parts of Solanum capsicoides All. (Solanaceae). Naunyn Schmiedebergs Arch Pharmacol 2019; 392:979-990. [PMID: 30982080 DOI: 10.1007/s00210-019-01648-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/22/2019] [Indexed: 11/28/2022]
Abstract
The present work describes the evaluation of the antidepressant-like activity of the extract, fractions, and compounds obtained from the aerial parts of Solanum capsicoides. The methanolic extract (MESC) obtained by conventional maceration was partitioned with solvents of increasing polarities yielding the respective fractions of hexane (HE), dichloromethane (DCM), and ethyl acetate (EA). The dichloromethane and ethyl acetate fractions were submitted to chromatographic and spectroscopic techniques, leading to the isolation and identification of cilistadiol (1), astragalin (2), and cilistol A (3). In relation to the antidepressant activity, the extract was active against the forced swimming test (FST) at a concentration of 300 mg/kg an ED50 (deffective dose that reduces 50% of immobility time) of 120.3 (117.3-123.4) mg/kg. Similar values were observed when evaluated in the tail suspension test (TST). In addition, the results showed no influence on motor behavior when evaluated in the open field test (OFT). Based on the observed profile of the MESC, dichloromethane fraction presenting the best profile, in both FST and TST test. Likewise, the fraction also did not present motor impairment when evaluated by the OFT test. Considering that the dichloromethane fraction was more effective, the isolated compounds cilistadiol and cilistol A were evaluated in the same experimental models. In FST, both compounds had a significant antidepressant-like effect, with ED50 values of 0.22 (0.16-0.28) and 1.03 (0.89-1.18) μmol/kg, respectively. When evaluated in the TST, showed ED50 values of 0.30 (0.18-0.52) and 1.49 (1.27-1.73) μmol/kg, respectively. The isolated compounds also did not present significant differences in the motor behavior when evaluated on OFT test in comparison with the control group. No toxicological parameters were observed until the highest dose of MESC (2000 mg/kg), demonstrating safety in the use of this plant.
Collapse
Affiliation(s)
- Marcel Petreanu
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Paula Maia
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Janieire L da Rocha Pittarello
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Lucimara C Loch
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Franco Delle Monache
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Alice L Perez
- Centro de Investigación en Productos Naturales (CIPRONA), Universidad de Costa Rica (UCR), San José, Costa Rica
| | - Godofredo Solano-Arias
- Centro de Investigación en Productos Naturales (CIPRONA), Universidad de Costa Rica (UCR), San José, Costa Rica
| | - Valdir Cechinel Filho
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Márcia Maria de Souza
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil
| | - Rivaldo Niero
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas (NIQFAR), Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, Santa Catarina, 88303-901, Brazil.
| |
Collapse
|
30
|
The internal link of serum steroid hormones levels in insomnia, depression, and Alzheimer's disease rats: Is there an effective way to distinguish among these three diseases based on potential biomarkers? J Sep Sci 2019; 42:1833-1841. [DOI: 10.1002/jssc.201801298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/20/2019] [Accepted: 03/02/2019] [Indexed: 11/07/2022]
|
31
|
Stukalin Y, Einat H. Analyzing test batteries in animal models of psychopathology with multivariate analysis of variance (MANOVA): One possible approach to increase external validity. Pharmacol Biochem Behav 2019; 178:51-55. [DOI: 10.1016/j.pbb.2017.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/22/2017] [Accepted: 11/27/2017] [Indexed: 10/18/2022]
|
32
|
Demin KA, Sysoev M, Chernysh MV, Savva AK, Koshiba M, Wappler-Guzzetta EA, Song C, De Abreu MS, Leonard B, Parker MO, Harvey BH, Tian L, Vasar E, Strekalova T, Amstislavskaya TG, Volgin AD, Alpyshov ET, Wang D, Kalueff AV. Animal models of major depressive disorder and the implications for drug discovery and development. Expert Opin Drug Discov 2019; 14:365-378. [PMID: 30793996 DOI: 10.1080/17460441.2019.1575360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Depression is a highly debilitating psychiatric disorder that affects the global population and causes severe disabilities and suicide. Depression pathogenesis remains poorly understood, and the disorder is often treatment-resistant and recurrent, necessitating the development of novel therapies, models and concepts in this field. Areas covered: Animal models are indispensable for translational biological psychiatry, and markedly advance the study of depression. Novel approaches continuously emerge that may help untangle the disorder heterogeneity and unclear categories of disease classification systems. Some of these approaches include widening the spectrum of model species used for translational research, using a broader range of test paradigms, exploring new pathogenic pathways and biomarkers, and focusing more closely on processes beyond neural cells (e.g. glial, inflammatory and metabolic deficits). Expert opinion: Dividing the core symptoms into easily translatable, evolutionarily conserved phenotypes is an effective way to reevaluate current depression modeling. Conceptually novel approaches based on the endophenotype paradigm, cross-species trait genetics and 'domain interplay concept', as well as using a wider spectrum of model organisms and target systems will enhance experimental modeling of depression and antidepressant drug discovery.
Collapse
Affiliation(s)
- Konstantin A Demin
- a Institute of Experimental Medicine , Almazov National Medical Research Centre , St. Petersburg , Russia.,b Institute of Translational Biomedicine , St. Petersburg State University , St. Petersburg , Russia
| | - Maxim Sysoev
- c Laboratory of Preclinical Bioscreening , Russian Research Center for Radiology and Surgical Technologies , St. Petersburg , Russia.,d Institute of Experimental Medicine , St. Petersburg , Russia
| | - Maria V Chernysh
- b Institute of Translational Biomedicine , St. Petersburg State University , St. Petersburg , Russia
| | - Anna K Savva
- e Faculty of Biology , St. Petersburg State University , St. Petersburg , Russia
| | | | | | - Cai Song
- h Research Institute of Marine Drugs and Nutrition , Guangdong Ocean University , Zhanjiang , China.,i Marine Medicine Development Center, Shenzhen Institute , Guangdong Ocean University , Shenzhen , China
| | - Murilo S De Abreu
- j Bioscience Institute , University of Passo Fundo (UPF) , Passo Fundo , Brazil
| | | | - Matthew O Parker
- l Brain and Behaviour Lab , School of Pharmacy and Biomedical Science, University of Portsmouth , Portsmouth , UK
| | - Brian H Harvey
- m Center of Excellence for Pharmaceutical Sciences , Division of Pharmacology, School of Pharmacy, North-West University , Potchefstroom , South Africa
| | - Li Tian
- n Institute of Biomedicine and Translational Medicine , University of Tartu , Tartu , Estonia
| | - Eero Vasar
- n Institute of Biomedicine and Translational Medicine , University of Tartu , Tartu , Estonia
| | - Tatyana Strekalova
- o Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, and Department of Normal Physiology , Sechenov First Moscow State Medical University , Moscow , Russia.,p Laboratory of Cognitive Dysfunctions , Institute of General Pathology and Pathophysiology , Moscow , Russia.,q Department of Neuroscience , Maastricht University , Maastricht , The Netherlands
| | | | - Andrey D Volgin
- g The International Zebrafish Neuroscience Research Consortium (ZNRC) , Slidell , LA , USA.,r Scientific Research Institute of Physiology and Basic Medicine , Novosibirsk , Russia
| | - Erik T Alpyshov
- s School of Pharmacy , Southwest University , Chongqing , China
| | - Dongmei Wang
- s School of Pharmacy , Southwest University , Chongqing , China
| | - Allan V Kalueff
- s School of Pharmacy , Southwest University , Chongqing , China.,t Almazov National Medical Research Centre , St. Petersburg , Russia.,u Ural Federal University , Ekaterinburg , Russia.,v Granov Russian Research Center of Radiology and Surgical Technologies , St. Petersburg , Russia.,w Laboratory of Biological Psychiatry, Institute of Translational Biomedicine , St. Petersburg State University , St. Petersburg , Russia.,x Laboratory of Translational Biopsychiatry , Scientific Research Institute of Physiology and Basic Medicine , Novosibirsk , Russia.,y ZENEREI Institute , Slidell , LA , USA.,z The International Stress and Behavior Society (ISBS), US HQ , New Orleans , LA , USA
| |
Collapse
|
33
|
Lima CNDC, da Silva FER, Chaves Filho AJM, Queiroz AIDG, Okamura AMNC, Fries GR, Quevedo J, de Sousa FCF, Vasconcelos SMM, de Lucena DF, Fonteles MMDF, Macedo DS. High Exploratory Phenotype Rats Exposed to Environmental Stressors Present Memory Deficits Accompanied by Immune-Inflammatory/Oxidative Alterations: Relevance to the Relationship Between Temperament and Mood Disorders. Front Psychiatry 2019; 10:547. [PMID: 31428001 PMCID: PMC6689823 DOI: 10.3389/fpsyt.2019.00547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
Low-exploratory (LE) and high-exploratory (HE) rodents mimic human depressive and hyperthymic temperaments, respectively. Mood disorders (MD) may be developed by the exposure of these temperaments to environmental stress (ES). Psychiatric symptoms severity in MD patients is related to the magnitude of memory impairment. Thus, we aimed at studying the consequences of the exposure of LE and HE male Wistar rats, during periadolescence, to a combination of ES, namely, paradoxical sleep deprivation (PSD) and unpredictable stress (US), on anxiety-related behavior in the plus maze test, working (WM) and declarative memory (DM) performance. We also evaluated hippocampal immune-inflammatory/oxidative, as consequences of ES, and prevention of ES-induced alterations by the mood-stabilizing drugs, lithium and valproate. Medium exploratory (ME) control rats were used for comparisons with HE- and LE-control rats. We observed that HE-controls presented increased anxiolytic behavior that was significantly increased by ES exposure, whereas LE-controls presented increased anxiety-like behavior relative to ME-controls. Lithium and valproate prevented anxiolytic alterations in HE+ES rats. HE+ES- and LE+ES-rats presented WM and DM deficits. Valproate and lithium prevented WM deficits in LE-PSD+US rats. Lithium prevented DM impairment in HE+ES-rats. Hippocampal levels of reduced glutathione (GSH) increased four-fold in HE+ES-rats, being prevented by valproate and lithium. All groups of LE+ES-rats presented increased levels of GSH in relation to controls. Increments in lipid peroxidation in LE+ES- and HE+ES-rats were prevented by valproate in HE+ES-rats and by both drugs in LE+ES-rats. Nitrite levels were increased in HE+ES- and LE+ES-rats (five-fold increase), which was prevented by both drugs in LE+ES-rats. HE+ES-rats presented a two-fold increase in the inducible nitric oxide synthase (iNOS) expression that was prevented by lithium. HE+ES-rats showed increased hippocampal and plasma levels of interleukin (IL)-1β and IL-4. Indoleamine 2, 3-dioxygenase 1 (IDO1) was increased in HE+ES- and LE+ES-rats, while tryptophan 2,3-dioxygenase (TDO2) was increased only in HE+ES-rats. Altogether, our results showed that LE- and HE-rats exposed to ES present distinct anxiety-related behavior and similar memory deficits. Furthermore, HE+ES-rats presented more brain and plasma inflammatory alterations that were partially prevented by the mood-stabilizing drugs. These alterations in HE+ES-rats may possibly be related to the development of mood symptoms.
Collapse
Affiliation(s)
- Camila Nayane de Carvalho Lima
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil.,Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - Francisco Eliclécio Rodrigues da Silva
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Ana Isabelle de Gois Queiroz
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Adriana Mary Nunes Costa Okamura
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Gabriel Rodrigo Fries
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - Francisca Cléa F de Sousa
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Silvania Maria Mendes Vasconcelos
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - David F de Lucena
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Marta Maria de França Fonteles
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceara, Fortaleza, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Neurosciences and Behavior Department, Faculdade de Medicina de Ribeirão Preto (FMRP), Ribeirão Preto, Brazil
| |
Collapse
|
34
|
Gorlova A, Pavlov D, Zubkov E, Zorkina Y, Inozemtsev A, Morozova A, Chekhonin V. Alteration of oxidative stress markers and behavior of rats in a novel model of depression. Acta Neurobiol Exp (Wars) 2019. [DOI: 10.21307/ane-2019-021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
35
|
Miranda ASD, Miranda ASD, Teixeira AL. Lamotrigine as a mood stabilizer: insights from the pre-clinical evidence. Expert Opin Drug Discov 2018; 14:179-190. [PMID: 30523725 DOI: 10.1080/17460441.2019.1553951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Lamotrigine (LTG) is a well-established anticonvulsant that is also approved for the prevention of mood relapses in bipolar disorder. However, the mechanisms underlying LTG mood stabilizing effects remain unclear. Areas covered: Herein, the pre-clinical evidence concerning LTG's' mode of action in depression and mania is reviewed. Bottlenecks and future perspectives for this expanding and promising field are also discussed. Pre-clinical studies have indicated that neurotransmitter systems, especially serotoninergic, noradrenergic and glutamatergic, as well as non-neurotransmitter pathways such as inflammation and oxidative processes might play a role in LTG's antidepressant effects. The mechanisms underlying LTG's anti-manic properties remain to be fully explored, but the available pre-clinical evidence points out to the role of glutamatergic neurotransmission, possibly through AMPA-receptors. Expert opinion: A major limitation of current pre-clinical investigations is that there are no experimental models that recapitulate the complexity of bipolar disorder. Significant methodological differences concerning time and dose of LTG treatment, administration route, animal strains, and behavioral paradigms also hamper the reproducibility of the findings, leading to contradictory conclusions. Moreover, the role of other mechanisms (e.g. inositol phosphate and GSK3β pathways) implicated in the mode of action of different mood-stabilizers must also be consolidated with LTG.
Collapse
Affiliation(s)
- Aline Silva de Miranda
- a Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,b Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Brasil
| | - Amanda Silva de Miranda
- c Departamento de Química , Instituto de Ciências Exatas, Universidade Federal de Minas Gerais , Belo Horizonte , Brasil
| | - Antônio Lúcio Teixeira
- a Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,d Neuropsychiatry Program & Immuno-Psychiatry Lab, Department of Psychiatry & Behavioral Sciences, McGovern Medical School , University of Texas Health Science Center at Houston , Houston , USA
| |
Collapse
|
36
|
Dal-Pont GC, Resende WR, Varela RB, Menegas S, Trajano KS, Peterle BR, Quevedo J, Valvassori SS. Inhibition of GSK-3β on Behavioral Changes and Oxidative Stress in an Animal Model of Mania. Mol Neurobiol 2018; 56:2379-2393. [DOI: 10.1007/s12035-018-1226-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/05/2018] [Indexed: 12/27/2022]
|
37
|
Cellular and behavioral effects of lipopolysaccharide treatment are dependent upon neurokinin-1 receptor activation. J Neuroinflammation 2018; 15:60. [PMID: 29486768 PMCID: PMC6389133 DOI: 10.1186/s12974-018-1098-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/19/2018] [Indexed: 11/10/2022] Open
Abstract
Background Several psychiatric conditions are affected by neuroinflammation and neuroimmune activation. The transcription factor nuclear factor kappa light-chain-enhancer of activated B cells (NFkB) plays a major role in inflammation and innate immunity. The neurokinin-1 receptor (NK1R) is the primary endogenous target of the neuroactive peptide substance P, and some data suggests that NK1R stimulation may influence NFkB activity. Both NK1R and NFkB have been shown to play a functional role in complex behaviors including stress responsivity, depression, and addiction. In this study, we test whether NFkB activity in the brain (stimulated by lipopolysaccharide administration) is dependent upon the NK1R. Methods Adult male Wistar rats were treated systemically with the NK1R antagonist L822429 followed by administration of systemic lipopolysaccharide (LPS, a strong activator of NFkB). Hippocampal extracts were used to assess expression of proinflammatory cytokines and NFkB-DNA-binding potential. For behavioral studies, rats were trained to consume 1% (w/v) sucrose solution in a continuous access two-bottle choice model. After establishment of baseline, animals were treated with L822429 and LPS and sucrose preference was measured 12 h post-treatment. Results Systemic LPS treatment causes a significant increase in proinflammatory cytokine expression and NFkB-DNA-binding activity within the hippocampus. These increases are attenuated by systemic pretreatment with the NK1R antagonist L822429. Systemic LPS treatment also led to the development of anhedonic-like behavior, evidenced by decreased sucrose intake in the sucrose preference test. This behavior was significantly attenuated by systemic pretreatment with the NK1R antagonist L822429. Conclusions Systemic LPS treatment induced significant increases in NFkB activity, evidenced by increased NFkB-DNA binding and by increased proinflammatory cytokine expression in the hippocampus. LPS also induced anhedonic-like behavior. Both the molecular and behavioral effects of LPS treatment were significantly attenuated by systemic NK1R antagonism, suggesting that NK1R stimulation lies upstream of NFkB activation following systemic LPS administration and is at least in part responsible for NFkB activation.
Collapse
|
38
|
Molina-Jiménez T, Limón-Morales O, Bonilla-Jaime H. Early postnatal treatment with clomipramine induces female sexual behavior and estrous cycle impairment. Pharmacol Biochem Behav 2018; 166:27-34. [PMID: 29407872 DOI: 10.1016/j.pbb.2018.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 01/10/2023]
Abstract
Administration of clomipramine (CMI), a tricyclic antidepressant, in early stages of development in rats, is considered an animal model for the study of depression. This pharmacological manipulation has induced behavioral and physiological alterations, i.e., less pleasure-seeking behaviors, despair, hyperactivity, cognitive dysfunction, alterations in neurotransmitter systems and in HPA axis. These abnormalities in adult male rats are similar to the symptoms observed in major depressive disorders. One of the main pleasure-seeking behaviors affected in male rats treated with CMI is sexual behavior. However, to date, no effects of early postnatal CMI treatment have been reported on female reproductive cyclicity and sexual behavior. Therefore, we explored CMI administration in early life (8-21 PN) on the estrous cycle and sexual behavior of adult female rats. Compared to the rats in the early postnatal saline treatment (CTRL group), the CMI rats had fewer estrous cycles, fewer days in the estrous stage, and longer cycles during a 20-day period of vaginal cytology analysis. On the behavioral test, the CMI rats displayed fewer proceptive behaviors (hopping, darting) and had lower lordosis quotients. Also, they usually failed to display lordosis and only rarely manifested marginal or normal lordosis. In contrast, the CTRL rats tended to display normal lordosis. These results suggest that early postnatal CMI treatment caused long-term disruptions of the estrous cycle and female sexual behavior, perhaps by alteration in the hypothalamic-pituitary-gonadal (HPG) axes and in neuronal circuits involved in the regulation of the performance and motivational of sexual behavior as the noradrenergic and serotonergic systems.
Collapse
Affiliation(s)
- Tania Molina-Jiménez
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Apartado, Postal 55 535, C.P. 09340 Ciudad de México, Mexico
| | - Ofelia Limón-Morales
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Av Universidad 3000, Cd. Universitaria, Coyoacán, 04510 Ciudad de México, Mexico
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Apartado Postal 55 535, C.P. 09340 Ciudad de México, Mexico.
| |
Collapse
|
39
|
Hodes A, Lifschytz T, Rosen H, Cohen Ben-Ami H, Lichtstein D. Reduction in endogenous cardiac steroids protects the brain from oxidative stress in a mouse model of mania induced by amphetamine. Brain Res Bull 2018; 137:356-362. [PMID: 29374602 DOI: 10.1016/j.brainresbull.2018.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Bipolar disorder (BD) is a severe mental illness characterized by episodes of mania and depression. Numerous studies have implicated the involvement of endogenous cardiac steroids (CS), and their receptor, Na+, K+ -ATPase, in BD. The aim of the present study was to examine the role of brain oxidative stress in the CS-induced behavioral effects in mice. METHODS Amphetamine (AMPH)-induced hyperactivity, assessed in the open-field test, served as a model for manic-like behavior in mice. A reduction in brain CS was obtained by specific and sensitive anti-ouabain antibodies. The level of oxidative stress was tested in the hippocampus and frontal cortex by measuring the activity of antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), as well as the levels of antioxidant non-protein thiols (NPSH) and oxidative damage biomarkers thiobarbituric acid reactive substances (TBARS) and protein carbonyl (PC). RESULTS AMPH administration resulted in a marked hyperactivity and increased oxidative stress, as manifested by increased SOD activity, decreased activities of CAT and GPx, reduced levels of NPSH and increased levels of TBARS and PC. The administration of anti-ouabain antibodies, which reduced the AMPH-induced hyperactivity, protected against the concomitant oxidative stress in the brain. CONCLUSIONS Our results demonstrate that oxidative stress participates in the effects of endogenous CS on manic-like behavior induced by AMPH. These finding support the notion that CS and oxidative stress may be associated with the pathophysiology of mania and BD.
Collapse
Affiliation(s)
- Anastasia Hodes
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tzuri Lifschytz
- Department of Psychiatry, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haim Rosen
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Hagit Cohen Ben-Ami
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - David Lichtstein
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
40
|
Valvassori SS, Dal-Pont GC, Resende WR, Varela RB, Peterle BR, Gava FF, Mina FG, Cararo JH, Carvalho AF, Quevedo J. Lithium and Tamoxifen Modulate Behavior and Protein Kinase C Activity in the Animal Model of Mania Induced by Ouabain. Int J Neuropsychopharmacol 2017; 20:877-885. [PMID: 29020306 PMCID: PMC5737643 DOI: 10.1093/ijnp/pyx049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/04/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The intracerebroventricular injection of ouabain, a specific inhibitor of the Na+/K+-adenosine-triphosphatase (Na+/K+-ATPase) enzyme, induces hyperactivity in rats in a putative animal model of mania. Several evidences have suggested that the protein kinase C signaling pathway is involved in bipolar disorder. In addition, it is known that protein kinase C inhibitors, such as lithium and tamoxifen, are effective in treating acute mania. METHODS In the present study, we investigated the effects of lithium and tamoxifen on the protein kinase C signaling pathway in the frontal cortex and hippocampus of rats submitted to the animal model of mania induced by ouabain. We showed that ouabain induced hyperlocomotion in the rats. RESULTS Ouabain increased the protein kinase C activity and the protein kinase C and MARCKS phosphorylation in frontal cortex and hippocampus of rats. Lithium and tamoxifen reversed the behavioral and protein kinase C pathway changes induced by ouabain. These findings indicate that the Na+/K+-ATPase inhibition can lead to protein kinase C alteration. CONCLUSIONS The present study showed that lithium and tamoxifen modulate changes in the behavior and protein kinase C signalling pathway alterations induced by ouabain, underlining the need for more studies of protein kinase C as a possible target for treatment of bipolar disorder.
Collapse
Affiliation(s)
- Samira S Valvassori
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo).,Correspondence: S. S. Valvassori, PhD, Laboratório de Sinalização Neural e Psicofarmacologia, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brasil, CEP 88806-000 ()
| | - Gustavo C Dal-Pont
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - Wilson R Resende
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - Roger B Varela
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - Bruna R Peterle
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - Fernanda F Gava
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - Francielle G Mina
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - José H Cararo
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - André F Carvalho
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| | - João Quevedo
- Laboratory of Neuronal Signaling and Psychopharmacology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, and Ms Gava); Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Valvassori, Mr Dal-Pont, Dr Resende, Mr Varela, Ms Peterle, Ms Gava, Ms Mina, and Dr Quevedo); Laboratory of Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil (Dr Cararo); Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil(Dr Carvalho); Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo);Center of Excellence on Mood Disorders, Department of Psychiatry and BehavioralSciences, The University of Texas Health Science Center at Houston Medical School, Houston, Texas (Dr Quevedo); Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas (Dr Quevedo)
| |
Collapse
|
41
|
Molina-Jímenez T, Landa-Cadena L, Bonilla-Jaime H. Chronic treatment with estradiol restores depressive-like behaviors in female Wistar rats treated neonatally with clomipramine. Horm Behav 2017; 94:61-68. [PMID: 28606740 DOI: 10.1016/j.yhbeh.2017.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 11/30/2022]
Abstract
Neonatal administration of clomipramine (CMI) induces diverse behavioral and neurochemical alterations in adult male rats that resemble major depression disorder. However, the possible behavioral alterations in adult female rats subjected to neonatal treatment with clomipramine are unknown. Therefore, the aim of this study was to explore the effect of neonatal treatment with CMI on adult female rats in relation to locomotion and behavioral despair during the estrus cycle. Also evaluated was the effect of chronic treatment with E2 on these female CMI rats. We found no effects on spontaneous locomotor activity due to neonatal treatment with CMI, or after 21days of E2 administration. In the FST, neonatal treatment with CMI increased immobility and decreased active swimming and climbing behaviors. Influence of the ovarian cycle was detected only in relation to climbing behavior, as the rats in the MD phase displayed less climbing activity. Chronic E2 administration decreased immobility but increased only swimming in CMI rats. These results suggest that neonatal treatment with CMI induces despair-like behavior in female rats, but that chronic E2 administration generates antidepressant-like behavior by decreasing immobility and increasing swimming, perhaps through interaction with the serotonergic system.
Collapse
Affiliation(s)
- Tania Molina-Jímenez
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Apartado Postal 55 535, C.P. 09340 Ciudad de México, México
| | - Liliana Landa-Cadena
- Facultad de Química Farmacéutica Biológica, Universidad Veracruzana, Circuito Gonzalo Aguirre Beltrán s/n, Zona Universitaria Xalapa, Veracruz, México
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Apartado Postal 55 535, C.P. 09340 Ciudad de México, México.
| |
Collapse
|
42
|
Valvassori SS, Dal-Pont GC, Resende WR, Jornada LK, Peterle BR, Machado AG, Farias HR, de Souza CT, Carvalho AF, Quevedo J. Lithium and valproate act on the GSK-3β signaling pathway to reverse manic-like behavior in an animal model of mania induced by ouabain. Neuropharmacology 2017; 117:447-459. [DOI: 10.1016/j.neuropharm.2016.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/04/2016] [Accepted: 10/13/2016] [Indexed: 11/28/2022]
|
43
|
Miranda ASD, Moreira FA, Teixeira AL. The preclinical discovery and development of quetiapine for the treatment of mania and depression. Expert Opin Drug Discov 2017; 12:525-535. [PMID: 28271741 DOI: 10.1080/17460441.2017.1304378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Bipolar disorder is a chronic disabling condition characterized by alternating manic and depressive episodes. Bipolar disorder has been associated with functional impairment, poor quality of life, morbidity and mortality. Despite its significant clinical, social and economic burden, treatment options for bipolar disorder are still limited. Several clinical trials have shown efficacy of the atypical antipsychotic quetiapine (QTP) in the treatment of this condition. However, the mechanisms underlying the antidepressant and anti-manic effects of QTP remain poorly understood. Areas covered: The article provides the emerging evidence from pre-clinical studies regarding the antidepressant and anti-manic mechanisms of action of QTP. In combination with its primary active metabolite norquetiapine, QTP modulates several neurotransmitter systems, including serotonin, dopamine, noradrenaline and histamine. QTP also seems to influence mediators of the immune system. Expert opinion: Pre-clinical studies have provided valuable information on the potential antidepressant mechanisms of action of QTP, but pre-clinical studies on QTP's anti-manic effects are still scarce. A major problem refers to the lack of valid experimental models for bipolar disorder. Additionally, immune and genetic based studies are largely descriptive. The role of the QTP metabolite norquetiapine in modulating non-neurotransmitter systems also needs to be further addressed.
Collapse
Affiliation(s)
- Aline Silva de Miranda
- a Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,b Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Brasil
| | - Fabrício A Moreira
- c Laboratório de Neuropsicofarmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Brasil
| | - Antônio Lúcio Teixeira
- a Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,d Neuropsychiatry Program, Department of Psychiatry & Behavioral Sciences, McGovern Medical School , University of Texas Health Science Center at Houston , Houston , TX , USA
| |
Collapse
|
44
|
Sharma AN, Fries GR, Galvez JF, Valvassori SS, Soares JC, Carvalho AF, Quevedo J. Modeling mania in preclinical settings: A comprehensive review. Prog Neuropsychopharmacol Biol Psychiatry 2016; 66:22-34. [PMID: 26545487 PMCID: PMC4728043 DOI: 10.1016/j.pnpbp.2015.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/29/2015] [Accepted: 11/03/2015] [Indexed: 12/17/2022]
Abstract
The current pathophysiological understanding of mechanisms leading to onset and progression of bipolar manic episodes remains limited. At the same time, available animal models for mania have limited face, construct, and predictive validities. Additionally, these models fail to encompass recent pathophysiological frameworks of bipolar disorder (BD), e.g. neuroprogression. Therefore, there is a need to search for novel preclinical models for mania that could comprehensively address these limitations. Herein we review the history, validity, and caveats of currently available animal models for mania. We also review new genetic models for mania, namely knockout mice for genes involved in neurotransmission, synapse formation, and intracellular signaling pathways. Furthermore, we review recent trends in preclinical models for mania that may aid in the comprehension of mechanisms underlying the neuroprogressive and recurring nature of BD. In conclusion, the validity of animal models for mania remains limited. Nevertheless, novel (e.g. genetic) animal models as well as adaptation of existing paradigms hold promise.
Collapse
Affiliation(s)
- Ajaykumar N Sharma
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Gabriel R Fries
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Juan F Galvez
- Department of Psychiatry, Pontificia Universidad Javeriana School of Medicine, Bogotá, Colombia
| | - Samira S Valvassori
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Jair C Soares
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Joao Quevedo
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil.
| |
Collapse
|
45
|
Lee SA, Holly KS, Voziyanov V, Villalba SL, Tong R, Grigsby HE, Glasscock E, Szele FG, Vlachos I, Murray TA. Gradient Index Microlens Implanted in Prefrontal Cortex of Mouse Does Not Affect Behavioral Test Performance over Time. PLoS One 2016; 11:e0146533. [PMID: 26799938 PMCID: PMC4723314 DOI: 10.1371/journal.pone.0146533] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 12/19/2015] [Indexed: 12/11/2022] Open
Abstract
Implanted gradient index lenses have extended the reach of standard multiphoton microscopy from the upper layers of the mouse cortex to the lower cortical layers and even subcortical regions. These lenses have the clarity to visualize dynamic activities, such as calcium transients, with subcellular and millisecond resolution and the stability to facilitate repeated imaging over weeks and months. In addition, behavioral tests can be used to correlate performance with observed changes in network function and structure that occur over time. Yet, this raises the questions, does an implanted microlens have an effect on behavioral tests, and if so, what is the extent of the effect? To answer these questions, we compared the performance of three groups of mice in three common behavioral tests. A gradient index lens was implanted in the prefrontal cortex of experimental mice. We compared their performance with mice that had either a cranial window or a sham surgery. Three presurgical and five postsurgical sets of behavioral tests were performed over seven weeks. Behavioral tests included rotarod, foot fault, and Morris water maze. No significant differences were found between the three groups, suggesting that microlens implantation did not affect performance. The results for the current study clear the way for combining behavioral studies with gradient index lens imaging in the prefrontal cortex, and potentially other regions of the mouse brain, to study structural, functional, and behavioral relationships in the brain.
Collapse
Affiliation(s)
- Seon A. Lee
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Kevin S. Holly
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Vladislav Voziyanov
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Stephanie L. Villalba
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Rudi Tong
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Holly E. Grigsby
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Francis G. Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ioannis Vlachos
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| | - Teresa A. Murray
- Center for Biomedical Research and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, United States of America
| |
Collapse
|
46
|
Fries GR, Valvassori SS, Bock H, Stertz L, Magalhães PVDS, Mariot E, Varela RB, Kauer-Sant'Anna M, Quevedo J, Kapczinski F, Saraiva-Pereira ML. Memory and brain-derived neurotrophic factor after subchronic or chronic amphetamine treatment in an animal model of mania. J Psychiatr Res 2015; 68:329-36. [PMID: 26026487 DOI: 10.1016/j.jpsychires.2015.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 05/01/2015] [Accepted: 05/07/2015] [Indexed: 11/26/2022]
Abstract
Progression of bipolar disorder (BD) has been associated with cognitive impairment and changes in neuroplasticity, including a decrease in serum brain-derived neurotrophic factor (BDNF). However, no study could examine BDNF levels directly in different brain regions after repeated mood episodes to date. The proposed animal model was designed to mimic several manic episodes and evaluate whether the performance in memory tasks and BDNF levels in hippocampus, prefrontal cortex, and amygdala would change after repeated amphetamine (AMPH) exposure. Adult male Wistar rats were divided into subchronic (AMPH for 7 days) and chronic groups (35 days), mimicking manic episodes at early and late stages of BD, respectively. After open field habituation or inhibitory avoidance test, rats were killed, brain regions were isolated, and BDNF mRNA and protein levels were measured by quantitative real-time PCR and ELISA, respectively. AMPH impaired habituation memory in both subchronic and chronic groups, and the impairment was worse in the chronic group. This was accompanied by increased Bdnf mRNA levels in the prefrontal cortex and amygdala region, as well as reduced BDNF protein in the hippocampus. In the inhibitory avoidance, AMPH significantly decreased the change from training to test when compared to saline. No difference was observed between subchronic and chronic groups, although chronically AMPH-treated rats presented increased Bdnf mRNA levels and decreased protein levels in hippocampus when compared to the subchronic group. Our results suggest that the cognitive impairment related to BD neuroprogression may be associated with BDNF alterations in hippocampus, prefrontal cortex, and amygdala.
Collapse
Affiliation(s)
- Gabriel R Fries
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Laboratory of Genetic Identification and Medical Genetic Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Samira S Valvassori
- INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Laboratory of Neurosciences, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, 88806-000, Criciúma, Santa Catarina, Brazil.
| | - Hugo Bock
- Laboratory of Genetic Identification and Medical Genetic Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-903, Rio Grande do Sul, Brazil.
| | - Laura Stertz
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Pedro Vieira da Silva Magalhães
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Department of Psychiatry, Universidade Federal Rio Grande do Sul, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil.
| | - Edimilson Mariot
- INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Laboratory of Neurosciences, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, 88806-000, Criciúma, Santa Catarina, Brazil.
| | - Roger B Varela
- INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Laboratory of Neurosciences, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, 88806-000, Criciúma, Santa Catarina, Brazil.
| | - Marcia Kauer-Sant'Anna
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-903, Rio Grande do Sul, Brazil; Department of Psychiatry, Universidade Federal Rio Grande do Sul, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil.
| | - João Quevedo
- INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA; Laboratory of Neurosciences, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, 88806-000, Criciúma, Santa Catarina, Brazil.
| | - Flávio Kapczinski
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; INCT of Translational Medicine, Hospital de Clinicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Psychiatry, Universidade Federal Rio Grande do Sul, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil.
| | - Maria Luiza Saraiva-Pereira
- Laboratory of Genetic Identification and Medical Genetic Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-903, Rio Grande do Sul, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-903, Rio Grande do Sul, Brazil.
| |
Collapse
|
47
|
Valvassori SS, Tonin PT, Varela RB, Carvalho AF, Mariot E, Amboni RT, Bianchini G, Andersen ML, Quevedo J. Lithium modulates the production of peripheral and cerebral cytokines in an animal model of mania induced by dextroamphetamine. Bipolar Disord 2015; 17:507-17. [PMID: 25929806 DOI: 10.1111/bdi.12299] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/09/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Several recent studies have suggested that the physiopathology of bipolar disorder (BD) is related to immune system alterations and inflammation. Lithium (Li) is a mood stabilizer that is considered the first-line treatment for this mood disorder. The goal of the present study was to investigate the effects of Li administration on behavior and cytokine levels [interleukin (IL)-1β, IL-4, IL-6, IL-10, and tumor necrosis factor-alpha (TNF-α)] in the periphery and brains of rats subjected to an animal model of mania induced by amphetamine (d-AMPH). METHODS Male Wistar rats were treated with d-AMPH or saline (Sal) for 14 days; on Day 8 of treatment, the rats were administered Li or Sal for the final seven days. Cytokine (IL-1β, IL-4, IL-6, IL-10, and TNF-α) levels were evaluated in the cerebrospinal fluid (CSF), serum, frontal cortex, striatum, and hippocampus. RESULTS The present study showed that d-AMPH induced hyperactivity in rats (p < 0.001), and Li treatment reversed this behavioral alteration (p < 0.001). In addition, d-AMPH increased the levels of IL-4, IL-6, IL-10, and TNF-α in the frontal cortex (p < 0.001), striatum (p < 0.001), and serum (p < 0.001), and treatment with Li reversed these cytokine alterations (p < 0.001). CONCLUSIONS Li modulates peripheral and cerebral cytokine production in an animal model of mania induced by d-AMPH, suggesting that its action on the inflammatory system may contribute to its therapeutic efficacy.
Collapse
Affiliation(s)
- Samira S Valvassori
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Paula T Tonin
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Roger B Varela
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - André F Carvalho
- Department of Psychiatry and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Edemilson Mariot
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Rafaela T Amboni
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Guilherme Bianchini
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Monica L Andersen
- Departamento de Psicologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - João Quevedo
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Center for Experimental Models in Psychiatry, Department of Psychiatry and Behavioral Sciences, University of Texas Medical School at Houston, Houston, TX, USA
| |
Collapse
|
48
|
Su M, Hong J, Zhao Y, Liu S, Xue X. MeCP2 controls hippocampal brain-derived neurotrophic factor expression via homeostatic interactions with microRNA‑132 in rats with depression. Mol Med Rep 2015; 12:5399-406. [PMID: 26239616 DOI: 10.3892/mmr.2015.4104] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 06/03/2015] [Indexed: 11/06/2022] Open
Abstract
Major depressive disorder (MDD) is a considerable public health concern, which affects patients worldwide. MDD is associated with psychosocial impairment, poor quality of life, and significant disability, morbidity and mortality. Stress is a major factor in depression, which impairs the structural and functional plasticity of the hippocampus. Previous studies have demonstrated that chronic unpredictable mild stress is able to downregulate the expression of brain‑derived neurotrophic factor (BDNF) and methyl‑CpG‑binding protein 2 (MeCP2), and alter the expression levels of certain microRNAs (miR). The aim of the present study was to investigate the regulatory association between BDNF, MeCP2 and miR-132 in an animal model of chronic stress‑induced depression. ELISA, western blot and qPCR were used to detect the expression levels of BDNF, MeCP2 and miR-132 in the peripheral blood samples of patients with MDD and in the hippocampi of depressed animals. In addition, a dual luciferase reporter gene system was used to determine whether miR-132 directly targets BDNF or MeCP2. The present study demonstrated that, as compared with normal subjects, miR‑132 expression was increased in the peripheral blood samples of patients with MDD, whereas the expression of MeCP2 and BDNF was decreased; thus, the expression levels of MeCP2 and BDNF were negatively correlated with those of miR‑132. In addition, in an animal model of chronic stress‑induced depression, increased expression levels of miR‑132, and decreased levels of MeCP2 and BDNF were detected in the hippocampi. Furthermore, knockdown of MeCP2 expression in primary hippocampal neurons increased the expression of miR‑132 and decreased the expression levels of BDNF. The results of the present study demonstrated that miR‑132 may directly target MeCP2, but not BDNF, and control its expression at the transcriptional and translational level. miR‑132 was also shown to negatively regulate BDNF expression. The reduced expression levels of BDNF, as induced by MeCP2 knockdown, were enhanced by miR‑132 mimics, and were rescued by miR‑132 inhibitors. These results suggested that homeostatic interactions between MeCP2 and miR‑132 may regulate hippocampal BDNF levels, which may have a role in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Meilei Su
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jun Hong
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yongzhi Zhao
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shuai Liu
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiang Xue
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
49
|
Luo YW, Xu Y, Cao WY, Zhong XL, Duan J, Wang XQ, Hu ZL, Li F, Zhang JY, Zhou M, Dai RP, Li CQ. Insulin-like growth factor 2 mitigates depressive behavior in a rat model of chronic stress. Neuropharmacology 2015; 89:318-24. [PMID: 25446675 DOI: 10.1016/j.neuropharm.2014.10.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 10/04/2014] [Accepted: 10/08/2014] [Indexed: 12/27/2022]
Abstract
Depression is a common psychiatric disorder associated with chronic stress. Insulin-like growth factor 2 (IGF2) is a growth factor that serves important roles in the brain during development and at adulthood. Here, the role of IGF2 expression in the hippocampus was investigated in a rat model of depression. A chronic restraint stress (CRS) model of depression was established in rats, exhibiting depression-like behavior as assessed with the sucrose preference test (SPT) and forced swimming test (FST), and with evaluation of the corticosterone levels. Hippocampal IGF2 levels were significantly lower in rats suffering CRS than in controls, as were levels of pERK1/2 and GluR1. Lentivirus-mediated hippocampal IGF2 overexpression alleviated depressive behavior in restrained rats, elevated the levels of pERK1/2 and GluR1 proteins, but it did not affect the expression of pGSK3β, GluR2, NMDAR1, and NMDAR2A. These results suggest the chronic restraint stress induces depressive behavior, which may be mediated by ERK-dependent IGF2 signaling, pointing to an antidepressant role for this molecular pathway.
Collapse
Affiliation(s)
- Yan-Wei Luo
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, 410013, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pathak G, Ibrahim BA, McCarthy SA, Baker K, Kelly MP. Amphetamine sensitization in mice is sufficient to produce both manic- and depressive-related behaviors as well as changes in the functional connectivity of corticolimbic structures. Neuropharmacology 2015; 95:434-47. [PMID: 25959066 DOI: 10.1016/j.neuropharm.2015.04.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 04/21/2015] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
Abstract
It has been suggested that amphetamine abuse and withdrawal mimics the diverse nature of bipolar disorder symptomatology in humans. Here, we determined if a single paradigm of amphetamine sensitization would be sufficient to produce both manic- and depressive-related behaviors in mice. CD-1 mice were subcutaneously dosed for 5 days with 1.8 mg/kg d-amphetamine or vehicle. On days 6-31 of withdrawal, amphetamine-sensitized (AS) mice were compared to vehicle-treated (VT) mice on a range of behavioral and biochemical endpoints. AS mice demonstrated reliable mania- and depression-related behaviors from day 7 to day 28 of withdrawal. Relative to VT mice, AS mice exhibited long-lasting mania-like hyperactivity following either an acute 30-min restraint stress or a low-dose 1 mg/kg d-amphetamine challenge, which was attenuated by the mood-stabilizers lithium and quetiapine. In absence of any challenge, AS mice showed anhedonia-like decreases in sucrose preference and depression-like impairments in the off-line consolidation of motor memory, as reflected by the lack of spontaneous improvement across days of training on the rotarod. AS mice also demonstrated a functional impairment in nest building, an ethologically-relevant activity of daily living. Western blot analyses revealed a significant increase in methylation of histone 3 at lysine 9 (H3K9), but not lysine 4 (H3K4), in hippocampus of AS mice relative to VT mice. In situ hybridization for the immediate-early gene activity-regulated cytoskeleton-associated protein (Arc) further revealed heightened activation of corticolimbic structures, decreased functional connectivity between frontal cortex and striatum, and increased functional connectivity between the amygdala and hippocampus of AS mice. The effects of amphetamine sensitization were blunted in C57BL/6J mice relative to CD-1 mice. These results show that a single amphetamine sensitization protocol is sufficient to produce behavioral, functional, and biochemical phenotypes in mice that are relevant to bipolar disorder.
Collapse
Affiliation(s)
- G Pathak
- University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - B A Ibrahim
- University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | | | - K Baker
- Pfizer, Neuroscience, Groton, CT 06340, USA
| | - M P Kelly
- University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|