1
|
Reguilón MD, Ferrer-Pérez C, Manzanedo C, Miñarro J, Rodríguez-Arias M. Voluntary wheel running during adolescence prevents the increase in ethanol intake induced by social defeat in male mice. Psychopharmacology (Berl) 2023:10.1007/s00213-023-06461-0. [PMID: 37736785 DOI: 10.1007/s00213-023-06461-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023]
Abstract
RATIONALE Exposure to social defeat (SD) induces a depressive phenotype, increased ethanol seeking and consumption, accompanied by activation of the neuroinflammatory response. However, a resilient response can be potentiated through physical exercise in the form of voluntary wheel running (VWR) during or after exposure to social stress. Therefore, the aim of this study was to test whether physical exercise during adolescence prior to being exposed to SD can enhance resilience to the increase in ethanol intake. METHODS Male mice had access to VWR during adolescence and the effects of social defeat (4 sessions every 72 h) on oral ethanol self-administration (SA) was evaluated. Based on the social interaction test, mice were classified as resilient or susceptible to depressive-like behavior. Two weeks after the last encounter, mice were subjected to the drinking in the dark and oral ethanol SA paradigms. Mice were then sacrificed to measure brain-derived neurotrophic factor (BDNF) levels in the striatum and hippocampus. RESULTS As expected, susceptible mice increased ethanol intake in the oral SA protocol. However, susceptible mice in the exercise condition did not increase ethanol intake, showing similar consumption and motivation for ethanol than the control and resilient groups. On the other hand, decreased BDNF levels were observed in susceptible mice in both experimental conditions compared to the control groups after ethanol SA. CONCLUSIONS The pre-exposure of VWR prevented the increase in consumption and motivation for ethanol induced by SD in susceptible mice. On the other hand, it appears that VWR did not exhibit any significant long-term effects on BDNF signaling, which is mainly affected in susceptible mice after ethanol intake.
Collapse
Affiliation(s)
- Marina D Reguilón
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - Carmen Ferrer-Pérez
- Departmento de Psicología Evolutiva, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Carmen Manzanedo
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - José Miñarro
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - Marta Rodríguez-Arias
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain.
| |
Collapse
|
2
|
Petković A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci 2022; 16:931964. [PMID: 36004305 PMCID: PMC9395206 DOI: 10.3389/fnbeh.2022.931964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Animal studies over the past two decades have led to extensive advances in our understanding of pathogenesis of depressive and mood disorders. Among these, rodent behavioural models proved to be of highest informative value. Here, we present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates. Behavioural stress paradigms and behavioural tests are assessed in terms of outcomes, strengths, weaknesses, and translational value, especially in the domain of pharmacological studies.
Collapse
Affiliation(s)
| | - Dipesh Chaudhury
- Laboratory of Neural Systems and Behaviour, Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
3
|
Parrott JM, Porter GA, Redus L, O'Connor JC. Brain derived neurotrophic factor deficiency exacerbates inflammation-induced anhedonia in mice. Psychoneuroendocrinology 2021; 134:105404. [PMID: 34601342 PMCID: PMC8934305 DOI: 10.1016/j.psyneuen.2021.105404] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 08/11/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is implicated in the pathology of major depression and influences the inflammatory response. Prolonged immune system activation can cause depression symptoms, and individuals with low BDNF expression may be vulnerable to inflammation-induced depression. We tested the hypothesis that BDNF deficient mice are vulnerable to the induction of depressive-like behavior following peripheral immune challenge. BDNF heterozygous (BDNF+/-) or wild-type (BDNF+/+) littermate mice were injected intraperitoneally (i.p.) with endotoxin (lipopolysaccharide, LPS) to trigger an acute pro-inflammatory response. After resolution of the acute sickness response, central expression of inflammatory genes, kynurenine metabolites, and depressive-like behaviors across multiple dimensions (symptoms) were measured. BDNF+/- mice displayed an exaggerated neuroinflammatory response following peripheral immune challenge. Pro-inflammatory cytokines interleukin-1β (IL-1β), tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) were overexpressed in BDNF+/- mice relative to BDNF+/+ littermate control mice. While behavioral despair and anxiety-like behavior was not different between genotypes, LPS-induced anhedonia-like behavior was significantly more pronounced in BDNF+/- mice relative to BDNF+/+ mice. The kynurenine pathway mediates the many depressive-like behavioral effects of peripheral LPS, and similar to pro-inflammatory cytokine gene expression, indoleamine 2,3-dioxygenase (IDO) expression and kynurenine metabolism was exaggerated in BDNF+/- mice. Genetic BDNF deficiency results in a dysregulated neuroinflammatory and metabolic response to peripheral immune challenge and in a specific vulnerability to the development of inflammation-induced anhedonia.
Collapse
Affiliation(s)
- Jennifer M Parrott
- Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States; Center for Biomedical Neuroscience and School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States
| | - Grace A Porter
- Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States; Center for Biomedical Neuroscience and School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States
| | - Laney Redus
- Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States
| | - Jason C O'Connor
- Audie L. Murphy VA Hospital, South Texas Veterans Heath System, San Antonio, TX 78229-4404, United States; Department of Pharmacology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States; Center for Biomedical Neuroscience and School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States; Mood Disorders Translational Research Core, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, United States.
| |
Collapse
|
4
|
Fallon IP, Tanner MK, Greenwood BN, Baratta MV. Sex differences in resilience: Experiential factors and their mechanisms. Eur J Neurosci 2020; 52:2530-2547. [PMID: 31800125 PMCID: PMC7269860 DOI: 10.1111/ejn.14639] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/31/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
Adverse life events can lead to stable changes in brain structure and function and are considered primary sources of risk for post-traumatic stress disorder, depression and other neuropsychiatric disorders. However, most individuals do not develop these conditions following exposure to traumatic experiences, and research efforts have identified a number of experiential factors associated with an individual's ability to withstand, adapt to and facilitate recovery from adversity. While multiple animal models of stress resilience exist, so that the detailed biological mechanisms can be explored, studies have been disproportionately conducted in male subjects even though the prevalence and presentation of stress-linked disorders differ between sexes. This review focuses on (a) the mechanisms by which experiential factors (behavioral control over a stressor, exercise) reduce the impact of adverse events as studied in males; (b) whether other manipulations (ketamine) that buffer against stress-induced sequelae engage the same circuit features; and (c) whether these processes operate similarly in females. We argue that investigation of experiential factors that produce resistance/resilience rather than vulnerability to adversity will generate a unique set of biological mechanisms that potentially underlie sex differences in mood disorders.
Collapse
Affiliation(s)
- Isabella P. Fallon
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Margaret K. Tanner
- Department of Integrative Biology, University of Colorado Denver, Denver, CO, 80217, USA
| | | | - Michael V. Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| |
Collapse
|
5
|
Zangiabadi I, Afarinesh MR, Shamsara A, Eftekhar-Vaghefi SH. Movento effects on learning and hippocampal brain-derived neurotrophic factor protein of adult male rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:36615-36622. [PMID: 31734837 DOI: 10.1007/s11356-019-06809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 10/16/2019] [Indexed: 06/10/2023]
Abstract
Spirotetramat is a toxic commercially known as Movento used to control pistachio psylla pests. In the present study, the effects of Movento on passive avoidance learning of rats and their ability to explore the novel object in the novel object recognition test were investigated. The changes in the concentration of hippocampal brain-derived neurotrophic factor (BDNF) proteins were evaluated, too. Male Wistar rats were gavaged at different dosages of the Movento (50, 100, 250, 500, 1000, 1250, and 1500 mg/kg) or saline for 7 days (administered every 2 days). We showed that Movento caused 50 and 100% mortality at the dose of 1250 and 1500 mg/kg, respectively. At the dose of 1000 mg/kg, Movento significantly decreased locomotor activity (P < 0.05). These rats also displayed a significant decrease in the number of training trials in the shuttle box and the ability to recognize a novel object compared with the control group (P < 0.01). The BDNF protein level of hippocampus also showed a significant decrease in the Movento (1000 mg/kg) compared with the control group (P < 0.01) while the number of pancellular necrosis pyramidal CA1 cells increased significantly in the Movento group (P < 0.001). We concluded that exposure to Movento can decline sensory, motor, and learning in rats.
Collapse
Affiliation(s)
- Iman Zangiabadi
- Department of Anatomy, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Afarinesh
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, 76198-13159, Kerman, Iran.
- Kerman Cognitive Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Ali Shamsara
- Department of Anatomy, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Hasan Eftekhar-Vaghefi
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, 76198-13159, Kerman, Iran.
- Department of Anatomy, Kerman Branch, Islamic Azad University, Kerman, Iran.
| |
Collapse
|
6
|
Storace A, Daniels S, Zhou Y, Kalisch B, Parker L, Rock E, Limebeer C, Lapointe T, Leri F. A study of limbic brain derived neurotrophic factor gene expression in male Sprague-Dawley rats trained on a learned helplessness task. Behav Brain Res 2019; 376:112174. [PMID: 31449911 DOI: 10.1016/j.bbr.2019.112174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/08/2019] [Accepted: 08/22/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Brain derived neurotrophic factor (BDNF) has been linked to the etiology and pathology of Major Depressive Disorder (MDD). Here, the relationship between learned helplessness (LH), a cognitive/motivational state relevant to MDD, and BDNF mRNA in various limbic regions, was investigated. METHODS In Sprague-Dawley male rats, LH was induced by escape training, using a triadic design of stressor controllability involving exposure to no shocks (NS), escapable shocks (ES) or yoked inescapable shocks (IES). LH was subsequently assessed in an active avoidance task, and levels of BDNF mRNA in limbic brain regions were compared across the triad following testing. RESULTS Although the IES group displayed greater LH, BDNF mRNA levels were lower in the hippocampus and higher in the nucleus accumbens of both IES and ES groups. In contrast, BDNF mRNA in the basolateral amygdala was elevated only in rats exposed to IES. CONCLUSION These results suggest that the inability to control an aversive stimulus can lead to a LH behavioural phenotype that is associated with region-specific alterations of BDNF gene expression in limbic nuclei.
Collapse
Affiliation(s)
- Alexandra Storace
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Stephen Daniels
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Yan Zhou
- Laboratory of the Biology of Addictive Disease, Rockefeller University, New York, NY, United States.
| | - Bettina Kalisch
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
| | - Linda Parker
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Erin Rock
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Cheryl Limebeer
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Thomas Lapointe
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Francesco Leri
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
7
|
Benatti C, Radighieri G, Alboni S, Blom JMC, Brunello N, Tascedda F. Modulation of neuroplasticity-related targets following stress-induced acute escape deficit. Behav Brain Res 2019; 364:140-148. [PMID: 30771367 DOI: 10.1016/j.bbr.2019.02.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/07/2019] [Indexed: 12/12/2022]
Abstract
Understanding resilience is a major challenge to improve current pharmacological therapies aimed at complementing psychological-based approaches of stress-related disorders. In particular, resilience is a multi-factorial construct where the complex network of molecular events that drive the process still needs to be resolved. Here, we exploit the acute escape deficit model, an animal model based on exposure to acute unavoidable stress followed by an escape test, to define vulnerable and resilient phenotypes in rats. Hippocampus and prefrontal cortex (PFC), two of the brain areas most involved in the stress response, were analysed for gene expression at two different time points (3 and 24 h) after the escape test. Total Brain-Derived Neurotrophic Factor (BDNF) was highly responsive in the PFC at 24-h after the escape test, while expression of BDNF transcript IV increased in the hippocampus of resistant animals 3 h post-test. Expression of memory enhancers like Neuronal PAS Domain Protein 4 (Npas4) and Activity-regulated cytoskeleton-associated protein (Arc) decreased in a time- and region-dependent fashion in both behavioural phenotypes. Also, the memory inhibitor Protein Phosphatase 1 (Ppp1ca) was increased in the hippocampus of resilient rats at 3 h post-test. Given the importance of neurotrophic factors and synaptic plasticity-related genes for the development of appropriate coping strategies, our data contribute to an additional step forward in the comprehension of the psychobiology of stress and resiliency.
Collapse
Affiliation(s)
- C Benatti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - G Radighieri
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - S Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - J M C Blom
- Department of Education and Human Sciences, University of Modena and Reggio Emilia, viale Antonio Allegri 9, 42121, Reggio Emilia, Italy; Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N Brunello
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - F Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
8
|
Wang X, Zou Z, Shen Q, Huang Z, Chen J, Tang J, Xue W, Tao W, Wu H, Wang D, Chen G. Involvement of NMDA-AKT-mTOR Signaling in Rapid Antidepressant-Like Activity of Chaihu-jia-Longgu-Muli-tang on Olfactory Bulbectomized Mice. Front Pharmacol 2019; 9:1537. [PMID: 30687098 PMCID: PMC6333740 DOI: 10.3389/fphar.2018.01537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Fast-onset antidepressants are urgently needed. Chaihu-jia-Longgu-Muli-tang (CLM), a classic Chinese herbal medicine, has been used for antidepressant treatment with long history. Olfactory bulbectomization (OB) model is validated for identification of rapid antidepressant efficacy. Here we used OB model for investigating the rapid onset activity of CLM in mice, and also tested the involvement of prefrontal Akt-mTOR and associated AMPA/NMDA receptors as well as hippocampal BDNF in the rapid antidepressant-like effect of CLM. Methods: The OB model was first characterized with depression-like behaviors and the time course changes of the behaviors. The fast onset of antidepressant effect of CLM was evaluated using sucrose preference test, tail suspension test and forced swim test in OB mice after a single administration. The expression of synaptic proteins of AMPA and NMDA subunits as well as Akt/mTOR signaling in the prefrontal cortex, and hippocampal BDNF was evaluated with the immunoblotting method. Results: A single dose of CLM significantly improved the deficiency in the sucrose preference and decreased the immobility time in the tail suspension test in OB mice. In the prefrontal cortex (PFC) in OB mice, there was lower expression level of the AMPA receptor subunit GluR1, rescued by a single dose of CLM. Additionally, the expression of NMDA subunit NR1 was up-regulated in OB mice, whereas mTOR and its upstream Akt signalings were both down-regulated. These deficiencies were reversed by a single dose of CLM. The CLM treatment also attenuated the expressions of NMDA receptor subunits NR2A and NR2B, which did not change in OB mice. In the hippocampus, expressions of GluR1 and brain derived neurotrophic factor (BDNF) were both up-regulated in OB mice, although CLM increased GluR1, but not BDNF. Conclusion: CLM elicited rapid antidepressant-like effects in the OB model mice, and CLM reversal of the abnormality in PFC expression of AMPA and NMDA receptors and associated Akt-mTOR signaling may underlie the effects.
Collapse
Affiliation(s)
- Xing Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhilu Zou
- Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qinqin Shen
- Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhiheng Huang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Chen
- Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Juanjuan Tang
- Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenda Xue
- Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Weiwei Tao
- Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Haoxin Wu
- Center for Translational Systems Biology and Neuroscience and Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dawei Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gang Chen
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, China
| |
Collapse
|
9
|
Depression and adult neurogenesis: Positive effects of the antidepressant fluoxetine and of physical exercise. Brain Res Bull 2018; 143:181-193. [PMID: 30236533 DOI: 10.1016/j.brainresbull.2018.09.002] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/03/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022]
Abstract
Of wide interest for health is the relation existing between depression, a very common psychological illness, accompanied by anxiety and reduced ability to concentrate, and adult neurogenesis. We will focus on two neurogenic stimuli, fluoxetine and physical exercise, both endowed with the ability to activate adult neurogenesis in the dentate gyrus of the hippocampus, known to be required for learning and memory, and both able to counteract depression. Fluoxetine belongs to the class of selective serotonin reuptake inhibitor (SSRI) antidepressants, which represent the most used pharmacological therapy; physical exercise has also been shown to effectively counteract depression symptoms in rodents as well as in humans. While there is evidence that the antidepressant effect of fluoxetine requires its pro-neurogenic action, exerted by promoting proliferation, differentiation and survival of progenitor cells of the hippocampus, on the other hand fluoxetine exerts also neurogenesis-independent antidepressant effects by influencing the plasticity of the new neurons generated. Similarly, the antidepressant action of running also correlates with an increase of hippocampal neurogenesis and plasticity, although the gene pathways involved are only partially coincident with those of fluoxetine, such as those involved in serotonin metabolism and synapse formation. We further discuss how extra-neurogenic actions are also suggested by the fact that, unlike running, fluoxetine is unable to stimulate neurogenesis during aging, but still displays antidepressant effects. Moreover, in specific conditions, fluoxetine or running activate not only progenitor but also stem cells, which normally are not stimulated; this fact reveals how stem cells have a long-term, hidden ability to self-renew and, more generally, that neurogenesis is subject to complex controls that may play a role in depression, such as the type of neurogenic stimulus or the state of the local niche. Finally, we discuss how fluoxetine or running are effective in counteracting depression originated from stress or neurodegenerative diseases.
Collapse
|
10
|
Phillips C. Physical Activity Modulates Common Neuroplasticity Substrates in Major Depressive and Bipolar Disorder. Neural Plast 2017; 2017:7014146. [PMID: 28529805 PMCID: PMC5424494 DOI: 10.1155/2017/7014146] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/10/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
Mood disorders (MDs) are chronic, recurrent mental diseases that affect millions of individuals worldwide. Although the biogenic amine model has provided some clinical utility, a need remains to better understand the interrelated mechanisms that contribute to neuroplasticity deficits in MDs and the means by which various therapeutics mitigate them. Of those therapeutics being investigated, physical activity (PA) has shown clear and consistent promise. Accordingly, the aims of this review are to (1) explicate key modulators, processes, and interactions that impinge upon multiple susceptibility points to effectuate neuroplasticity deficits in MDs; (2) explore the putative mechanisms by which PA mitigates these features; (3) review protocols used to induce the positive effects of PA in MDs; and (4) highlight implications for clinicians and researchers.
Collapse
|
11
|
Yin X, Guven N, Dietis N. Stress-based animal models of depression: Do we actually know what we are doing? Brain Res 2016; 1652:30-42. [DOI: 10.1016/j.brainres.2016.09.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/03/2016] [Accepted: 09/19/2016] [Indexed: 01/10/2023]
|
12
|
Mika A, Day HEW, Martinez A, Rumian NL, Greenwood BN, Chichlowski M, Berg BM, Fleshner M. Early life diets with prebiotics and bioactive milk fractions attenuate the impact of stress on learned helplessness behaviours and alter gene expression within neural circuits important for stress resistance. Eur J Neurosci 2016; 45:342-357. [PMID: 27763700 DOI: 10.1111/ejn.13444] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/27/2016] [Accepted: 10/17/2016] [Indexed: 12/25/2022]
Abstract
Manipulating gut microbes may improve mental health. Prebiotics are indigestible compounds that increase the growth and activity of health-promoting microorganisms, yet few studies have examined how prebiotics affect CNS function. Using an acute inescapable stressor known to produce learned helplessness behaviours such as failure to escape and exaggerated fear, we tested whether early life supplementation of a blend of two prebiotics, galactooligosaccharide (GOS) and polydextrose (PDX), and the glycoprotein lactoferrin (LAC) would attenuate behavioural and biological responses to stress later in life. Juvenile, male F344 rats were fed diets containing either GOS and PDX alone, LAC alone, or GOS, PDX and LAC. All diets altered gut bacteria, while diets containing GOS and PDX increased Lactobacillus spp. After 4 weeks, rats were exposed to inescapable stress, and either immediately killed for blood and tissues, or assessed for learned helplessness 24 h later. Diets did not attenuate stress effects on spleen weight, corticosterone and blood glucose; however, all diets differentially attenuated stress-induced learned helplessness. Notably, in situ hybridization revealed that all diets reduced stress-evoked cfos mRNA in the dorsal raphe nucleus (DRN), a structure important for learned helplessness behaviours. In addition, GOS, PDX and LAC diet attenuated stress-evoked decreases in mRNA for the 5-HT1A autoreceptor in the DRN and increased basal BDNF mRNA within the prefrontal cortex. These data suggest early life diets containing prebiotics and/or LAC promote behavioural stress resistance and uniquely modulate gene expression in corresponding circuits.
Collapse
Affiliation(s)
- Agnieszka Mika
- Department of Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO, 80309, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Heidi E W Day
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Alexander Martinez
- Department of Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO, 80309, USA
| | - Nicole L Rumian
- Department of Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO, 80309, USA
| | | | | | - Brian M Berg
- Mead Johnson Pediatric Nutrition Institute, Evansville, IN, USA
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO, 80309, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
13
|
Nicastro TM, Greenwood BN. Central monoaminergic systems are a site of convergence of signals conveying the experience of exercise to brain circuits involved in cognition and emotional behavior. Curr Zool 2016; 62:293-306. [PMID: 29491917 PMCID: PMC5804240 DOI: 10.1093/cz/zow027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 02/11/2016] [Indexed: 01/04/2023] Open
Abstract
Physical activity can enhance cognitive function and increase resistance against deleterious effects of stress on mental health. Enhanced cognitive function and stress resistance produced by exercise are conserved among vertebrates, suggesting that ubiquitous mechanisms may underlie beneficial effects of exercise. In the current review, we summarize the beneficial effects of exercise on cognitive function and stress resistance and discuss central and peripheral signaling factors that may be critical for conferring the effects of physical activity to brain circuits involved in cognitive function and stress. Additionally, it is suggested that norepinephrine and serotonin, highly conserved monoamines that are sensitive to exercise and able to modulate behavior in multiple species, could represent a convergence between peripheral and central exercise signals that mediate the beneficial effects of exercise. Finally, we offer the novel hypothesis that thermoregulation during exercise could contribute to the emotional effects of exercise by activating a subset of temperature-sensitive serotonergic neurons in the dorsal raphe nucleus that convey anxiolytic and stress-protective signals to forebrain regions. Throughout the review, we discuss limitations to current approaches and offer strategies for future research in exercise neuroscience.
Collapse
|
14
|
Su CL, Su CW, Hsiao YH, Gean PW. Epigenetic regulation of BDNF in the learned helplessness-induced animal model of depression. J Psychiatr Res 2016; 76:101-10. [PMID: 26921875 DOI: 10.1016/j.jpsychires.2016.02.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 02/03/2016] [Accepted: 02/12/2016] [Indexed: 01/22/2023]
Abstract
Major depressive disorder (MDD), one of the most common mental disorders, is a significant risk factor for suicide and causes a low quality of life for many people. However, the causes and underlying mechanism of depression remain elusive. In the current work, we investigated epigenetic regulation of BDNF in the learned helplessness-induced animal model of depression. Mice were exposed to inescapable stress and divided into learned helplessness (LH) and resilient (LH-R) groups depending on the number they failed to escape. We found that the LH group had longer immobility duration in the forced swimming test (FST) and tail suspension tests (TST), which is consistent with a depression-related phenotype. Western blotting analysis and enzyme-linked immunosorbent assay (ELISA) revealed that the LH group had lower BDNF expression than that of the LH-R group. The LH group consistently had lower BDNF mRNA levels, as detected by qPCR assay. In addition, we found BDNF exon IV was down-regulated in the LH group. Intraperitoneal injection of imipramine or histone deacetylase inhibitors (HDACi) to the LH mice for 14 consecutive days ameliorated depression-like behaviors and reversed the decrease in BDNF. The expression of HDAC5 was up-regulated in the LH mice, and a ChIP assay revealed that the level of HDAC5 binding to the promoter region of BDNF exon IV was higher than that seen in other groups. Knockdown of HDAC5 reduced depression-like behaviors in the LH mice. Taken together, these results suggest that epigenetic regulation of BDNF by HDAC5 plays an important role in the learned helplessness model of depression.
Collapse
Affiliation(s)
- Chun-Lin Su
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan
| | - Chun-Wei Su
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan
| | - Ya-Hsin Hsiao
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan
| | - Po-Wu Gean
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan.
| |
Collapse
|
15
|
Rao R, Ennis K, Mitchell EP, Tran PV, Gewirtz JC. Recurrent Moderate Hypoglycemia Suppresses Brain-Derived Neurotrophic Factor Expression in the Prefrontal Cortex and Impairs Sensorimotor Gating in the Posthypoglycemic Period in Young Rats. Dev Neurosci 2016; 38:74-82. [PMID: 26820887 DOI: 10.1159/000442878] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/29/2015] [Indexed: 01/04/2023] Open
Abstract
Recurrent hypoglycemia is common in infants and children. In developing rat models, recurrent moderate hypoglycemia leads to neuronal injury in the medial prefrontal cortex. To understand the effects beyond neuronal injury, 3-week-old male rats were subjected to 5 episodes of moderate hypoglycemia (blood glucose concentration, approx. 30 mg/dl for 90 min) once daily from postnatal day 24 to 28. Neuronal injury was determined using Fluoro-Jade B histochemistry on postnatal day 29. The effects on brain-derived neurotrophic factor (BDNF) and its cognate receptor, tyrosine kinase receptor B (TrkB) expression, which is critical for prefrontal cortex development, were determined on postnatal day 29 and at adulthood. The effects on prefrontal cortex-mediated function were determined by assessing the prepulse inhibition of the acoustic startle reflex on postnatal day 29 and 2 weeks later, and by testing for fear-potentiated startle at adulthood. Recurrent hypoglycemia led to neuronal injury confined primarily to the medial prefrontal cortex. BDNF/TrkB expression in the prefrontal cortex was suppressed on postnatal day 29 and was accompanied by lower prepulse inhibition, suggesting impaired sensorimotor gating. Following the cessation of recurrent hypoglycemia, the prepulse inhibition had recovered at 2 weeks. BDNF/TrkB expression in the prefrontal cortex had normalized and fear-potentiated startle was intact at adulthood. Recurrent moderate hypoglycemia during development has significant adverse effects on the prefrontal cortex in the posthypoglycemic period.
Collapse
|
16
|
Sciolino NR, Smith JM, Stranahan AM, Freeman KG, Edwards GL, Weinshenker D, Holmes PV. Galanin mediates features of neural and behavioral stress resilience afforded by exercise. Neuropharmacology 2015; 89:255-64. [PMID: 25301278 PMCID: PMC4250306 DOI: 10.1016/j.neuropharm.2014.09.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/23/2014] [Accepted: 09/28/2014] [Indexed: 12/15/2022]
Abstract
Exercise promotes resilience to stress and increases galanin in the locus coeruleus (LC), but the question of whether changes in galanin signaling mediate the stress-buffering effects of exercise has never been addressed. To test the contributions of galanin to stress resilience, male Sprague Dawley rats received intracerebroventricular (ICV) cannulation for drug delivery and frontocortical cannulation for microdialysis, and were housed with or without a running wheel for 21d. Rats were acutely injected with vehicle or the galanin receptor antagonist M40 and exposed to a single session of either footshock or no stress. Other groups received galanin, the galanin receptor antagonist M40, or vehicle chronically for 21d prior to the stress session. Microdialysis sampling occurred during stress exposure and anxiety-related behavior was measured on the following day in the elevated plus maze. Dendritic spines were visualized by Golgi impregnation in medial prefrontal cortex (mPFC) pyramidal neurons and quantified. Exercise increased galanin levels in the LC. Under non-stressed conditions, anxiety-related behavior and dopamine levels were comparable between exercised and sedentary rats. In contrast, exposure to stress reduced open arm exploration in sedentary rats but not in exercise rats or those treated chronically with ICV galanin, indicating improved resilience. Both exercise and chronic, ICV galanin prevented the increased dopamine overflow and loss of dendritic spines observed after stress in sedentary rats. Chronic, but not acute M40 administration blocked the resilience-promoting effects of exercise. The results indicate that increased galanin levels promote features of resilience at both behavioral and neural levels.
Collapse
Affiliation(s)
- N R Sciolino
- Neuroscience Program, Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA.
| | - J M Smith
- Neuroscience Program, Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA.
| | - A M Stranahan
- Physiology Department, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| | - K G Freeman
- Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA.
| | - G L Edwards
- Neuroscience Program, Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA; Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA.
| | - D Weinshenker
- Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - P V Holmes
- Neuroscience Program, Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA; Psychology Department, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
17
|
Cechella JL, Leite MR, Rosario AR, Sampaio TB, Zeni G. Diphenyl diselenide-supplemented diet and swimming exercise enhance novel object recognition memory in old rats. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9666. [PMID: 24994534 PMCID: PMC4150883 DOI: 10.1007/s11357-014-9666-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/18/2014] [Indexed: 06/03/2023]
Abstract
The benefits of exercise and the element selenium on mental health and cognitive performance are well documented. The purpose of the present study was to investigate whether the intake of a diet supplemented with diphenyl diselenide [(PhSe)2] and the swimming exercise could enhance memory in old Wistar rats. Male Wistar rats (24 months) were fed daily with standard diet chow or standard chow supplemented with 1 ppm of (PhSe)2 during 4 weeks. Animals were submitted to swimming training with a workload (3 % of body weight, 20 min/day for 4 weeks). After 4 weeks, the object recognition test (ORT) and the object location test (OLT) were performed. The results of this study demonstrated that intake of a supplemented diet with (PhSe)2 and swimming exercise was effective in improving short-term and long-term memory as well as spatial learning, increasing the hippocampal levels of phosphorylated cAMP-response element-binding protein (CREB) in old rats. This study also provided evidence that (PhSe)2-supplemented diet facilitated memory of old rats by modulating cAMP levels and stimulating CREB phosphorylation, without altering the levels of Akt.
Collapse
Affiliation(s)
- José L. Cechella
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul 97105-900 Brazil
| | - Marlon R. Leite
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul 97105-900 Brazil
| | - Alisson R. Rosario
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul 97105-900 Brazil
| | - Tuane B. Sampaio
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul 97105-900 Brazil
| | - Gilson Zeni
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul 97105-900 Brazil
| |
Collapse
|
18
|
Rodent models of depression: neurotrophic and neuroinflammatory biomarkers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:932757. [PMID: 24999483 PMCID: PMC4066721 DOI: 10.1155/2014/932757] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/18/2014] [Indexed: 12/13/2022]
Abstract
Rodent models are an indispensable tool for studying etiology and progress of depression. Since interrelated systems of neurotrophic factors and cytokines comprise major regulatory mechanisms controlling normal brain plasticity, impairments of these systems form the basis for development of cerebral pathologies, including mental diseases. The present review focuses on the numerous experimental rodent models of depression induced by different stress factors (exteroceptive and interoceptive) during early life (including prenatal period) or adulthood, giving emphasis to the data on the changes of neurotrophic factors and neuroinflammatory indices in the brain. These parameters are closely related to behavioral depression-like symptoms and impairments of neuronal plasticity and are both gender- and genotype-dependent. Stress-related changes in expression of neurotrophins and cytokines in rodent brain are region-specific. Some contradictory data reported by different groups may be a consequence of differences of stress paradigms or their realization in different laboratories. Like all experimental models, stress-induced depression-like conditions are experimental simplification of clinical depression states; however, they are suitable for understanding the involvement of neurotrophic factors and cytokines in the pathogenesis of the disease—a goal unachievable in the clinical reality. These major regulatory systems may be important targets for therapeutic measures as well as for development of drugs for treatment of depression states.
Collapse
|
19
|
Abstract
Exposure to an uncontrollable stressor elicits a constellation of physiological and behavioral sequel in laboratory rats that often reflect aspects of anxiety and other emotional disruptions. We review evidence suggesting that plasticity within the serotonergic dorsal raphe nucleus (DRN) is critical to the expression of uncontrollable stressor-induced anxiety. Specifically, after uncontrollable stressor exposure subsequent anxiogenic stimuli evoke greater 5-HT release in DRN terminal regions including the amygdala and striatum; and pharmacological blockade of postsynaptic 5-HT(2C) receptors in these regions prevents expression of stressor-induced anxiety. Importantly, the controllability of stress, the presence of safety signals, and a history of exercise mitigate the expression of stressor-induced anxiety. These stress-protective factors appear to involve distinct neural substrates; with stressor controllability requiring the medial prefrontal cortex, safety signals the insular cortex and exercise affecting the 5-HT system directly. Knowledge of the distinct yet converging mechanisms underlying these stress-protective factors could provide insight into novel strategies for the treatment and prevention of stress-related psychiatric disorders.
Collapse
|
20
|
Fleshner M, Greenwood BN, Yirmiya R. Neuronal-glial mechanisms of exercise-evoked stress robustness. Curr Top Behav Neurosci 2014; 18:1-12. [PMID: 24481547 DOI: 10.1007/7854_2014_277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stress robustness by definition, incorporates both stress resistance (organisms endure greater stressor intensity or duration before suffering negative consequences) and stress resilience (organisms recover faster after suffering negative consequences). Factors that influence stress robustness include the nature of the stressor, (i.e., controllability, intensity, chronicity) and features of the organism (i.e., age, genetics, sex, and physical activity status). Here we present a novel hypothesis for how physically active versus sedentary living promotes stress robustness in the face of intense uncontrollable stress. Advances in neurobiology have established microglia as an active player in the regulation of synaptic activity, and recent work has revealed mechanisms for modulating glial function, including cross talk between neurons and glia. This chapter presents supporting evidence that the physical activity status of an organism may modulate stress-evoked neuronal-glial responses by changing the CX3CL1-CX3CR1 axis. Specifically, we propose that sedentary animals respond to an intense acute uncontrollable stressor with excessive serotonin (5-HT) and noradrenergic (NE) activity and/or prolonged down-regulation of the CX3CL1-CX3CR1 axis resulting in activation and proliferation of hippocampal microglia in the absence of pathogenic signals and consequent hippocampal-dependent memory deficits and reduced neurogenesis. In contrast, physically active animals respond to the same stressor with constrained 5-HT and NE activity and rapidly recovering CX3CL1-CX3CR1 axis responses resulting in the quieting of microglia, and protection from negative cognitive and neurobiological effects of stress.
Collapse
Affiliation(s)
- Monika Fleshner
- Department of Integrative Physiology and The Center for Neuroscience, University of Colorado, Boulder, CO, 80309, USA,
| | | | | |
Collapse
|
21
|
Holmes PV. Trophic Mechanisms for Exercise-Induced Stress Resilience: Potential Role of Interactions between BDNF and Galanin. Front Psychiatry 2014; 5:90. [PMID: 25120496 PMCID: PMC4112800 DOI: 10.3389/fpsyt.2014.00090] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/14/2014] [Indexed: 12/11/2022] Open
Abstract
Current concepts of the neurobiology of stress-related disorders, such as anxiety and depression emphasize disruptions in neural plasticity and neurotrophins. The potent trophic actions of exercise, therefore, represent not only an effective means for prevention and treatment of these disorders, they also afford the opportunity to employ exercise paradigms as a basic research tool to uncover the neurobiological mechanisms underlying these disorders. Novel approaches to studying stress-related disorders focus increasingly on trophic factor signaling in corticolimbic circuits that both mediate and regulate cognitive, behavioral, and physiological responses to deleterious stress. Recent evidence demonstrates that the neural plasticity supported by these trophic mechanisms is vital for establishing and maintaining resilience to stress. Therapeutic interventions that promote these mechanisms, be they pharmacological, behavioral, or environmental, may therefore prevent or reverse stress-related mental illness by enhancing resilience. The present paper will provide an overview of trophic mechanisms responsible for the enhancement of resilience by voluntary exercise with an emphasis on brain-derived neurotrophic factor, galanin, and interactions between these two trophic factors.
Collapse
Affiliation(s)
- Philip V Holmes
- Neuroscience Program, Psychology Department, Biomedical and Health Sciences Institute, The University of Georgia , Athens, GA , USA
| |
Collapse
|
22
|
Sickmann HM, Li Y, Mørk A, Sanchez C, Gulinello M. Does stress elicit depression? Evidence from clinical and preclinical studies. Curr Top Behav Neurosci 2014; 18:123-159. [PMID: 24633891 DOI: 10.1007/7854_2014_292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Exposure to stressful situations may induce or deteriorate an already existing depression. Stress-related depression can be elicited at an adolescent/adult age but evidence also shows that early adverse experiences even at the fetal stage may predispose the offspring for later development of depression. The hypothalamus-pituitary-adrenal axis (HPA-axis) plays a key role in regulating the stress response and dysregulation in the system has been linked to depression both in humans and in animal models. This chapter critically reviews clinical and preclinical findings that may explain how stress can cause depression, including HPA-axis changes and alterations beyond the HPA-axis. As stress does not elicit depression in the majority of the population, this motivated research to focus on understanding the biology underlying resilient versus sensitive subjects. Animal models of depression have contributed to a deeper understanding of these mechanisms. Findings from these models will be presented.
Collapse
Affiliation(s)
- Helle M Sickmann
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
23
|
Sasse SK, Nyhuis TJ, Masini CV, Day HEW, Campeau S. Central gene expression changes associated with enhanced neuroendocrine and autonomic response habituation to repeated noise stress after voluntary wheel running in rats. Front Physiol 2013; 4:341. [PMID: 24324441 PMCID: PMC3839297 DOI: 10.3389/fphys.2013.00341] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/05/2013] [Indexed: 01/15/2023] Open
Abstract
Accumulating evidence indicates that regular physical exercise benefits health in part by counteracting some of the negative physiological impacts of stress. While some studies identified reductions in some measures of acute stress responses with prior exercise, limited data were available concerning effects on cardiovascular function, and reported effects on hypothalamic-pituitary-adrenocortical (HPA) axis responses were largely inconsistent. Given that exposure to repeated or prolonged stress is strongly implicated in the precipitation and exacerbation of illness, we proposed the novel hypothesis that physical exercise might facilitate adaptation to repeated stress, and subsequently demonstrated significant enhancement of both HPA axis (glucocorticoid) and cardiovascular (tachycardia) response habituation to repeated noise stress in rats with long-term access to running wheels compared to sedentary controls. Stress habituation has been attributed to modifications of brain circuits, but the specific sites of adaptation and the molecular changes driving its expression remain unclear. Here, in situ hybridization histochemistry was used to examine regulation of select stress-associated signaling systems in brain regions representing likely candidates to underlie exercise-enhanced stress habituation. Analyzed brains were collected from active (6 weeks of wheel running) and sedentary rats following control, acute, or repeated noise exposures that induced a significantly faster rate of glucocorticoid response habituation in active animals but preserved acute noise responsiveness. Nearly identical experimental manipulations also induce a faster rate of cardiovascular response habituation in exercised, repeatedly stressed rats. The observed regulation of the corticotropin-releasing factor and brain-derived neurotrophic factor systems across several brain regions suggests widespread effects of voluntary exercise on central functions and related adaptations to stress across multiple response modalities.
Collapse
Affiliation(s)
- Sarah K Sasse
- Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | | | | | | | | |
Collapse
|
24
|
Forced and voluntary exercises equally improve spatial learning and memory and hippocampal BDNF levels. Behav Brain Res 2013; 247:34-9. [DOI: 10.1016/j.bbr.2013.03.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/01/2013] [Accepted: 03/05/2013] [Indexed: 11/23/2022]
|
25
|
Greenwood BN, Loughridge AB, Sadaoui N, Christianson JP, Fleshner M. The protective effects of voluntary exercise against the behavioral consequences of uncontrollable stress persist despite an increase in anxiety following forced cessation of exercise. Behav Brain Res 2012; 233:314-21. [PMID: 22610051 DOI: 10.1016/j.bbr.2012.05.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 05/09/2012] [Accepted: 05/10/2012] [Indexed: 10/28/2022]
Abstract
Humans who exercise are less likely to suffer from stress-related mood disorders. Similarly, rats allowed voluntary access to running wheels have constrained corticosterone responses to mild stressors and are protected against several behavioral consequences of uncontrollable stress which resemble symptoms of human anxiety and depression, including exaggerated fear and deficits in shuttle box escape learning. Although exercise conveys clear stress resistance, the duration of time the protective effects of exercise against the behavioral consequences of uncontrollable stress persist following exercise cessation is unknown. The current studies investigated (1) whether exercise-induced stress resistance extends to social avoidance, another anxiety-like behavior elicited by uncontrollable stressor exposure, and (2) the duration of time the protective effects of exercise persist following forced cessation of exercise. Six weeks of wheel running constrained the increase in corticosterone elicited by social exploration testing, and prevented the reduction in social exploration, exaggerated shock-elicited fear, and deficits in escape learning produced by uncontrollable stress. The protective effect of voluntary exercise against stress-induced interference with escape learning persisted for 15 days, but was lost by 25 days, following cessation of exercise. An anxiogenic effect, as revealed by a reduction in social exploration and an increase in fear behavior immerged as a function of time following cessation of exercise. Results demonstrate that the protective effect of voluntary exercise against the behavioral consequences of uncontrollable stress extends to include social avoidance, and can persist for several days following exercise cessation despite an increase in anxiety produced by forced cessation of exercise.
Collapse
Affiliation(s)
- Benjamin N Greenwood
- Department of Integrative Physiology, University of Colorado-Boulder, CO 80309-0354, USA.
| | | | | | | | | |
Collapse
|
26
|
Andero R, Ressler KJ. Fear extinction and BDNF: translating animal models of PTSD to the clinic. GENES BRAIN AND BEHAVIOR 2012; 11:503-12. [PMID: 22530815 DOI: 10.1111/j.1601-183x.2012.00801.x] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is the most studied neurotrophin involved in synaptic plasticity processes that are required for long-term learning and memory. Specifically, BDNF gene expression and activation of its high-affinity tropomyosin-related kinase B (TrkB) receptor are necessary in the amygdala, hippocampus and prefrontal cortex for the formation of emotional memories, including fear memories. Among the psychiatric disorders with altered fear processing, there is post-traumatic stress disorder (PTSD) which is characterized by an inability to extinguish fear memories. Since BDNF appears to enhance extinction of fear, targeting impaired extinction in anxiety disorders such as PTSD via BDNF signalling may be an important and novel way to enhance treatment efficacy. The aim of this review is to provide a translational point of view that stems from findings in the BDNF regulation of synaptic plasticity and fear extinction. In addition, there are different systems that seem to alter fear extinction through BDNF modulation like the endocannabinoid system and the hypothalamic-pituitary adrenal axis. Recent work also finds that the pituitary adenylate cyclase-activating polypeptide and PAC1 receptor, which are upstream of BDNF activation, may be implicated in PTSD. Especially interesting are data that exogenous fear extinction enhancers such as antidepressants, histone deacetylases inhibitors and D-cycloserine, a partial N-methyl d-aspartate agonist, may act through or in concert with the BDNF-TrkB system. Finally, we review studies where recombinant BDNF and a putative TrkB agonist, 7,8-dihydroxyflavone, may enhance extinction of fear. These approaches may lead to novel agents that improve extinction in animal models and eventually humans.
Collapse
Affiliation(s)
- R Andero
- Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
27
|
Zhou W, Chen L, Yang S, Li F, Li X. Behavioral stress-induced activation of FoxO3a in the cerebral cortex of mice. Biol Psychiatry 2012; 71:583-92. [PMID: 21978520 PMCID: PMC3254805 DOI: 10.1016/j.biopsych.2011.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/22/2011] [Accepted: 08/25/2011] [Indexed: 01/22/2023]
Abstract
BACKGROUND The transcription factor FoxO3a is highly expressed in brain, but little is known about the response of FoxO3a to behavioral stress and its impact in the associated behavioral changes. METHODS We tested the response of brain FoxO3a in the learned helplessness (LH) paradigm and tested signaling pathways that mediate the response of FoxO3a. RESULTS A single session of inescapable shocks (IES) in mice reduced FoxO3a phosphorylation at the Akt-regulating serine/threonine residues and induced prolonged nuclear accumulation of FoxO3a in the cerebral cortex, both indicating activation of FoxO3a in brain. The response of FoxO3a is accompanied by a transient inactivation of Akt and a prolonged activation of glycogen synthase kinase-3beta (GSK3β). Noticeably, FoxO3a formed a protein complex with GSK3β in the cerebral cortex, and the interaction between the two proteins was stronger in IES-treated mice. Inhibition of glycogen synthase kinase-3 was able to abolish IES-induced LH behavior, disrupt IES-induced GSK3β-FoxO3a interaction, and reduce nuclear FoxO3a accumulation. In vitro approaches further revealed that the interaction between GSK3β and FoxO3a was strongest when both were active; FoxO3a was phosphorylated by recombinant GSK3β; and glycogen synthase kinase-3 inhibitors effectively reduced FoxO3a transcriptional activity. Importantly, IES-induced LH behavior was markedly diminished in FoxO3a-deficient mice that had minimal FoxO3a expression and reduced levels of FoxO3a-inducible genes. CONCLUSIONS FoxO3a is activated in response to IES by interacting with GSK3β, and inhibition of GSK3β or reducing FoxO3a expression promotes resistance to stress-induced behavioral disturbance by disrupting this signaling mechanism.
Collapse
Affiliation(s)
- Wenjun Zhou
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 35294, USA
| | | | | | | | | |
Collapse
|
28
|
Novak CM, Burghardt PR, Levine JA. The use of a running wheel to measure activity in rodents: relationship to energy balance, general activity, and reward. Neurosci Biobehav Rev 2012; 36:1001-1014. [PMID: 22230703 DOI: 10.1016/j.neubiorev.2011.12.012] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 12/07/2011] [Accepted: 12/22/2011] [Indexed: 12/21/2022]
Abstract
Running wheels are commonly employed to measure rodent physical activity in a variety of contexts, including studies of energy balance and obesity. There is no consensus on the nature of wheel-running activity or its underlying causes, however. Here, we will begin by systematically reviewing how running wheel availability affects physical activity and other aspects of energy balance in laboratory rodents. While wheel running and physical activity in the absence of a wheel commonly correlate in a general sense, in many specific aspects the two do not correspond. In fact, the presence of running wheels alters several aspects of energy balance, including body weight and composition, food intake, and energy expenditure of activity. We contend that wheel-running activity should be considered a behavior in and of itself, reflecting several underlying behavioral processes in addition to a rodent's general, spontaneous activity. These behavioral processes include defensive behavior, predatory aggression, and depression- and anxiety-like behaviors. As it relates to energy balance, wheel running engages several brain systems-including those related to the stress response, mood, and reward, and those responsive to growth factors-that influence energy balance indirectly. We contend that wheel-running behavior represents factors in addition to rodents' tendency to be physically active, engaging additional neural and physiological mechanisms which can then independently alter energy balance and behavior. Given the impact of wheel-running behavior on numerous overlapping systems that influence behavior and physiology, this review outlines the need for careful design and interpretation of studies that utilize running wheels as a means for exercise or as a measurement of general physical activity.
Collapse
Affiliation(s)
- Colleen M Novak
- Department of Biological Sciences, Kent State University, PO Box 5190, 222 Cunningham Hall, Kent, OH 44242, United States
| | | | - James A Levine
- Mayo Clinic, Endocrine Research Unit, Rochester, MN 55905, United States
| |
Collapse
|
29
|
Abstract
Voluntary exercise reduces the incidence of stress-related psychiatric disorders in humans and prevents serotonin-dependent behavioral consequences of stress in rodents. Evidence reviewed herein is consistent with the hypothesis that exercise increases stress resistance by producing neuroplasticity at multiple sites of the central serotonergic system, which all help to limit the behavioral impact of acute increases in serotonin during stressor exposure.
Collapse
|
30
|
Strong PV, Christianson JP, Loughridge AB, Amat J, Maier SF, Fleshner M, Greenwood BN. 5-hydroxytryptamine 2C receptors in the dorsal striatum mediate stress-induced interference with negatively reinforced instrumental escape behavior. Neuroscience 2011; 197:132-44. [PMID: 21958863 DOI: 10.1016/j.neuroscience.2011.09.041] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 09/19/2011] [Accepted: 09/19/2011] [Indexed: 12/28/2022]
Abstract
Uncontrollable stress can interfere with instrumental learning and induce anxiety in humans and rodents. While evidence supports a role for serotonin (5-HT) and serotonin 2C receptors (5-HT(2C)R) in the behavioral consequences of uncontrollable stress, the specific sites of action are unknown. These experiments sought to delineate the role of 5-HT and 5-HT(2C)R in the dorsal striatum (DS) and the lateral/basolateral amygdala (BLA) in the expression of stress-induced instrumental escape deficits and exaggerated fear, as these structures are critical to instrumental learning and fear behaviors. Using in vivo microdialysis, we first demonstrated that prior uncontrollable, but not controllable, stress sensitizes extracellular 5-HT in the dorsal striatum, a result that parallels prior work in the BLA. Additionally, rats were implanted with bi-lateral cannula in either the DS or the BLA and exposed to uncontrollable tail shock stress. One day later, rats were injected with 5-HT(2C)R antagonist (SB242084) and fear and instrumental learning behaviors were assessed in a shuttle box. Separately, groups of non-stressed rats received an intra-DS or an intra-BLA injection of the 5-HT(2C)R agonist (CP809101) and behavior was observed. Intra-DS injections of the 5-HT(2C)R antagonist prior to fear/escape tests completely blocked the stress-induced interference with instrumental escape learning; a partial block was observed when injections were in the BLA. Antagonist administration in either region did not influence stress-induced fear behavior. In the absence of prior stress, intra-DS administration of the 5-HT(2C)R agonist was sufficient to interfere with escape behavior without enhancing fear, while intra-BLA administration of the 5-HT(2C)R agonist increased fear behavior but had no effect on escape learning. Results reveal a novel role of the 5-HT(2C)R in the DS in the expression of instrumental escape deficits produced by uncontrollable stress and demonstrate that the involvement of 5-HT(2C)R activation in stress-induced behaviors is regionally specific.
Collapse
Affiliation(s)
- P V Strong
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80309-0354, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Farley S, Dumas S, El Mestikawy S, Giros B. Increased expression of the Vesicular Glutamate Transporter-1 (VGLUT1) in the prefrontal cortex correlates with differential vulnerability to chronic stress in various mouse strains: effects of fluoxetine and MK-801. Neuropharmacology 2011; 62:503-17. [PMID: 21945287 DOI: 10.1016/j.neuropharm.2011.09.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 09/07/2011] [Accepted: 09/08/2011] [Indexed: 12/27/2022]
Abstract
Major depression is a chronic psychiatric illness that is highly prevalent and disabling. The available medications are ineffective for many patients suggesting that differents molecular pathways could be specifically altered in the unresponsive patients. Recently, the glutamatergic system has emerged as a target in the research on depression and acute NMDA receptor blockade has been shown to produce strong antidepressant effects. We have studied the adaptations of four mice strains (C57BL/6, DBA/2, C3H and BALB/c) to a chronic unpredictable stress protocol, a widely used model of depression in rodents. BALB/c mice displayed strikingly different behavioral and neurochemical adaptations compared to the other strains tested, suggesting that different molecular pathways are involved in their specific vulnerability. They became hyperactive during the dark period, anhedonic-like and displayed no alterations in the tail suspension test (TST). After chronic stress, only the BALB/c displayed an increased frontocortical VGLUT1 expression which is suggestive of a dysregulation of their prefrontal glutamatergic system, and no BDNF mRNA alteration, although the acute stress modulation of this mRNA is similar to the other strains. Chronic administration of an antagonist of NMDA receptors, MK-801, induced antidepressant-like effects in the TST for stressed BALB/c, but was ineffective for the hyperactivity and anhedonia-like behavior, in contrast to fluoxetine. Chronic MK-801 was totally inactive on the behavior of stressed C57BL/6 mice. MK-801, but not fluoxetine, inhibited the VGLUT1 prefrontal increase in BALB/c. Fluoxetine increased VGLUT1 and BDNF mRNA expression in the hippocampus of the C57BL/6 but not in the BALB/c strain, suggesting a different reactivity in-between strain to both stress and antidepressant. Interestingly enough, the BDNF or VGLUT1 increase is not necessary to reverse the stress induced behavioral alterations in our experimental settings. This observation supports the conclusion that BDNF and VGLUT1 are depressive state markers, but not involved in its etiology. Finally, there is a substantial similarity between the phenotypes that are observed in the BALB/c mice and endogenous depression in humans, as well as between C57BL/6 mice and atypical depression. To have a better understanding of the variability of depression etiologies in human, and the implication of the glutamatergic system, it may be suggested that future animal studies in the mouse would systematically compare the two strains BALB/c and C57BL/6 for the identification of relevant biological mechanisms. This article is part of a special Issue entitled 'Anxiety and Depression'.
Collapse
Affiliation(s)
- Séverine Farley
- INSERM U952 Physiopathologie des Maladies du Système nerveux Central, 9 Quai St Bernard, 75005 Paris, France
| | | | | | | |
Collapse
|
32
|
Kubera M, Obuchowicz E, Goehler L, Brzeszcz J, Maes M. In animal models, psychosocial stress-induced (neuro)inflammation, apoptosis and reduced neurogenesis are associated to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:744-59. [PMID: 20828592 DOI: 10.1016/j.pnpbp.2010.08.026] [Citation(s) in RCA: 328] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 08/28/2010] [Accepted: 08/29/2010] [Indexed: 12/13/2022]
Abstract
Recently, the inflammatory and neurodegenerative (I&ND) hypothesis of depression was formulated (Maes et al., 2009), i.e. the neurodegeneration and reduced neurogenesis that characterize depression are caused by inflammation, cell-mediated immune activation and their long-term sequels. The aim of this paper is to review the body of evidence that external stressors may induce (neuro)inflammation, neurodegeneration and reduced neurogenesis; and that antidepressive treatments may impact on these pathways. The chronic mild stress (CMS) and learned helplessness (LH) models show that depression-like behaviors are accompanied by peripheral and central inflammation, neuronal cell damage, decreased neurogenesis and apoptosis in the hippocampus. External stress-induced depression-like behaviors are associated with a) increased interleukin-(IL)1β, tumor necrosis factor-α, IL-6, nuclear factor κB, cyclooxygenase-2, expression of Toll-like receptors and lipid peroxidation; b) antineurogenic effects and reduced brain-derived neurotrophic factor (BDNF) levels; and c) apoptosis with reduced levels of Bcl-2 and BAG1 (Bcl-2 associated athanogene 1), and increased levels of caspase-3. Stress-induced inflammation, e.g. increased IL-1β, but not reduced neurogenesis, is sufficient to cause depression. Antidepressants a) reduce peripheral and central inflammatory pathways by decreasing IL-1β, TNFα and IL-6 levels; b) stimulate neuronal differentiation, synaptic plasticity, axonal growth and regeneration through stimulatory effects on the expression of different neurotrophic factors, e.g. trkB, the receptor for brain-derived neurotrophic factor; and c) attenuate apoptotic pathways by activating Bcl-2 and Bcl-xl proteins, and suppressing caspase-3. It is concluded that external stressors may provoke depression-like behaviors through activation of inflammatory, oxidative, apoptotic and antineurogenic mechanisms. The clinical efficacity of antidepressants may be ascribed to their ability to reverse these different pathways.
Collapse
Affiliation(s)
- Marta Kubera
- Department of Experimental Endocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, PL 31-343 Kraków, Poland.
| | | | | | | | | |
Collapse
|
33
|
Nyhuis TJ, Masini CV, Sasse SK, Day HEW, Campeau S. Physical activity, but not environmental complexity, facilitates HPA axis response habituation to repeated audiogenic stress despite neurotrophin mRNA regulation in both conditions. Brain Res 2010; 1362:68-77. [PMID: 20851112 DOI: 10.1016/j.brainres.2010.09.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 09/03/2010] [Accepted: 09/10/2010] [Indexed: 02/05/2023]
Abstract
Stress exacerbates several physical and psychological disorders. Voluntary exercise can reduce susceptibility to many of these stress-associated disorders. In rodents, voluntary exercise can reduce hypothalamic-pituitary-adrenocortical (HPA) axis activity in response to various stressors as well as upregulate several brain neurotrophins. An important issue regarding voluntary exercise is whether its effect on the reduction of HPA axis activation in response to stress is due to the physical activity itself or simply the enhanced environmental complexity provided by the running wheels. The present study compared the effects of physical activity and environmental complexity (that did not increase physical activity) on HPA axis habituation to repeated stress and modulation of brain neurotrophin mRNA expression. For six weeks, male rats were given free access to running wheels (exercise group), given 4 objects that were repeatedly exchanged (increased environmental complexity group), or housed in standard cages. On week 7, animals were exposed to 11 consecutive daily 30-min sessions of 98-dBA noise. Plasma corticosterone and adrenocorticotropic hormone were measured from blood collected directly after noise exposures. Tissue, including brains, thymi, and adrenal glands was collected on Day 11. Although rats in both the exercise and enhanced environmental complexity groups expressed higher levels of BDNF and NGF mRNA in several brain regions, only exercise animals showed quicker glucocorticoid habituation to repeated audiogenic stress. These results suggest that voluntary exercise, independent from other environmental manipulations, accounts for the reduction in susceptibility to stress.
Collapse
Affiliation(s)
- Tara J Nyhuis
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO 80309, USA.
| | | | | | | | | |
Collapse
|
34
|
Rajkumar R, Mahesh R. Assessing the neuronal serotonergic target-based antidepressant stratagem: impact of in vivo interaction studies and knockout models. Curr Neuropharmacol 2010; 6:215-34. [PMID: 19506722 PMCID: PMC2687932 DOI: 10.2174/157015908785777256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 06/05/2008] [Accepted: 06/12/2008] [Indexed: 12/26/2022] Open
Abstract
Depression remains a challenge in the field of affective neuroscience, despite a steady research progress. Six out of nine basic antidepressant mechanisms rely on serotonin neurotransmitter system. Preclinical studies have demonstrated the significance of serotonin receptors (5-HT1-3,6,7), its signal transduction pathways and classical down stream targets (including neurotrophins, neurokinins, other peptides and their receptors) in antidepressant drug action. Serotonergic control of depression embraces the recent molecular requirements such as influence on proliferation, neurogenesis, plasticity, synaptic (re)modeling and transmission in the central nervous system. The present progress report analyses the credibility of each protein as therapeutically relevant target of depression. In vivo interaction studies and knockout models which identified these targets are foreseen to unearth new ligands and help them transform to drug candidates. The importance of the antidepressant assay selection at the preclinical level using salient animal models/assay systems is discussed. Such test batteries would definitely provide antidepressants with faster onset, efficacy in resistant (and co-morbid) types and with least adverse effects. Apart from the selective ligands, only those molecules which bring an overall harmony, by virtue of their affinities to various receptor subtypes, could qualify as effective antidepressants. Synchronised modulation of various serotonergic sub-pathways is the basis for a unique and balanced antidepressant profile, as that of fluoxetine (most exploited antidepressant) and such a profile may be considered as a template for the upcoming antidepressants. In conclusion, 5-HT based multi-targeted antidepressant drug discovery supported by in vivo interaction studies and knockout models is advocated as a strategy to provide classic molecules for clinical trials.
Collapse
Affiliation(s)
- R Rajkumar
- Pharmacy Group, FD-III, Vidya Vihar, Birla Institute of Technology & Science, Pilani, Rajasthan-333031, India.
| | | |
Collapse
|
35
|
Campeau S, Nyhuis TJ, Kryskow EM, Masini CV, Babb JA, Sasse SK, Greenwood BN, Fleshner M, Day HEW. Stress rapidly increases alpha 1d adrenergic receptor mRNA in the rat dentate gyrus. Brain Res 2010; 1323:109-18. [PMID: 20138850 DOI: 10.1016/j.brainres.2010.01.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 01/26/2010] [Accepted: 01/28/2010] [Indexed: 01/30/2023]
Abstract
The hippocampal formation is a highly plastic brain region that is sensitive to stress. It receives extensive noradrenergic projections, and noradrenaline is released in the hippocampus in response to stressor exposure. The hippocampus expresses particularly high levels of the alpha(1D) adrenergic receptor (ADR) and we have previously demonstrated that alpha(1d) ADR mRNA expression in the rat hippocampus is modulated by corticosterone. One of the defining features of a stress response is activation of the hypothalamic pituitary adrenal (HPA) axis, resulting in the release of corticosterone from the adrenal glands. However, the effect of stress on hippocampal expression of alpha(1d) ADR mRNA has not been determined. In this study, male rats were exposed to inescapable tail shock, loud noise or restraint, and the effect on alpha(1d) ADR mRNA expression in the hippocampus was determined by semi-quantitative in situ hybridization. All three stressors resulted in a rapid upregulation of alpha(1d) ADR mRNA in the dentate gyrus, with expression peaking at approximately 90min after the start of the stressor. Physical activity has previously been reported to counteract some of the effects of stress that occur within the dentate gyrus. However, 6weeks of voluntary wheel running in rats did not prevent the restraint stress-induced increase in alpha(1d) ADR mRNA expression in the dentate gyrus. Although the function of the alpha(1D) ADR in the dentate gyrus is not known, these data provide further evidence for a close interaction between stress and the noradrenergic system in the hippocampus.
Collapse
Affiliation(s)
- Serge Campeau
- Department of Psychology and Neuroscience, University of Colorado, Boulder, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hajszan T, Szigeti-Buck K, Sallam NL, Bober J, Parducz A, MacLusky NJ, Leranth C, Duman RS. Effects of estradiol on learned helplessness and associated remodeling of hippocampal spine synapses in female rats. Biol Psychiatry 2010; 67:168-74. [PMID: 19811775 PMCID: PMC2794927 DOI: 10.1016/j.biopsych.2009.08.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 08/12/2009] [Accepted: 08/15/2009] [Indexed: 01/18/2023]
Abstract
BACKGROUND Despite the fact that women are twice as likely to develop depression as men, our understanding of depression neurobiology in female subjects is limited. We have recently reported in male rats that development of helpless behavior is associated with a severe loss of hippocampal spine synapses, which is reversed by treatment with the antidepressant desipramine. Considering that estradiol has a hippocampal synaptogenic effect similar to those of antidepressants, the presence of estradiol during the female reproductive life might influence behavioral and synaptic responses to stress and depression. METHODS With electron microscopic stereology, we analyzed hippocampal spine synapses in association with helpless behavior in ovariectomized female rats (n = 70), under different conditions of estradiol exposure. RESULTS Stress induced an acute and persistent loss of hippocampal spine synapses, whereas subchronic treatment with desipramine reversed the stress-induced synaptic loss. Estradiol supplementation given either before stress or before escape testing of nonstressed animals increased the number of hippocampal spine synapses. Correlation analysis demonstrated a statistically significant negative correlation between the severity of helpless behavior and hippocampal spine synapse numbers. CONCLUSIONS These findings suggest that hippocampal spine synapse remodeling might be a critical factor underlying learned helplessness and, possibly, the neurobiology of depression.
Collapse
Affiliation(s)
- Tibor Hajszan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | - Klara Szigeti-Buck
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine; New Haven, CT 06520, USA,Department of Pharmacology, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Nermin L Sallam
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Jeremy Bober
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Arpad Parducz
- Institute of Biophysics, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Neil J MacLusky
- Department of Biomedical Sciences, Ontario Veterinary College, Guelph, Ontario, Canada N1G 2W1
| | - Csaba Leranth
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine; New Haven, CT 06520, USA,Department of Neurobiology; Yale University School of Medicine; New Haven, CT 06520, USA
| | - Ronald S Duman
- Department of Pharmacology, Yale University School of Medicine; New Haven, CT 06520, USA,Department of Psychiatry; Yale University School of Medicine; New Haven, CT 06520, USA
| |
Collapse
|
37
|
Guimarães FS, Zangrossi H, Del Ben CM, Graeff FG. Serotonin in Panic and Anxiety Disorders. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2010. [DOI: 10.1016/s1569-7339(10)70105-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
38
|
Greenwood BN, Strong PV, Foley TE, Fleshner M. A behavioral analysis of the impact of voluntary physical activity on hippocampus-dependent contextual conditioning. Hippocampus 2009; 19:988-1001. [PMID: 19115374 DOI: 10.1002/hipo.20534] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Voluntary physical activity induces molecular changes in the hippocampus consistent with improved hippocampal function, but few studies have explored the effects of wheel running on specific hippocampal-dependent learning and memory processes. The current studies investigated the impact of voluntary wheel running on learning and memory for context and extinction using contextual fear conditioning which is known to be dependent on the hippocampus. When conditioning occurred prior to the start of 6 weeks of wheel running, wheel running had no effect on memory for context or extinction (assessed with freezing). In contrast, when wheel running occurred for 6 weeks prior to conditioning, physical activity improved contextual memory during a retention test 24 h later, but did not affect extinction learning or memory. Wheel running had no effect on freezing immediately after foot shock presentation during conditioning, suggesting that physical activity does not affect the acquisition of the context-shock association or alter the expression of freezing, per se. Instead, it is argued that physical activity improves the consolidation of contextual memories in the hippocampus. Consistent with improved hippocampus-dependent context learning and memory, 6 weeks of wheel running also improved context discrimination and reduced the context pre-exposure time required to form a strong contextual memory. The effect of wheel running on brain-derived neurotrophic factor (BDNF) messenger ribonucleic acid (mRNA) in hippocampal and amygdala subregions was also investigated. Wheel running increased BDNF mRNA in the dentate gyrus, CA1, and the basolateral amygdala. Results are consistent with improved hippocampal function following physical activity.
Collapse
Affiliation(s)
- Benjamin N Greenwood
- Department of Integrative Physiology and the Center for Neuroscience, University of Colorado, Boulder, CO 80309-0354, USA.
| | | | | | | |
Collapse
|
39
|
Strong PV, Greenwood BN, Fleshner M. The effects of the selective 5-HT(2C) receptor antagonist SB 242084 on learned helplessness in male Fischer 344 rats. Psychopharmacology (Berl) 2009; 203:665-75. [PMID: 19037632 DOI: 10.1007/s00213-008-1413-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 11/03/2008] [Indexed: 12/21/2022]
Abstract
RATIONALE Rats exposed to an uncontrollable stressor demonstrate a constellation of behaviors such as exaggerated freezing and deficits in shuttle box escape learning. These behaviors in rats have been called learned helplessness and have been argued to model human stress-related mood disorders. Learned helplessness is thought to be caused by hyperactivation of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) and a subsequent exaggerated release of 5-HT in DRN projection sites. Blocking 5-HT(2C) receptors in the face of an increase in serotonin can alleviate anxiety behaviors in some animal models. However, specific 5-HT receptor subtypes involved in learned helplessness remain unknown. OBJECTIVES The current experiments tested the hypothesis that 5-HT(2C) receptor activation is necessary and sufficient for the expression of learned helplessness. RESULTS The selective 5-HT(2C) receptor antagonist SB 242084 (1.0 mg/kg) administered i.p. to adult male Fischer 344 rats prior to shuttle box behavioral testing, but not before stress, blocked stress-induced deficits in escape learning but had no effect on the exaggerated shock-elicited freezing. The selective 5-HT(2C) receptor agonist CP-809101 was sufficient to produce learned helplessness-like behaviors in the absence of prior stress and these effects were blocked by pretreatment with SB 242084. CONCLUSIONS Results implicate the 5-HT(2C) receptor subtype in mediating the shuttle box escape deficits produced by exposure to uncontrollable stress and suggest that different postsynaptic 5-HT receptor subtypes underlie the different learned helplessness behaviors.
Collapse
Affiliation(s)
- Paul V Strong
- Department of Integrative Physiology and the Center for Neuroscience, University of Colorado, Clare Small Room 104, Campus Box 354, Boulder, CO 80309, USA.
| | | | | |
Collapse
|
40
|
Stress impairs 5-HT2A receptor-mediated serotonergic facilitation of GABA release in juvenile rat basolateral amygdala. Neuropsychopharmacology 2009; 34:410-23. [PMID: 18536707 DOI: 10.1038/npp.2008.71] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The occurrence of stress and anxiety disorders has been closely associated with alterations of the amygdala GABAergic system. In these disorders, dysregulation of the serotonergic system, a very important modulator of the amygdala GABAergic system, is also well recognized. The present study, utilizing a learned helplessness stress rat model, was designed to determine whether stress is capable of altering serotonergic modulation of the amygdala GABAergic system. In control rats, administration of 5-HT or alpha-methyl-5-HT, a 5-HT(2) receptor agonist, to basolateral amygdala (BLA) slices dramatically enhanced frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs). This effect was blocked by selective 5-HT(2A) receptor antagonists while a selective 5-HT(2B) receptor agonist and a selective 5-HT(2C) receptor agonist were without effect on sIPSCs. Double immunofluorescence labeling demonstrated that the 5-HT(2A) receptor is primarily localized to parvalbumin-containing BLA interneurons. Thus, serotonin primarily acts via 5-HT(2A) receptors to facilitate BLA GABAergic inhibition. In stressed rats, the 5-HT(2A) receptor-mediated facilitative actions were severely impaired. Quantitative RT-PCR and western blot analysis showed that the impairment of 5-HT(2A) receptor signaling primarily resulted from receptor downregulation. The stress-induced effect appeared to be specific to 5-HT(2A) receptors because stress had no significant impact on other serotonin receptors, as well as histamine H(3) receptor and alpha(2) adrenoceptor signaling in the BLA. This severe impairment of 5-HT(2A) receptor-mediated facilitation of BLA GABAergic inhibition might result in an amygdala circuitry with hyperexcitability, and a lower threshold of activation, and thus be an important mechanism underlying the emergence of stress-associated psychiatric symptoms.
Collapse
|
41
|
Caldwell KK, Sheema S, Paz RD, Samudio-Ruiz SL, Laughlin MH, Spence NE, Roehlk MJ, Alcon SN, Allan AM. Fetal alcohol spectrum disorder-associated depression: evidence for reductions in the levels of brain-derived neurotrophic factor in a mouse model. Pharmacol Biochem Behav 2008; 90:614-24. [PMID: 18558427 DOI: 10.1016/j.pbb.2008.05.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 05/05/2008] [Accepted: 05/09/2008] [Indexed: 01/06/2023]
Abstract
Prenatal ethanol exposure is associated with an increased incidence of depressive disorders in patient populations. However, the mechanisms that link prenatal ethanol exposure and depression are unknown. Several recent studies have implicated reduced brain-derived neurotrophic factor (BDNF) levels in the hippocampal formation and frontal cortex as important contributors to the etiology of depression. In the present studies, we sought to determine whether prenatal ethanol exposure is associated with behaviors that model depression, as well as with reduced BDNF levels in the hippocampal formation and/or medial frontal cortex, in a mouse model of fetal alcohol spectrum disorder (FASD). Compared to control adult mice, prenatal ethanol-exposed adult mice displayed increased learned helplessness behavior and increased immobility in the Porsolt forced swim test. Prenatal ethanol exposure was associated with decreased BDNF protein levels in the medial frontal cortex, but not the hippocampal formation, while total BDNF mRNA and BDNF transcripts containing exons III, IV or VI were reduced in both the medial frontal cortex and the hippocampal formation of prenatal ethanol-exposed mice. These results identify reduced BDNF levels in the medial frontal cortex and hippocampal formation as potential mediators of depressive disorders associated with FASD.
Collapse
Affiliation(s)
- Kevin K Caldwell
- Department of Neurosciences, MSC 08 4740, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Greenwood BN, Strong PV, Brooks L, Fleshner M. Anxiety-like behaviors produced by acute fluoxetine administration in male Fischer 344 rats are prevented by prior exercise. Psychopharmacology (Berl) 2008; 199:209-22. [PMID: 18454279 PMCID: PMC4356177 DOI: 10.1007/s00213-008-1167-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 04/08/2008] [Indexed: 11/26/2022]
Abstract
RATIONALE Although selective 5-HT reuptake inhibitors (SSRIs) can reduce anxiety after chronic treatment, acute SSRI administration is associated with an increase in anxiety consistent with an acute increase in 5-HT neurotransmission. Exercise is anxiolytic in humans, and wheel running prevents anxiety-like behavioral consequences of uncontrollable stress in rats, but the effects of exercise on acute fluoxetine-induced anxiety-like behaviors are unknown. OBJECTIVES The current studies tested the hypothesis that acute administration of the SSRI fluoxetine would produce behaviors in rats resembling those produced by uncontrollable stress and that these behaviors would be blocked by prior wheel running. RESULTS Adult, male Fisher 344 rats administered moderate (10 mg/kg) or high (20 mg/kg) doses of fluoxetine demonstrated exaggerated shock-elicited freezing and an interference with shuttle box escape compared to rats given either saline or low-dose fluoxetine (2.5 mg/kg). Fluoxetine-induced behaviors were similar to, but smaller in magnitude than, those produced by uncontrollable stress and were blocked by pretreatment with the 5-HT(2C) receptor antagonist SB 242084 (1 mg/kg). Rats allowed access to running wheels for 6 weeks were protected against the anxiety-like behaviors produced by a single injection of fluoxetine (10 mg/kg). CONCLUSIONS Behavioral effects of acute fluoxetine administration resemble those produced by uncontrollable stress. Results are consistent with the idea that exercise can produce resistance against the anxiogenic effects of acute increases in 5-HT and suggest that acute behavioral effects of antidepressants can depend on history of physical activity.
Collapse
Affiliation(s)
- Benjamin N Greenwood
- Department of Integrative Physiology and the Center for Neuroscience, University of Colorado-Boulder, Boulder, CO 80309, USA.
| | | | | | | |
Collapse
|
43
|
Exercise, learned helplessness, and the stress-resistant brain. Neuromolecular Med 2008; 10:81-98. [PMID: 18300002 DOI: 10.1007/s12017-008-8029-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 01/16/2008] [Indexed: 02/02/2023]
Abstract
Exercise can prevent the development of stress-related mood disorders, such as depression and anxiety. The underlying neurobiological mechanisms of this effect, however, remain unknown. Recently, researchers have used animal models to begin to elucidate the potential mechanisms underlying the protective effects of physical activity. Using the behavioral consequences of uncontrollable stress or "learned helplessness" as an animal analog of depression- and anxiety-like behaviors in rats, we are investigating factors that could be important for the antidepressant and anxiolytic properties of exercise (i.e., wheel running). The current review focuses on the following: (1) the effect of exercise on the behavioral consequences of uncontrollable stress and the implications of these effects on the specificity of the "learned helplessness" animal model; (2) the neurocircuitry of learned helplessness and the role of serotonin; and (3) exercise-associated neural adaptations and neural plasticity that may contribute to the stress-resistant brain. Identifying the mechanisms by which exercise prevents learned helplessness could shed light on the complex neurobiology of depression and anxiety and potentially lead to novel strategies for the prevention of stress-related mood disorders.
Collapse
|
44
|
Duman CH, Schlesinger L, Russell DS, Duman RS. Voluntary exercise produces antidepressant and anxiolytic behavioral effects in mice. Brain Res 2008; 1199:148-58. [PMID: 18267317 DOI: 10.1016/j.brainres.2007.12.047] [Citation(s) in RCA: 300] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 12/18/2007] [Accepted: 12/20/2007] [Indexed: 01/18/2023]
Abstract
Reports of beneficial effects of exercise on psychological health in humans are increasingly supported by basic research studies. Exercise is hypothesized to regulate antidepressant-related mechanisms and we therefore characterized the effects of chronic exercise in mouse behavioral paradigms relevant to antidepressant actions. Mice given free access to running wheels showed antidepressant-like behavior in learned helplessness, forced-swim (FST) and tail suspension paradigms. These responses were similar to responses of antidepressant drug-treated animals. When tested under conditions where locomotor activity was not altered, exercising mice also showed reduced anxiety compared to sedentary control mice. In situ hybridization analysis showed that BDNF mRNA was increased in specific subfields of hippocampus after wheel running. We chose one paradigm, the FST, in which to investigate a functional role for brain-derived neurotrophic factor (BDNF) in the behavioral response to exercise. We tested mice heterozygous for a deletion of the BDNF gene in the FST after wheel-running. Exercising wild-type mice showed the expected antidepressant-like behavioral response in the FST but exercise was ineffective in improving FST performance in heterozygous BDNF knockout mice. A possible functional contribution of a BDNF signaling pathway to FST performance in exercising mice was investigated using the specific MEK inhibitor PD184161 to block the MAPK signaling pathway. Subchronic administration of PD184161 to exercising mice blocked the antidepressant-like behavioral response seen in vehicle-treated exercising mice in the FST. In summary, chronic wheel-running exercise in mice results in antidepressant-like behavioral changes that may involve a BDNF related mechanism similar to that hypothesized for antidepressant drug treatment.
Collapse
Affiliation(s)
- Catharine H Duman
- Division of Molecular Psychiatry, Abraham Ribicoff Research Facilities, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, CT 06508, USA.
| | | | | | | |
Collapse
|
45
|
Schmidt HD, Duman RS. The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behavior. Behav Pharmacol 2007; 18:391-418. [PMID: 17762509 DOI: 10.1097/fbp.0b013e3282ee2aa8] [Citation(s) in RCA: 494] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Major depressive disorder (MDD) is characterized by structural and neurochemical changes in limbic structures, including the hippocampus, that regulate mood and cognitive functions. Hippocampal atrophy is observed in patients with depression and this effect is blocked or reversed by antidepressant treatments. Brain-derived neurotrophic factor and other neurotrophic/growth factors are decreased in postmortem hippocampal tissue from suicide victims, which suggests that altered trophic support could contribute to the pathophysiology of MDD. Preclinical studies demonstrate that exposure to stress leads to atrophy and cell loss in the hippocampus as well as decreased expression of neurotrophic/growth factors, and that antidepressant administration reverses or blocks the effects of stress. Accumulating evidence suggests that altered neurogenesis in the adult hippocampus mediates the action of antidepressants. Chronic antidepressant administration upregulates neurogenesis in the adult hippocampus and this cellular response is required for the effects of antidepressants in certain animal models of depression. Here, we review cellular (e.g. adult neurogenesis) and behavioral studies that support the neurotrophic/neurogenic hypothesis of depression and antidepressant action. Aberrant regulation of neuronal plasticity, including neurogenesis, in the hippocampus and other limbic nuclei may result in maladaptive changes in neural networks that underlie the pathophysiology of MDD.
Collapse
Affiliation(s)
- Heath D Schmidt
- Division of Molecular Psychiatry, Abraham Ribicoff Research Facilities, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|