451
|
Gardier AM, Guiard BP, Guilloux JP, Repérant C, Coudoré F, David DJ. Interest of using genetically manipulated mice as models of depression to evaluate antidepressant drugs activity: a review. Fundam Clin Pharmacol 2009; 23:23-42. [PMID: 19267769 DOI: 10.1111/j.1472-8206.2008.00640.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Among the multiple possibilities to study human depressive disorders, animal models remain important preclinical tools. They allow the understanding of the mechanisms of action of antidepressant drugs. Primarily developed in rat, animal models of depression have been adapted to the mouse, an easy-to-use mammal with better genetic possibilities than rats. As an example, genetic manipulation of the serotoninergic 5-hydroxytryptamine-HT; (5-HT) system provided important opportunities to investigate the role of this monoamine in mood disorders. The contribution of either constitutive knockout (KO), tissue specific, or inducible KO mice and animal models in the current knowledge of the pathophysiology and treatment of depression is unanimously recognized. The phenotype of genetically manipulated animals is strongly influenced by both the genetic background of the animal as well as environmental factors. For these reasons, it is necessary to underline that KO mice have been generated on various genetic backgrounds, which strongly influence the behavioral and neurochemical responses to the tests. The present review will thus focus on KO mice lacking G protein-coupled monoaminergic receptors (e.g; 5-HT1B, 5-HT1A, and 5-HT4 receptors) and the 5-HT serotonin transporter, which is the main target of antidepressant drugs (or strategies). The importance of KO mice for neurotrophic factors, particularly for brain-derived neurotrophic factor and its main receptor displaying a tyrosine kinase activity, will also be addressed to illustrate the fact that in preclinical studies, combination of genetic manipulations with pharmacological ones should allow further progress in the field of neuropsychopharmacology.
Collapse
Affiliation(s)
- Alain M Gardier
- Fac. Pharmacie, Univ. Paris Sud, EA 3544, Chatenay-Malabry Cedex F-92296, France.
| | | | | | | | | | | |
Collapse
|
452
|
Enhanced sensitivity of the MRL/MpJ mouse to the neuroplastic and behavioral effects of chronic antidepressant treatments. Neuropsychopharmacology 2009; 34:1764-73. [PMID: 19177066 PMCID: PMC2680932 DOI: 10.1038/npp.2008.234] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chronic administration of antidepressant drugs produce changes in neuroplasticity and behavior in rodents, effects that may be associated with the slow emergence of clinical therapeutic effects. Owing to the uncertainty over the effects of chronic antidepressant treatments in mice, these experiments compared the regulation of neurogenesis, neurotrophin levels, and behavior produced by chronic antidepressant treatments between two inbred mouse strains, MRL/MpJ and C57BL/6J. The MRL/MpJ strain is associated with enhanced wound healing and tissue regeneration, whereas C57BL/6J mice are used commonly for behavioral studies. Proliferation and survival of hippocampal progenitor cells were measured using flow cytometry, a new platform that rapidly quantifies the incorporation of 5-bromo-2-deoxyuridine (BrdU). Hippocampal cell proliferation was increased significantly after chronic administration of fluoxetine (FLX: 5, 10 mg/kg, intraperitoneal (i.p.), b.i.d.) or desipramine (DMI: 5, 10 mg/kg, i.p., b.i.d.) for 21 days in MRL/MpJ mice, but not in C57BL/6J mice. Hippocampal progenitor cells born prior to chronic antidepressant treatments were not affected in either mouse strain. Protein levels of brain-derived neurotrophic factor (BDNF) in MRL/MpJ mice were elevated significantly in the frontal cortex, hippocampus, and amygdala after chronic FLX treatment, but increased only in the frontal cortex by chronic DMI. In contrast, BDNF levels in C57BL/6J mice were decreased in the hippocampus and increased in the amygdala after chronic FLX, and were decreased in the brain stem after chronic DMI. Novelty-induced hypophagia (NIH) was used to examine a behavioral effect produced by chronic antidepressant treatment. MRL/MpJ mice, chronically administered FLX or DMI, had significantly shorter latencies to consume food when exposed to a novel environment than untreated mice, whereas there were no effects on the behavior of C57BL/6J mice. In conclusion, robust effects of chronic antidepressant treatments on hippocampal cell proliferation and BDNF levels paralleled the ability of these drugs to produce changes in NIH behavior in MRL/MpJ, while none of these effects were produced in C57BL/6J mice. The greater responsiveness of MRL/MpJ mice may be important for drug discovery, for genetic studies, and for understanding the neural mechanisms underlying the physiological and behavioral effects of chronic antidepressant treatments.
Collapse
|
453
|
TRH-receptor-type-2-deficient mice are euthyroid and exhibit increased depression and reduced anxiety phenotypes. Neuropsychopharmacology 2009; 34:1601-8. [PMID: 19078951 PMCID: PMC2669701 DOI: 10.1038/npp.2008.217] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Thyrotropin-releasing hormone (TRH) is a neuropeptide that initiates its effects in mice by interacting with two G-protein-coupled receptors, TRH receptor type 1 (TRH-R1) and TRH receptor type 2 (TRH-R2). Two previous reports described the effects of deleting TRH-R1 in mice. TRH-R1 knockout mice exhibit hypothyroidism, hyperglycemia, and increased depression and anxiety-like behavior. Here we report the generation of TRH-R2 knockout mice. The phenotype of these mice was characterized using gross and histological analyses along with blood hematological assays and chemistries. Standard metabolic tests to assess glucose and insulin tolerance were performed. Behavioral testing included elevated plus maze, open field, tail suspension, forced swim, and novelty-induced hypophagia tests. TRH-R2 knockout mice are euthyroid with normal basal and TRH-stimulated serum levels of thyroid-stimulating hormone (thyrotropin), are normoglycemic, and exhibit normal development and growth. Female, but not male, TRH-R2 knockout mice exhibit moderately increased depression-like and reduced anxiety-like phenotypes. Because the behavioral changes in TRH-R1 knockout mice may have been caused secondarily by their hypothyroidism whereas TRH-R2 knockout mice are euthyroid, these data provide the first evidence for the involvement of the TRH/TRH-R system, specifically extrahypothalamic TRH/TRH-R2, in regulating mood and affect.
Collapse
|
454
|
Gundersen BB, Blendy JA. Effects of the histone deacetylase inhibitor sodium butyrate in models of depression and anxiety. Neuropharmacology 2009; 57:67-74. [PMID: 19393671 DOI: 10.1016/j.neuropharm.2009.04.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 04/02/2009] [Accepted: 04/15/2009] [Indexed: 11/27/2022]
Abstract
Histone modification, which affects the rate of transcription without altering DNA sequence, occurs in response to various psychiatric drugs and in several models of psychiatric disease. As increases in histone acetylation have been seen after treatment with antidepressants, we investigated whether directly increasing histone acetylation using a histone deacetylase inhibitor would have antidepressant effects. We administered sodium butyrate (NaB, 100 mg/kg, i.p.) to mice acutely (3 injections over 24 h) or chronically (twice daily for 21 days) and subjected them to a number of behavioral tests of antidepressant response. This dose of NaB had no effect on overall locomotor activity after either acute or chronic treatment. Acutely treated mice showed an increase in immobility in the forced-swim test (FST) and an increase in latency to consume in the novel environment of the novelty-induced hypophagia (NIH) paradigm, an anxiogenic effect. The effect of NaB on anxiety did not generalize to another test, the elevated zero maze, where it had no effect. Chronic treatment with NaB had no effect on latency to consume in the NIH or immobility in the FST. However, this dose did alter histone acetylation in the hippocampus. While H4 acetylation increased in the hippocampus 30 min following acute NaB, chronic treatment caused a decrease in AcH4. There were no changes in AcH3 following either treatment. While changes in chromatin structure may be involved in the mechanism of action of antidepressant drugs, these data suggest that increasing histone acetylation pharmacologically is not sufficient to produce antidepressant effects.
Collapse
Affiliation(s)
- Brigitta B Gundersen
- Department of Pharmacology, University of Pennysylvania, School of Medicine, Translational Research Labs, 125 S. 31st St, Philadelphia, PA 19104, USA
| | | |
Collapse
|
455
|
Mutant mouse models and antidepressant drug research: focus on serotonin and brain-derived neurotrophic factor. Behav Pharmacol 2009; 20:18-32. [PMID: 19179848 DOI: 10.1097/fbp.0b013e3283243fcd] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Several lines of knockout (KO) mice have been evaluated as models of depression-related behavioral and neurobiological changes, and used to investigate molecular and cellular mechanisms underlying the activity of antidepressant drugs. Adult neurogenesis and brain 5-hydroxytryptamine (5-HT)/neurotrophic factor interactions have recently attracted great interest in relation to the mechanism of action of antidepressant drugs. The present review focuses primarily on genetic manipulation of the serotoninergic (5-HT) system. Basal neurochemical and behavioral changes occurring in mice lacking the 5-HT transporter (SERT), which is the main target of antidepressant drugs, as well as in those lacking G protein-coupled serotonin receptors (e.g. 5-HT1B, 5-HT1A, and 5-HT4 receptors) are described and evaluated. The importance of KO mice for neurotrophic factors, particularly for brain-derived neurotrophic factor and its high-affinity receptor (R-TrkB), is also addressed. Constitutive KO, tissue specific, or inducible KO mice targeting both 5-HT and brain-derived neurotrophic factor systems may potentially make an important contribution to knowledge of the pathophysiology and treatment of depression.
Collapse
|
456
|
Shanahan NA, Holick KA, Masten VL, Waeber C, Ansorge M, Gingrich JA, Geyer MA, Hen R, Dulawa SC. Chronic reductions in serotonin transporter function prevent 5-HT1B-induced behavioral effects in mice. Biol Psychiatry 2009; 65:401-8. [PMID: 19013555 PMCID: PMC2674010 DOI: 10.1016/j.biopsych.2008.09.026] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 08/26/2008] [Accepted: 09/16/2008] [Indexed: 11/28/2022]
Abstract
BACKGROUND Obsessive-compulsive disorder (OCD) is characterized by intrusive thoughts, images, or impulses and/or repetitive stereotypical behavior. Obsessive-compulsive disorder patients exhibit reduced prepulse inhibition (PPI) and symptom exacerbation after challenge with 5-HT1B receptor agonists. Recently, gain-of-function alleles of the serotonin transporter (5-HTT) have been associated with OCD. We tested the hypothesis that reducing 5-HTT function chronically, either genetically or via serotonin reuptake inhibitor (SRI) treatment, attenuates PPI deficits and perseverative hyperlocomotion induced by 5-HT1B agonists in mice. METHODS Mice received subchronic or chronic pretreatment with the SRI fluoxetine and acute treatment with RU24969 (5-HT1A/1B agonist) or 8-OH-DPAT (5-HT1A agonist) and were assessed for PPI, locomotor activity, and spatial patterns of locomotion. The same measures were evaluated in 5-HTT wild-type (WT), heterozygous (HT), and knockout (KO) mice after RU24969 treatment. The effects of WAY100635 (5-HTA antagonist) or GR127935 (5-HT1B/D antagonist) pretreatment on RU24969-induced effects were evaluated. Finally, 5-HT1B binding and functional coupling were assessed in 5-HTT-WT, -HT, and -KO mice, and normal fluoxetine-treated mice. RESULTS Chronic, but not subchronic, fluoxetine treatment prevented RU24969-induced PPI deficits and perseverative hyperlocomotion. These RU24969-induced effects were mediated via 5-HT1B and not 5-HT1A receptors. 5-HTT-KO mice showed no effects of RU24969, and 5-HTT-HT mice exhibited intermediate phenotypes. 5-HT1B binding and functional coupling were reduced in the globus pallidus and substantia nigra of 5-HTT-KO mice. CONCLUSIONS Our results demonstrate that chronic, but not subchronic, fluoxetine treatment and 5-HTT knockout robustly attenuate 5-HT1B agonist-induced PPI deficits and perseverative hyperlocomotion. These results may have implications for the etiology and treatment of OCD.
Collapse
Affiliation(s)
- Nancy A. Shanahan
- Committee on Neurobiology, University of Chicago, Chicago, IL, 60637
| | - Kerri A. Holick
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Virginia L. Masten
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093
| | - Christian Waeber
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Mark Ansorge
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Jay A. Gingrich
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Mark A. Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093
| | - Rene Hen
- Department of Psychiatry, Columbia University, New York, NY 10032, Department of Pharmacology, Columbia University, New York, NY 10032, Center for Neurobiology and Behavior, Columbia University, New York, NY 10032
| | - Stephanie C. Dulawa
- Department of Psychiatry, University of Chicago, Chicago, IL, 60637, Committee on Neurobiology, University of Chicago, Chicago, IL, 60637,Correspondence should be addressed to: Dr. Stephanie Dulawa, Department of Psychiatry, 924 East 57 Street, Room R018, MC 3077, Chicago, IL 60637, Tel: 773-834-0241, Fax: 773-834-2970,
| |
Collapse
|
457
|
Fluoxetine directly counteracts the adverse effects of chronic stress on T cell immunity by compensatory and specific mechanisms. Brain Behav Immun 2009; 23:36-40. [PMID: 18625298 DOI: 10.1016/j.bbi.2008.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 06/12/2008] [Accepted: 06/20/2008] [Indexed: 11/22/2022] Open
Abstract
Chronic stress and depression are widely known to down-regulate the immune system, and several antidepressants can reverse this impairment, with or without effects in normal subjects. Although the central nervous system is undoubtedly involved in these events, some psychotropic drugs can also exert direct effects on lymphoid cells. We have recently shown that the antidepressant fluoxetine enhances T cell proliferation and T(H)1 cytokine production in vivo, without changes on CD4/CD8 subsets. In vitro, a direct action of fluoxetine upon T lymphocyte reactivity by complex mechanisms was also described. In another work, we also found that chronic stress reduces T cell mediated immunity, namely a decrease of T cell response to mitogens, T(H)1 cytokine production and CD4+-but not CD8+--T lymphocytes. Here we investigated the effects of fluoxetine on chronic stress-driven immune system depression. We found that fluoxetine restored T cell proliferation and interleukin-2, interferon-gamma and tumor necrosis factor-alpha production by compensatory mechanisms. In addition, CD4/CD8 ratio was also normalized by antidepressant administration, but this seems to be a non-compensatory effect associated specifically to stress. No changes were observed in other lymphoid cells, i.e. natural killer cells and B lymphocytes. Finally, we observed that fluoxetine is able to reverse T cell reactivity impairment in vitro by a direct action at clinically relevant doses. These results highlight the relevance of pharmacological treatment of stress and depression, and may help to begin elucidating the complex events triggered--directly and/or indirectly--by antidepressants in non-neuronal cell types.
Collapse
|
458
|
Gould TD, Dao DT, Kovacsics CE. The Open Field Test. MOOD AND ANXIETY RELATED PHENOTYPES IN MICE 2009. [DOI: 10.1007/978-1-60761-303-9_1] [Citation(s) in RCA: 191] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
459
|
Hascoët M, Bourin M. The Forced Swimming Test in Mice: A Suitable Model to Study Antidepressants. MOOD AND ANXIETY RELATED PHENOTYPES IN MICE 2009. [DOI: 10.1007/978-1-60761-303-9_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
460
|
Animal models of depression in dopamine, serotonin, and norepinephrine transporter knockout mice: prominent effects of dopamine transporter deletions. Behav Pharmacol 2008; 19:566-74. [PMID: 18690111 DOI: 10.1097/fbp.0b013e32830cd80f] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Antidepressant drugs produce therapeutic actions and many of their side effects via blockade of the plasma membrane transporters for serotonin (SERT/SLC6A2), norepinephrine (NET/SLC6A1), and dopamine (DAT/SLC6A3). Many antidepressants block several of these transporters; some are more selective. Mouse gene knockouts of these transporters provide interesting models for possible effects of chronic antidepressant treatments. To examine the role of monoamine transporters in models of depression DAT, NET, and SERT knockout (KO) mice and wild-type littermates were studied in the forced swim test (FST), the tail suspension test, and for sucrose consumption. To dissociate general activity from potential antidepressant effects three types of behavior were assessed in the FST: immobility, climbing, and swimming. In confirmation of earlier reports, both DAT KO and NET KO mice exhibited less immobility than wild-type littermates whereas SERT KO mice did not. Effects of DAT deletion were not simply because of hyperactivity, as decreased immobility was observed in DAT+/- mice that were not hyperactive as well as in DAT-/- mice that displayed profound hyperactivity. Climbing was increased, whereas swimming was almost eliminated in DAT-/- mice, and a modest but similar effect was seen in NET KO mice, which showed a modest decrease in locomotor activity. Combined increases in climbing and decreases in immobility are characteristic of FST results in antidepressant animal models, whereas selective effects on swimming are associated with the effects of stimulant drugs. Therefore, an effect on climbing is thought to more specifically reflect antidepressant effects, as has been observed in several other proposed animal models of reduced depressive phenotypes. A similar profile was observed in the tail suspension test, where DAT, NET, and SERT knockouts were all found to reduce immobility, but much greater effects were observed in DAT KO mice. However, to further determine whether these effects of DAT KO in animal models of depression may be because of the confounding effects of hyperactivity, mice were also assessed in a sucrose consumption test. Sucrose consumption was increased in DAT KO mice consistent with reduced anhedonia, and inconsistent with competitive hyperactivity; no increases were observed in SERT KO or NET KO mice. In summary, the effects of DAT KO in animal models of depression are larger than those produced by NET or SERT KO, and unlikely to be simply the result of the confounding effects of locomotor hyperactivity; thus, these data support reevaluation of the role that DAT expression could play in depression and the potential antidepressant effects of DAT blockade.
Collapse
|
461
|
Serotonin (5-hydroxytryptamine) transporter inhibition causes bone loss in adult mice independently of estrogen deficiency. Menopause 2008; 15:1176-83. [DOI: 10.1097/gme.0b013e318173566b] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
462
|
Duman CH, Schlesinger L, Terwilliger R, Russell DS, Newton SS, Duman RS. Peripheral insulin-like growth factor-I produces antidepressant-like behavior and contributes to the effect of exercise. Behav Brain Res 2008; 198:366-71. [PMID: 19056428 DOI: 10.1016/j.bbr.2008.11.016] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 08/11/2008] [Accepted: 11/10/2008] [Indexed: 11/30/2022]
Abstract
Growth factors in the brain are important to depression and it's treatment and we assessed the ability of peripherally administered insulin-like growth factor-I (IGF-I) to influence behavior related to depression. We found that mice that received chronic IGF-I treatment showed antidepressant-like behavior in forced-swim and novelty-induced hypophagia (NIH) tests and increased sucrose consumption after chronic mild unpredictable stress exposure. Additionally, peripheral anti-IGF-I administration blocked exercise-induced antidepressant effects in the forced-swim test (FST). These results support the functional relevance of neurotrophic mechanisms to depression and extend this idea to include neurotrophic factors in the periphery.
Collapse
Affiliation(s)
- Catharine H Duman
- Division of Molecular Psychiatry, Abraham Ribicoff Research Facilities, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA.
| | | | | | | | | | | |
Collapse
|
463
|
Abstract
Mast cells are resident in the brain and contain numerous mediators, including neurotransmitters, cytokines, and chemokines, that are released in response to a variety of natural and pharmacological triggers. The number of mast cells in the brain fluctuates with stress and various behavioral and endocrine states. These properties suggest that mast cells are poised to influence neural systems underlying behavior. Using genetic and pharmacological loss-of-function models we performed a behavioral screen for arousal responses including emotionality, locomotor, and sensory components. We found that mast cell deficient Kit(W-sh/W-sh) (sash(-/-)) mice had a greater anxiety-like phenotype than WT and heterozygote littermate control animals in the open field arena and elevated plus maze. Second, we show that blockade of brain, but not peripheral, mast cell activation increased anxiety-like behavior. Taken together, the data implicate brain mast cells in the modulation of anxiety-like behavior and provide evidence for the behavioral importance of neuroimmune links.
Collapse
|
464
|
Pollak DD, Monje FJ, Zuckerman L, Denny CA, Drew MR, Kandel ER. An animal model of a behavioral intervention for depression. Neuron 2008; 60:149-61. [PMID: 18940595 PMCID: PMC3417703 DOI: 10.1016/j.neuron.2008.07.041] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 08/26/2008] [Accepted: 07/30/2008] [Indexed: 11/25/2022]
Abstract
Although conditioned inhibition of fear (or learned safety) is a learning process critical for preventing chronic stress, a predisposing factor for depression and other psychopathologies, little is known about its functional purposes or molecular mechanisms. To obtain better insight into learned safety, we investigated its behavioral and molecular characteristics and found that it acts as a behavioral antidepressant in two animal models. Learned safety promotes the survival of newborn cells in the dentate gyrus of the hippocampus, while its antidepressant effect is abolished in mice with ablated hippocampal neurogenesis. Learned safety also increases the expression of BDNF in the hippocampus and leads to downregulation of genes involved in the dopaminergic and neuropeptidergic but not the serotonergic system in the basolateral amygdala. These data suggest that learned safety is an animal model of a behavioral antidepressant that shares some neuronal hallmarks of pharmacological antidepressants but is mediated by different molecular pathways.
Collapse
Affiliation(s)
- Daniela D Pollak
- Department of Neuroscience, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
465
|
Norcross M, Poonam M, Enoch AJ, Karlsson RM, Brigman JL, Cameron HA, Harvey-White J, Holmes A, Holmes A. Effects of adolescent fluoxetine treatment on fear-, anxiety- or stress-related behaviors in C57BL/6J or BALB/cJ mice. Psychopharmacology (Berl) 2008; 200:413-24. [PMID: 18594797 PMCID: PMC2574726 DOI: 10.1007/s00213-008-1215-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2007] [Accepted: 05/28/2008] [Indexed: 11/27/2022]
Abstract
RATIONALE 5-Hydroxytryptamine (5-HT, serotonin) plays a major role in brain ontogeny. Disruption of 5-HT during early postnatal development produces lasting changes in rodent 'emotion-related' behaviors. Adverse effects of treatment with serotonin reuptake inhibitor (SRI) antidepressants have been reported in human adolescents. However, the long-term effects of chronic SRI treatment during adolescence in rodents remain unclear. OBJECTIVES The objectives of the study are to assess the effects of fluoxetine treatment throughout the adolescent period in measures of fear-, anxiety- and stress-related endpoints in drug-free adults and to examine these effects in two genetic strains of mice differing in baseline stress- and anxiety-related behaviors and sensitivity to SRIs. MATERIALS AND METHODS C57BL/6J and BALB/cJ mice received one of two fluoxetine doses for 4 weeks during adolescence (3-7 weeks old). A separate group of C57BL/6J and BALB/cJ mice received fluoxetine for 4 weeks during adulthood (8-12 weeks old). After a 3-week washout period, mice were tested for anxiety-like behaviors (novel open field, elevated plus-maze), fear conditioning and extinction, and stress-related responses to forced swim, as well as serotonin brain levels. RESULTS Adolescent fluoxetine treatment did not increase adult measures of anxiety-, fear- or stress-related behaviors, or brain serotonin levels. The same duration of treatment in adulthood also had no effects on these measures when tested after a 3-week washout period. CONCLUSIONS In clear contrast with emotion-related abnormalities caused by preadolescent fluoxetine treatment or genetic inactivation of fluoxetine's pharmacological target, the 5-HT transporter, fluoxetine treatment throughout mouse adolescence did not produce detectable, lasting abnormalities in either "high" or "low anxiety" inbred mouse strains.
Collapse
Affiliation(s)
- Maxine Norcross
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA
| | - Mathur Poonam
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA
| | - Abigail J. Enoch
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA
| | - Rose-Marie Karlsson
- Laboratory of Translational and Clinical Studies, Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jonathan L. Brigman
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA
| | - Heather A. Cameron
- Unit on Neuroplasticity, Mood and Anxiety Disorders Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Judith Harvey-White
- Laboratory for Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Rockville, MD, USA
| | - Andrew Holmes
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA, e-mail:
| | | |
Collapse
|
466
|
Navailles S, Hof PR, Schmauss C. Antidepressant drug-induced stimulation of mouse hippocampal neurogenesis is age-dependent and altered by early life stress. J Comp Neurol 2008; 509:372-81. [PMID: 18512685 DOI: 10.1002/cne.21775] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The continuous generation of new neurons in the adult hippocampus exhibits remarkable plasticity. Decreased neurogenesis is thought to underlie depression-like behaviors, and increased neurogenesis is thought to occur following antidepressant drug treatment. Studies on different strains of mice, however, yielded contrasting results with regard to the link between behavioral modifications induced by antidepressant drugs or environmental enrichment and changes in adult hippocampal neurogenesis. Therefore, we conducted a comparative study on the inbred strains Balb/c and C57Bl/6 that differ substantially in emotionality, stress reactivity, and behavioral responses to chronic antidepressant drugs. Quantitative assessments of progenitor cell proliferation and immature neuronal differentiation in the dentate gyrus revealed that, despite significantly different basal proliferation rates between both strains, neither strain exhibited changes in adult neurogenesis after exposure to early life stress or adult chronic fluoxetine treatment. A stimulatory effect of fluoxetine on adult hippocampal neurogenesis was only detected when treatment was initiated during adolescence, and this effect was abolished in mice exposed to early life stress, a prominent risk factor for developing adult-onset depression-like behaviors. Thus, in both strains of mice neither adult fluoxetine treatment nor adolescent fluoxetine treatment following early life stress exposure increased the proliferation and early differentiation of adult neural progenitor cells.
Collapse
Affiliation(s)
- Sylvia Navailles
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, USA
| | | | | |
Collapse
|
467
|
Catts VS, Al-Menhali N, Burne THJ, Colditz MJ, Coulson EJ. The p75 neurotrophin receptor regulates hippocampal neurogenesis and related behaviours. Eur J Neurosci 2008; 28:883-92. [PMID: 18717734 DOI: 10.1111/j.1460-9568.2008.06390.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although changes to neural circuitry are believed to underlie behavioural characteristics mediated by the hippocampus, the contribution of neurogenesis to this process remains controversial. This is partially because the molecular regulators of neurogenesis remain to be fully elucidated, and experiments generically preventing neurogenesis have, for the most part, depended on paradigms involving irradiation. Here we show that mice lacking the p75 neurotrophin receptor (p75(NTR-/-)) have 25% fewer neuroblasts and 50% fewer newborn neurons in the dentate gyrus, coincident with increased rates of cell death of newly born cells and a significantly smaller granular cell layer and dentate gyrus, than those of p75(NTR+/+) mice. Whereas p75(NTR-/-) mice had increased latency to feed in a novelty-suppressed feeding paradigm they had increased mobility in another test of "depression", the tail-suspension test. p75(NTR-/-) mice also had subtle behavioural impairment in Morris water maze tasks compared to wild-type animals. No difference between genotypes was found in relation to anxiety or exploration behaviour based on the elevated-plus maze, light-dark, hole-board, T-maze or forced-swim tests. Overall, this study demonstrates that p75(NTR) is an important regulator of hippocampal neurogenesis, with concomitant effects on associated behaviours. However, the behavioural attributes of the p75(NTR-/-) mice may be better explained by altered circuitry driven by the loss of p75(NTR) in the basal forebrain, rather than direct changes to neurogenesis.
Collapse
Affiliation(s)
- Vibeke S Catts
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | | | | | | | | |
Collapse
|
468
|
Chen PJ, Hsieh CL, Su KP, Hou YC, Chiang HM, Lin IH, Sheen LY. The antidepressant effect of Gastrodia elata Bl. on the forced-swimming test in rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2008; 36:95-106. [PMID: 18306453 DOI: 10.1142/s0192415x08005618] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Depression is a common psychiatric disorder with a high morbidity and mortality rate. The pharmacotherapy used in clinic today is not suitable for all patients and causes certain side-effects. Thus, looking for alternative treatments with antidepressant effect and minimal side-effect is important. Gastrodia elata Bl. is a famous Chinese traditional medicine used for centuries. The aim of this study is to test the antidepressant effect of Gastrodia elata Bl. extract (GE) using forced-swimming test (FST). Sprague-Dawley rats were assigned to control, GE, and fluoxetine groups, treated with 10 ml/kg bw (body weight) water, 1 g/kg bw of GE, and 15 mg/kg bw of fluoxetine, respectively. Those samples were administered by gavage to rats 23.5, 4.5 and 1 hour prior to the test session of FST. After FST, the animals were sacrificed and their brains were collected for monoamines analysis. The results indicated that the duration of immobility was significantly decreased in GE group compared to the control (p < 0.05). The concentration of serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), and the ratio of 5-HIAA/5-HT in frontal cortex, amygdala, and hippocampus were not significantly different between GE and the control groups. However, administration of GE significantly increased the dopamine (DA) concentration (p < 0.05) and decreased the concentration of 3,4-dihydroxyphenylacetic acid (DOPAC) (p < 0.01) and DA turnover (p < 0.05) in striatum compared to the control. The results of this study show that Gastrodia elata Bl. extract modulates the turnover of DA in rats, and thus probably possesses antidepressant effect in rats.
Collapse
Affiliation(s)
- Pei-Ju Chen
- Graduate Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
469
|
Onishchenko N, Karpova N, Sabri F, Castrn E, Ceccatelli S. Long-lasting depression-like behavior and epigenetic changes of BDNF gene expression induced by perinatal exposure to methylmercury. J Neurochem 2008; 106:1378-87. [DOI: 10.1111/j.1471-4159.2008.05484.x] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
470
|
Bechtholt AJ, Valentino RJ, Lucki I. Overlapping and distinct brain regions associated with the anxiolytic effects of chlordiazepoxide and chronic fluoxetine. Neuropsychopharmacology 2008; 33:2117-30. [PMID: 17987061 DOI: 10.1038/sj.npp.1301616] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Little is known about the sites of action for the behavioral effects of chronic antidepressants. The novelty-induced hypophagia (NIH) test is one of few animal behavioral tests sensitive to acute benzodiazepines and chronic antidepressants. The goals of these experiments were to examine patterns of brain activation associated with the behavioral response to novelty and identify regions that could regulate the anxiolytic effects of acute benzodiazepine and chronic antidepressant treatments, measured using the NIH test. In the first experiment, rats were treated acutely with the anxiolytic, chlordiazepoxide (2.5 or 5 mg/kg, i.p.). In separate experiments, animals were implanted with osmotic minipumps delivering vehicle or fluoxetine (5 or 20 mg/kg per day s.c.) for 3 or 28 days. NIH was assessed by giving animals access to a familiar palatable food in a novel environment. Associated brain areas were identified using c-fos immunohistochemistry. NIH was mitigated by acute chlordiazepoxide and chronic fluoxetine. Both drugs reversed novelty-induced changes in c-fos expression in the lateral division of the posterolateral part of the bed nucleus of the stria terminalis (STLP), cingulate cortex (Cg), and dorsal field CA2 of the hippocampus (dCA2). Chronic fluoxetine additionally increased c-fos expression in the anterior nucleus accumbens (aAcb) and the piriform cortex (Pir). The effects of the drugs on c-fos expression in many regions correlated with anxiolytic efficacy. These findings identified brain regions where the effects of chronic antidepressants and benzodiazepines may converge to produce anxiolytic activity, as well as distinct sites of action for the two classes of drugs.
Collapse
Affiliation(s)
- Anita J Bechtholt
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403, USA
| | | | | |
Collapse
|
471
|
Guzzetti S, Calcagno E, Canetta A, Sacchetti G, Fracasso C, Caccia S, Cervo L, Invernizzi RW. Strain differences in paroxetine-induced reduction of immobility time in the forced swimming test in mice: role of serotonin. Eur J Pharmacol 2008; 594:117-24. [PMID: 18691569 DOI: 10.1016/j.ejphar.2008.07.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 07/11/2008] [Accepted: 07/21/2008] [Indexed: 11/16/2022]
Abstract
We studied the antidepressant-like effect of paroxetine in strains of mice carrying different isoforms of tryptophan hydroxylase-2 (TPH-2), the enzyme responsible for the synthesis of brain serotonin (5-HT). The effect of paroxetine alone and in combination with pharmacological treatments enhancing or lowering 5-HT synthesis or melatonin was assessed in the forced swimming test in mice carrying allelic variants of TPH-2 (1473C in C57BL/6 and 1473G in DBA/2 and BALB/c). Changes in brain 5-hydroxytryptophan (5-HTP) accumulation and melatonin levels were measured by high-performance liquid chromatography. Paroxetine (2.5 and 5 mg/kg) reduced immobility time in C57BL/6J and C57BL/6N mice but had no such effect in DBA/2J, DBA/2N and BALB/c mice, even at 10 mg/kg. Enhancing 5-HT synthesis with tryptophan reinstated the antidepressant-like effect of paroxetine in DBA/2J, DBA/2N and BALB/c mice whereas inhibition of 5-HT synthesis prevented the effect of paroxetine in C57BL/6N mice. The response to paroxetine was not associated with changes in locomotor activity, brain melatonin or brain levels of the drug measured at the end of the behavioral test. These results support the importance of 5-HT synthesis in the response to SSRIs and suggest that melatonin does not contribute to the ability of tryptophan to rescue the antidepressant-like effect of paroxetine.
Collapse
Affiliation(s)
- Sara Guzzetti
- Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Laboratory of Experimental Psychopharmacology, Via La Masa 19, 20156 Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
472
|
Chronic fluoxetine bidirectionally modulates potentiating effects of serotonin on the hippocampal mossy fiber synaptic transmission. J Neurosci 2008; 28:6272-80. [PMID: 18550770 DOI: 10.1523/jneurosci.1656-08.2008] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) have been used to treat various psychiatric disorders. Although the cellular mechanisms underlying amelioration of particular symptoms are mostly unknown, recent studies have shown critical importance of the dentate gyrus of the hippocampus in behavioral effects of SSRIs in rodents. Here, we show that serotonin potentiates synaptic transmission between mossy fibers, the sole output of the dentate granule cells, and CA3 pyramidal cells in mouse hippocampal slices. This potentiation is mediated by activation of 5-HT(4) receptors and intracellular cAMP elevation. A chronic treatment of mice with fluoxetine, a widely used SSRI, bidirectionally modulates the 5-HT-induced potentiation: Fluoxetine enhances the potentiation induced by lower concentrations of serotonin, while attenuates that by the higher concentration, which represents stabilization of synaptic 5-HT action. In contrast to the chronic treatment, an acute application of fluoxetine in slices induces a leftward shift in the dose-response curve of the 5-HT-induced potentiation. Thus, acute and chronic fluoxetine treatments have distinct effects on the serotonergic modulation of the mossy fiber synaptic transmission. Exposure of mice to novel environments induces increases in locomotor activity and hippocampal extracellular 5-HT levels. In mice chronically treated with fluoxetine, the novelty-induced hyperactivity is reduced without significant alterations in home cage activity and motor skills. Our results suggest that the chronic fluoxetine treatment can stabilize the serotonergic modulation of the central synaptic transmission, which may contribute to attenuation of hyperactive behaviors.
Collapse
|
473
|
Reinés A, Cereseto M, Ferrero A, Sifonios L, Podestá MF, Wikinski S. Maintenance treatment with fluoxetine is necessary to sustain normal levels of synaptic markers in an experimental model of depression: correlation with behavioral response. Neuropsychopharmacology 2008; 33:1896-908. [PMID: 17955054 DOI: 10.1038/sj.npp.1301596] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dysfunction of hippocampal plasticity has been proposed to play a critical role in the pathophysiology of depression. However, antidepressant drug effects on synaptic plasticity and cytoskeletal remodeling remain controversial. The aim of the present study was to evaluate in animals exposed to the learned helplessness (LH) paradigm, an accepted experimental model of depression, the effect of chronic treatment with fluoxetine (FLX) on synaptic and cytoskeletal proteins known to undergo plastic changes. Synaptophysin (SYN), postsynaptic density 95 (PSD-95), axon growth-associated protein 43 (GAP-43), and cytoskeletal proteins (intermediate neurofilaments and MAP-2) were studied in the hippocampus by immunohistochemistry. Whereas LH animals treated 21 days with saline (LH-S group) displayed diminished SYN and PSD-95 immunostainings in the CA3 but not in the DG, chronic treatment with FLX not only reversed the despaired behavior induced by exposure to LH paradigm, but also fully recovered SYN and PSD-95 labeling to control values. Similar results were obtained for the axonal remodeling marker GAP-43. FLX treatment did not modify either the decreased light neurofilament subunit (NFL) observed in the hippocampus of LH animals or any other cytoskeletal protein studied. When FLX treatment was withdrawn for 90 days in those LH-FLX animals in which reversion of despair had been observed at day 25, recurrence of despaired behavior was found accompanied by decreased SYN, PSD-95, and NFL labelings. Results indicate that the synapse remodeling induced by FLX in the CA3 region could underlie its behavioral efficacy despite the absence of cytoskeletal remodeling and that the stability of synaptic changes would depend on the continuous administration of the drug.
Collapse
Affiliation(s)
- Analía Reinés
- Instituto de Investigaciones Farmacológicas (ININFA), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
474
|
Miller BH, Schultz LE, Gulati A, Cameron MD, Pletcher MT. Genetic regulation of behavioral and neuronal responses to fluoxetine. Neuropsychopharmacology 2008; 33:1312-22. [PMID: 17609676 DOI: 10.1038/sj.npp.1301497] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Despite widespread use of antidepressants, the factors underlying the behavioral response to antidepressants are unknown. It has been shown that antidepressant treatment promotes the proliferation and survival of neurons in the adult hippocampus via enhanced serotonergic signaling, but it is unclear whether hippocampal neurogenesis is responsible for the behavioral response to antidepressants. Furthermore, a large subpopulation of patients fails to respond to antidepressant treatment due to presumed underlying genetic factors. In the present study, we have used the phenotypic and genotypic variability of inbred mouse strains to show that there is a genetic component to both the behavioral and neuronal effects of chronic fluoxetine treatment, and that this antidepressant induces an increase in hippocampal cell proliferation only in the strains that also show a positive behavioral response to treatment. Furthermore, the behavioral and neuronal responses are associated with an upregulation of genes known to promote neuronal proliferation and survival. These results suggest that inherent genetic predisposition to increased serotonin-induced neurogenesis may be a determinant of antidepressant efficacy.
Collapse
Affiliation(s)
- Brooke H Miller
- Department of Molecular Therapeutics, The Scripps Research Institute--Scripps Florida, Jupiter, FL 33458, USA
| | | | | | | | | |
Collapse
|
475
|
Tatapudy S, Bruening S, Gleason G, Toth M. Validation and use of a computer-assisted counting procedure to quantify BrdU-labeled proliferating cells in the early postnatal mouse hippocampus. J Neurosci Methods 2008; 172:173-7. [PMID: 18533271 DOI: 10.1016/j.jneumeth.2008.04.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 02/29/2008] [Accepted: 04/15/2008] [Indexed: 02/03/2023]
Abstract
The dentate gyrus is one of the few brain regions that show proliferation of neuronal precursors postnatally and in adult life. Proliferation in the dentate gyrus has been shown to be influenced by exercise, stress and drugs such as antidepressants. Traditionally, proliferation studies rely on the time consuming and subjective manual count of labeled cells. Here we adapted the Metamorph software to automatically count cells labeled in the S phase in the developing dentate gyrus of mice. The validity of the computer-assisted method was established by showing an outcome similar to that obtained with the established manual counting procedure. In addition, by using a genetically modified mouse line with increased proliferation, the ability of the computer-assisted method to detect changes in proliferation was demonstrated.
Collapse
Affiliation(s)
- Sonali Tatapudy
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021, USA
| | | | | | | |
Collapse
|
476
|
Holmes A. Genetic variation in cortico-amygdala serotonin function and risk for stress-related disease. Neurosci Biobehav Rev 2008; 32:1293-314. [PMID: 18439676 DOI: 10.1016/j.neubiorev.2008.03.006] [Citation(s) in RCA: 201] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 02/20/2008] [Indexed: 01/09/2023]
Abstract
The serotonin system is strongly implicated in the pathophysiology and therapeutic alleviation of stress-related disorders such as anxiety and depression. Serotonergic modulation of the acute response to stress and the adaptation to chronic stress is mediated by a myriad of molecules controlling serotonin neuron development (Pet-1), synthesis (tryptophan hydroxylase 1 and 2 isozymes), packaging (vesicular monoamine transporter 2), actions at presynaptic and postsynaptic receptors (5-HT1A, 5-HT1B, 5-HT2A, 5-HT2C, 5-HT3A, 5-HT4, 5-HT5A, 5-HT6, 5-HT7), reuptake (serotonin transporter), and degradation (monoamine oxidase A). A growing body of evidence from preclinical rodents models, and especially genetically modified mice and inbred mouse strains, has provided significant insight into how genetic variation in these molecules can affect the development and function of a key neural circuit between the dorsal raphe nucleus, medial prefrontal cortex and amygdala. By extension, such variation is hypothesized to have a major influence on individual differences in the stress response and risk for stress-related disease in humans. The current article provides an update on this rapidly evolving field of research.
Collapse
Affiliation(s)
- Andrew Holmes
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcoholism and Alcohol Abuse, NIH, 5625 Fishers Lane Room 2N09, Rockville, MD 20852-9411, USA.
| |
Collapse
|
477
|
Inhibition of serotonin but not norepinephrine transport during development produces delayed, persistent perturbations of emotional behaviors in mice. J Neurosci 2008; 28:199-207. [PMID: 18171937 DOI: 10.1523/jneurosci.3973-07.2008] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Serotonin (5-HT) acts as a neurotransmitter, but also modulates brain maturation during early development. The demonstrated influence of genetic variants on brain function, personality traits, and susceptibility to neuropsychiatric disorders suggests a critical importance of developmental mechanisms. However, little is known about how and when developmentally perturbed 5-HT signaling affects circuitry and resulting behavior. The 5-HT transporter (5-HTT) is a key regulator of extracellular 5-HT levels and we used pharmacologic strategies to manipulate 5-HTT function during development and determine behavioral consequences. Transient exposure to the 5-HTT inhibitors fluoxetine, clomipramine, and citalopram from postnatal day 4 (P4) to P21 produced abnormal emotional behaviors in adult mice. Similar treatment with the norepinephrine transporter (NET) inhibitor, desipramine, did not adversely affect adult behavior, suggesting that 5-HT and norepinephrine (NE) do not share the same effects on brain development. Shifting our period of treatment/testing to P90/P185 failed to mimic the effect of earlier exposure, demonstrating that 5-HT effects on adult behavior are developmentally specific. We have hypothesized that early-life perturbations of 5-HT signaling affect corticolimbic circuits that do not reach maturity until the peri-adolescent period. In support of this idea, we found that abnormal behaviors resulting from postnatal fluoxetine exposure have a post-pubescent onset and persist long after reaching adult age. A better understanding of the underlying 5-HT sensitive circuits and how they are perturbed should lead to new insights into how various genetic polymorphisms confer their risk to carriers. Furthermore, these studies should help determine whether in utero exposure to 5-HTT blocking drugs poses a risk for behavioral abnormalities in later life.
Collapse
|
478
|
Chronic fluoxetine treatment alters behavior, but not adult hippocampal neurogenesis, in BALB/cJ mice. Mol Psychiatry 2008; 13:119-21. [PMID: 18202694 DOI: 10.1038/sj.mp.4002104] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
479
|
Marcussen AB, Flagstad P, Kristjansen PEG, Johansen FF, Englund U. Increase in neurogenesis and behavioural benefit after chronic fluoxetine treatment in Wistar rats. Acta Neurol Scand 2008; 117:94-100. [PMID: 18184344 DOI: 10.1111/j.1600-0404.2007.00910.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Disturbances in hippocampal neurogenesis may be involved in the pathophysiology of depression and it has been argued that an increase in the generation of new nerve cells in the hippocampus is involved in the mechanism of action of antidepressants. MATERIALS AND METHODS Adult Wistar rats were treated with fluoxetine (10 mg/kg) 1 h, daily for 5 (subchronic) or 28 days (chronic) before the Novelty Suppressed Feeding test was performed. Cell proliferation and neurogenesis were analysed using the markers 5-bromo-deoxy-2'-uridine, Ki-67, and doublecortin. RESULTS A significant behavioural effect was found after 28 days of fluoxetine administration. However, no behavioural improvement was demonstrated after acute and subchronic treatment with fluoxetine. We further demonstrate that chronic antidepressant treatment increases cell proliferation as well as neurogenesis in the dentate gyrus, here using Wistar rats. CONCLUSIONS In further development of antidepressants, neurogenesis may serve as an important parameter to examine the efficacy and mechanism of action of novel drugs.
Collapse
Affiliation(s)
- A B Marcussen
- Department of Pharmacology Target Research, H. Lundbeck A/S, Valby, Denmark
| | | | | | | | | |
Collapse
|
480
|
Karlsson RM, Choe JS, Cameron HA, Thorsell A, Crawley JN, Holmes A, Heilig M. The neuropeptide Y Y1 receptor subtype is necessary for the anxiolytic-like effects of neuropeptide Y, but not the antidepressant-like effects of fluoxetine, in mice. Psychopharmacology (Berl) 2008; 195:547-57. [PMID: 17891380 DOI: 10.1007/s00213-007-0945-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 09/05/2007] [Indexed: 12/13/2022]
Abstract
RATIONALE Neuropeptide Y (NPY) is implicated in the pathophysiology of affective illness. Multiple receptor subtypes (Y1R, Y2R, and Y5R) have been suggested to contribute to NPY's effects on rodent anxiety and depression-related behaviors. OBJECTIVES To further elucidate the role of Y1R in (1) NPY's anxiolytic-like effects and (2) fluoxetine's antidepressant-like and neurogenesis-inducing effects. METHODS Mice lacking Y1R were assessed for spontaneous anxiety-like behavior (open field, elevated plus-maze, and light/dark exploration test) and Pavlovian fear conditioning, and for the anxiolytic-like effects of intracerebroventricularly (icv)-administrated NPY (elevated plus-maze). Next, Y1R -/- were assessed for the antidepressant-like effects of acute fluoxetine in the forced swim test and chronic fluoxetine in the novelty-induced hypophagia test, as well as for chronic fluoxetine-induced hippocampal neurogenesis. RESULTS Y1R -/- exhibited largely normal baseline behavior as compared to +/+ littermate controls. Intraventricular administration of NPY in Y1R -/- mice failed to produce the normal anxiolytic-like effect in the elevated plus-maze test seen in +/+ mice. Y1R mutant mice showed higher immobility in the forced swim test and longer latencies in the novelty-induced hypophagia test. In addition, Y1R -/- mice responded normally to the acute and chronic effects of fluoxetine treatment in the forced swim test and the novelty-induced hypophagia test, respectively, as well as increased neuronal precursor cell proliferation in the hippocampus. CONCLUSIONS These data demonstrate that Y1R is necessary for the anxiolytic-like effects of icv NPY, but not for the antidepressant-like or neurogenesis-inducing effects of fluoxetine. The present study supports targeting Y1R as a novel therapeutic target for anxiety disorders.
Collapse
MESH Headings
- Animals
- Antidepressive Agents, Second-Generation/pharmacology
- Anxiety/physiopathology
- Arousal/drug effects
- Arousal/physiology
- Cell Count
- Cell Division/drug effects
- Cell Division/physiology
- Conditioning, Classical/drug effects
- Conditioning, Classical/physiology
- Depression/physiopathology
- Fear/drug effects
- Fear/physiology
- Female
- Fluoxetine/pharmacology
- Hippocampus/drug effects
- Hippocampus/physiopathology
- Injections, Intraventricular
- Male
- Maze Learning/drug effects
- Maze Learning/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons/drug effects
- Neurons/physiology
- Neuropeptide Y/pharmacology
- Neuropeptide Y/physiology
- Receptors, Neuropeptide Y/drug effects
- Receptors, Neuropeptide Y/genetics
- Receptors, Neuropeptide Y/physiology
Collapse
Affiliation(s)
- Rose-Marie Karlsson
- Laboratory of Clinical and Translational Studies, NIH, National Institute of Alcohol Abuse and Alcoholism, NIH, 10 Center Drive, 1-15330, Bethesda, MD 20892-1375, USA.
| | | | | | | | | | | | | |
Collapse
|
481
|
Holick KA, Lee DC, Hen R, Dulawa SC. Behavioral effects of chronic fluoxetine in BALB/cJ mice do not require adult hippocampal neurogenesis or the serotonin 1A receptor. Neuropsychopharmacology 2008; 33:406-17. [PMID: 17429410 DOI: 10.1038/sj.npp.1301399] [Citation(s) in RCA: 233] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We previously reported that chronic, but not subchronic, treatment with the selective serotonin reuptake inhibitor (SSRI) fluoxetine altered behavior in the forced swimming test (FST) in BALB/cJ mice. We now use this model to investigate mechanisms underlying the delayed onset of the behavioral response to antidepressants, specifically (1) adult hippocampal neurogenesis and (2) expression of the 5-HT1A receptor. Here, we show data validating this model of chronic antidepressant action. We found the FST to be selectively responsive to chronic administration of the SSRI fluoxetine (18 mg/kg/day) and the tricyclic antidepressant desipramine (20 mg/kg/day), but not to the antipsychotic haloperidol (1 mg/kg/day) in BALB/cJ mice. The behavioral effects of fluoxetine emerged by 12 days of treatment, and were affected neither by ablation of progenitor cells of the hippocampus nor by genetic deletion of the 5-HT1A receptor. The effect of fluoxetine in the BALB/cJ mice was also neurogenesis-independent in the novelty-induced hypophagia test. We also found that chronic fluoxetine does not induce an increase in cell proliferation or the number of young neurons as measured by BrdU and doublecortin immunolabeling, respectively, in BALB/cJ mice. These data are in contrast to our previous report using a different strain of mice (129SvEvTac). In conclusion, we find that BALB/cJ mice show a robust response to chronic SSRI treatment in the FST, which is not mediated by an increase in new neurons in the hippocampus, and does not require the 5-HT1A receptor. These findings suggest that SSRIs can produce antidepressant-like effects via distinct mechanisms in different mouse strains.
Collapse
Affiliation(s)
- Kerri A Holick
- Department of Pharmacology, Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
482
|
Kinsey SG, Bailey MT, Sheridan JF, Padgett DA. The inflammatory response to social defeat is increased in older mice. Physiol Behav 2007; 93:628-36. [PMID: 18068740 DOI: 10.1016/j.physbeh.2007.11.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 10/18/2007] [Accepted: 11/01/2007] [Indexed: 12/26/2022]
Abstract
KINSEY, S. G., BAILEY, M. T., SHERIDAN, J. F., PADGETT, D. A. The inflammatory response to social defeat is increased in older mice. PHYSIOL BEHAV 91(0) 000-000, 2007. Previous research indicates that repeated social defeat of mice causes increased lymphocyte trafficking to the spleen, elevated proinflammatory cytokine production, and induced glucocorticoid insensitivity in splenocytes. Social defeat also causes increases in anxiety-like behavior. This study investigated whether repeated social defeat results in similar immunoregulatory and behavioral changes in older mice as those seen previously in young adult mice. The data revealed that, regardless of age, defeated mice had significantly more splenic CD11b+ Gr-1+ monocytes and neutrophils than controls. Supernatants harvested from cultured splenocytes from older mice contained comparatively higher IL-6 and TNF-alpha than supernatants from younger animals. In addition, those same cells derived from older defeated mice were hypersensitive to lipopolysaccharide (LPS) and insensitive to glucocorticoids in vitro. As seen previously in young adult mice, social defeat caused an increase in anxiety-like behavior in the open field test, but had no effect on learned helplessness in the forced swim test. These data indicated that repeated social defeat results in a proinflammatory state that is exacerbated in older mice. The implications of these data are noteworthy, given the strong role of inflammation in many age-related diseases.
Collapse
Affiliation(s)
- Steven G Kinsey
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH 43210 USA
| | | | | | | |
Collapse
|
483
|
Siegmund A, Wotjak CT. A mouse model of posttraumatic stress disorder that distinguishes between conditioned and sensitised fear. J Psychiatr Res 2007; 41:848-60. [PMID: 17027033 DOI: 10.1016/j.jpsychires.2006.07.017] [Citation(s) in RCA: 194] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 07/11/2006] [Accepted: 07/27/2006] [Indexed: 11/21/2022]
Abstract
The pathomechanisms of posttraumatic stress disorder (PTSD) are still unknown, but both fear conditioning and stress sensitisation are supposed to play a crucial role. Hence, valid animal models that model both associative and non-associative components of fear will facilitate elucidation of the biological substrates of the illness, and to develop novel and specific approaches for its prevention and therapy. Here we applied a single electric footshock to C57BL/6N (B6N) and C57BL/6JOla (B6JOla) mice and recorded the conditioned response to contextual trauma reminders (associative fear), the sensitised reaction to a neutral tone in a novel environment (non-associative fear, hyperarousal), social interaction and various emotional behaviours using Modified Holeboard, Test for Novelty-Induced Suppression of Feeding and Forced Swimming Test, after different incubation times (1, 14, 28 days). Freezing generally increased as a function of shock intensity. In B6N mice, sensitised fear was maximal 28 days after trauma and was accompanied by signs of emotional blunting and social withdrawal. B6JOla mice, in contrast, were less susceptible to develop PTSD-like symptoms. The phenotype of B6N exhibited high behavioural variance, allowing distinction between vulnerable and resilient individuals. Only in vulnerable B6N mice, chronic fluoxetine treatment - initiated after an incubation period of 28 days - ameliorated sensitised fear. This new mouse model fulfils common criteria for face and predictive validity and can be used to investigate the biological correlates of individual fear susceptibility, as well as the impact and interrelationship of associative and non-associative fear components in the development and maintenance of PTSD.
Collapse
Affiliation(s)
- Anja Siegmund
- Max-Planck-Institut für Psychiatrie, AG Neuronale Plastizität, Kraepelinstr. 2, D-80804 Munich, Germany
| | | |
Collapse
|
484
|
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
|
485
|
Shishkina GT, Kalinina TS, Dygalo NN. Up-regulation of tryptophan hydroxylase-2 mRNA in the rat brain by chronic fluoxetine treatment correlates with its antidepressant effect. Neuroscience 2007; 150:404-12. [PMID: 17950541 DOI: 10.1016/j.neuroscience.2007.09.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 09/01/2007] [Accepted: 10/03/2007] [Indexed: 12/25/2022]
Abstract
Tryptophan hydroxylase-2 (TPH2), the rate-limiting enzyme in 5-HT synthesis in the brain, is a candidate for participation in a mechanism mediating the antidepressant effect of selective 5-HT reuptake inhibitors such as fluoxetine. Using real-time reverse transcription-polymerase chain reaction (RT-PCR) and semi-quantitative RT-PCR techniques, we have examined the effects of fluoxetine administration with drinking water (7.5 mg/kg/day) for 2, 4 and 8 weeks on TPH2 mRNA expression in the midbrain part of the dorsal raphe nucleus (DRN) and in the brainstem containing the rest of the raphe complex. Fluoxetine treatment for 4 and 8 weeks significantly increased basal TPH2 mRNA levels in the midbrain, an effect that was correlated with the appearance of antidepressant-like effects in the forced swim test. A significant induction of TPH2 and 5-HT transporter (5-HTT) mRNAs was detected in the midbrain of untreated rats 24 h after the swim test. In these animals, the swim test also produced a marked decrease in 5-HT metabolite (5-hydroxyindoleacetic acid (5-HIAA)) content in the amygdala. Fluoxetine treatment for 4 and 8, but not for 2 weeks, abolished these swim-induced changes in TPH2 and 5-HTT mRNAs levels in the midbrain and 5-HIAA content in the amygdala. The results of the present study suggest that TPH2 gene expression in the midbrain part of the DRN is implicated in depression and stress response, as well as in the antidepressant fluoxetine action.
Collapse
Affiliation(s)
- G T Shishkina
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk 630090, Russia
| | | | | |
Collapse
|
486
|
Conley RK, Hutson PH. Effects of acute and chronic treatment with fluoxetine on stress-induced hyperthermia in telemetered rats and mice. Eur J Pharmacol 2007; 564:138-45. [PMID: 17462624 DOI: 10.1016/j.ejphar.2007.02.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 02/16/2007] [Accepted: 02/20/2007] [Indexed: 10/23/2022]
Abstract
Preclinical and clinical evidence suggests that anxiolytic effects are observed after chronic administration of the selective serotonin reuptake inhibitor fluoxetine. In contrast, acute treatment may increase signs of anxiety. The present study examined the effects of acute and chronic administration of fluoxetine on a physiological measure of anxiety, stress-induced hyperthermia, in rats and mice using radiotelemetry to record core temperature and locomotor activity and ethologically relevant stressors to evoke the hyperthermic response. In both species, the benzodiazepine agonist chlordiazepoxide reduced stress-induced hyperthermia at doses (5 mg/kg i.p. rat, 10 mg/kg p.o. mouse) that had no significant effect on locomotor activity. Similarly, in both species, chronic (21 days) treatment with fluoxetine attenuated the hyperthermic response without significantly affecting locomotor activity. However, acute fluoxetine elicited species-specific effects. Thus in mice, stress-induced hyperthermia and activity were unaffected by fluoxetine (20 mg/kg p.o.) consistent with a lack of anxiolytic or anxiogenic activity. In contrast, in rats, fluoxetine (10 mg/kg i.p.) caused a significant baseline hypothermia in the absence of stress, confounding further interpretation. In conclusion, stress-induced hyperthermia in mice was unaffected by acute treatment and significantly reduced by chronic treatment with fluoxetine. However, in rats chronic administration of fluoxetine significantly reduced stress-induced hyperthermia while the effects of acute treatment were confounded by a decrease in body temperature in the absence of stress. Together, these observations support the view that chronic administration of fluoxetine is anxiolytic; however, the stress-induced hyperthermia assay does not reveal anxiogenic effects of acute administration of fluoxetine in rats or mice.
Collapse
Affiliation(s)
- Rachel K Conley
- Merck Sharp & Dohme, Neuroscience Research Centre, Harlow, Essex, UK.
| | | |
Collapse
|
487
|
Earnheart JC, Schweizer C, Crestani F, Iwasato T, Itohara S, Mohler H, Lüscher B. GABAergic control of adult hippocampal neurogenesis in relation to behavior indicative of trait anxiety and depression states. J Neurosci 2007; 27:3845-54. [PMID: 17409249 PMCID: PMC2441879 DOI: 10.1523/jneurosci.3609-06.2007] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stressful experiences in early life are known risk factors for anxiety and depressive illnesses, and they inhibit hippocampal neurogenesis and the expression of GABA(A) receptors in adulthood. Conversely, deficits in GABAergic neurotransmission and reduced neurogenesis are implicated in the etiology of pathological anxiety and diverse mood disorders. Mice that are heterozygous for the gamma2 subunit of GABA(A) receptors exhibit a modest functional deficit in mainly postsynaptic GABA(A) receptors that is associated with a behavioral, cognitive, and pharmacological phenotype indicative of heightened trait anxiety. Here we used cell type-specific and developmentally controlled inactivation of the gamma2 subunit gene to further analyze the mechanism and brain substrate underlying this phenotype. Heterozygous deletion of the gamma2 subunit induced selectively in immature neurons of the embryonic and adult forebrain resulted in reduced adult hippocampal neurogenesis associated with heightened behavioral inhibition to naturally aversive situations, including stressful situations known to be sensitive to antidepressant drug treatment. Reduced adult hippocampal neurogenesis was associated with normal cell proliferation, indicating a selective vulnerability of postmitotic immature neurons to modest functional deficits in gamma2 subunit-containing GABA(A) receptors. In contrast, a comparable forebrain-specific GABA(A) receptor deficit induced selectively in mature neurons during adolescence lacked neurogenic and behavioral consequences. These results suggest that modestly reduced GABA(A) receptor function in immature neurons of the developing and adult brain can serve as a common molecular substrate for deficits in adult neurogenesis and behavior indicative of anxious and depressive-like mood states.
Collapse
Affiliation(s)
- John C Earnheart
- Department of Biology and Penn State Neuroscience Institute, Penn State University, University Park, Pennsylvania 16802, USA
| | | | | | | | | | | | | |
Collapse
|
488
|
Kinsey SG, Bailey MT, Sheridan JF, Padgett DA, Avitsur R. Repeated social defeat causes increased anxiety-like behavior and alters splenocyte function in C57BL/6 and CD-1 mice. Brain Behav Immun 2007; 21:458-66. [PMID: 17178210 PMCID: PMC1941837 DOI: 10.1016/j.bbi.2006.11.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 11/01/2006] [Accepted: 11/02/2006] [Indexed: 11/28/2022] Open
Abstract
The experimental model, social disruption (SDR), is a model of social stress in which mice are repeatedly attacked and defeated in their home cage by an aggressive conspecific. In terms of the impact of this stressor on the immune response, SDR has been reported to cause hyperinflammation and glucocorticoid insensitivity. To this point however, the behavioral consequences of SDR have not been thoroughly characterized. Because social defeat has been reported to cause anxiety- and depressive-like behaviors, the current study was designed to assess whether SDR also causes anxiety- and depressive-like behaviors. Using the light/dark preference test and the open field test as tools to measure behaviors characteristic of anxiety, the data showed that C57BL/6 and CD-1 male mice subjected to SDR displayed increased anxiety-like behavior. The increase in anxiety-like behaviors persisted for at least 1 week after the cessation of the stressor. In contrast, depressive-like behaviors were not elicited by SDR as assessed by the forced swim test or the tail suspension test. These data indicate that social disruption stress causes an increase in anxiety-like behaviors, but not depressive-like behaviors.
Collapse
Affiliation(s)
- Steven G. Kinsey
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Michael T. Bailey
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
- The Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - John F. Sheridan
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
- The Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - David A. Padgett
- Department of Psychology, The Ohio State University, Columbus, OH, USA
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
- The Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Ronit Avitsur
- The Academic College of Tel Aviv-Yaffo, School of Behavioral Sciences
| |
Collapse
|
489
|
Bhansali P, Dunning J, Singer SE, David L, Schmauss C. Early life stress alters adult serotonin 2C receptor pre-mRNA editing and expression of the alpha subunit of the heterotrimeric G-protein G q. J Neurosci 2007; 27:1467-73. [PMID: 17287521 PMCID: PMC6673584 DOI: 10.1523/jneurosci.4632-06.2007] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Infant maternal separation, a paradigm of early life stress in rodents, elicits long-lasting changes in gene expression that persist into adulthood. In BALB/c mice, an inbred strain with spontaneously elevated anxiety and stress reactivity, infant maternal separation led to increased depression-like behavioral responses to adult stress and robustly increased editing of serotonin 2C receptor pre-mRNA. Chronic fluoxetine treatment of adult BALB/c mice exposed to early life stress affected neither their behavioral responses to stress nor their basal 5-HT2C pre-mRNA editing phenotype. However, when fluoxetine was administered during adolescence, depression-like behavioral responses to stress were significantly diminished in these mice, and their basal and stress-induced 5-HT2C pre-mRNA editing phenotypes were significantly lower. Moreover, when BALB/c mice exposed to early life stress were raised in an enriched postweaning environment, their depression-like behavioral responses to adult stress were also significantly diminished. However, their 5-HT2C pre-mRNA editing phenotype remained unaltered. Hence, the similar behavioral effects of enrichment and fluoxetine treatment during adolescence were not accompanied by similar changes in 5-HT2C pre-mRNA editing. Enriched and nonenriched BALB/c mice exposed to early life stress also exhibited significantly increased expression of mRNA and protein encoding the G alpha q subunit of G-protein that couples to 5-HT2A/2C receptors. In contrast, G alpha q expression levels were significantly lower in fluoxetine-treated mice. These findings suggest that compensatory changes in G alpha q expression occur in mice with persistently altered 5-HT2C pre-mRNA editing and provide an explanation for the dissociation between 5-HT2C receptor editing phenotypes and behavioral stress responses.
Collapse
MESH Headings
- Age Factors
- Animals
- Anxiety, Separation/complications
- Anxiety, Separation/genetics
- Anxiety, Separation/psychology
- Body Weight
- Depressive Disorder/drug therapy
- Depressive Disorder/etiology
- Depressive Disorder/genetics
- Depressive Disorder/physiopathology
- Depressive Disorder/psychology
- Emotions
- Environment
- Female
- Fluoxetine/pharmacology
- Fluoxetine/therapeutic use
- GTP-Binding Protein alpha Subunits, Gq-G11/biosynthesis
- GTP-Binding Protein alpha Subunits, Gq-G11/genetics
- Genetic Predisposition to Disease
- Helplessness, Learned
- Male
- Mice
- Mice, Inbred BALB C/genetics
- Mice, Inbred BALB C/physiology
- Mice, Inbred BALB C/psychology
- Mice, Inbred C57BL
- Neocortex/metabolism
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- Protein Isoforms/physiology
- RNA Editing
- RNA Precursors/genetics
- Random Allocation
- Receptor, Serotonin, 5-HT2C/biosynthesis
- Receptor, Serotonin, 5-HT2C/genetics
- Receptor, Serotonin, 5-HT2C/physiology
- Serotonin/physiology
- Selective Serotonin Reuptake Inhibitors/pharmacology
- Selective Serotonin Reuptake Inhibitors/therapeutic use
- Species Specificity
- Swimming
Collapse
Affiliation(s)
- Punita Bhansali
- Department of Psychiatry and
- Department of Neuroscience, New York State Psychiatric Institute, New York, New York 10032
| | - Jane Dunning
- Department of Neuroscience, New York State Psychiatric Institute, New York, New York 10032
| | - Sarah E. Singer
- Center for Neurobiology and Behavior, Columbia University, and
| | - Leora David
- Department of Psychiatry and
- Department of Neuroscience, New York State Psychiatric Institute, New York, New York 10032
| | - Claudia Schmauss
- Department of Psychiatry and
- Department of Neuroscience, New York State Psychiatric Institute, New York, New York 10032
| |
Collapse
|
490
|
Bechtholt AJ, Hill TE, Lucki I. Anxiolytic effect of serotonin depletion in the novelty-induced hypophagia test. Psychopharmacology (Berl) 2007; 190:531-40. [PMID: 17165096 DOI: 10.1007/s00213-006-0615-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Accepted: 10/12/2006] [Indexed: 10/23/2022]
Abstract
RATIONALE Relatively little is known about the neural mechanisms underlying anxiety in the novelty-induced hypophagia test, the only known anxiety test that is responsive to chronic but not acute or subchronic antidepressant treatment. OBJECTIVES The goal of the present experiment was to characterize the role of serotonin in the ability of novelty to suppress feeding. MATERIALS AND METHODS Pair-housed male Sprague-Dawley rats were trained to eat graham cracker crumbs individually in their home cage (15 min/day). After stable daily intakes were obtained, the animals were depleted of serotonin using 4-chloro-DL -phenylalanine (150 mg kg(-1) day(-1) x 2 days). Forty-eight hours later, central serotonin was restored by the administration of the peripheral L -aromatic amino acid decarboxylase inhibitor, benserazide (10 mg/kg), followed 15 min later with the immediate precursor of serotonin, 5-hydroxy-L -tryptophan (30 mg/kg). Thirty minutes later, the animals were given access to graham cracker crumbs in a novel environment. RESULTS The animals demonstrated increased latencies to approach the food and reduced food intake in the novel environment. This effect was attenuated by serotonin depletion. Repletion of central serotonin restored the inhibitory response to novelty. The analysis of serotonin content in different brain regions confirmed that serotonin was depleted by greater than 90%, whereas the repletion treatment resulted in serotonin levels similar to nondepleted animals. CONCLUSIONS Acute depletion of serotonin acts to reduce anxiety behavior as measured by an inhibitory anxiety response during exposure to novel stimuli. These findings are in agreement with the proposed general role for serotonin in behavioral inhibition and that reductions of serotonin facilitate the adoption of more active coping responses to stress.
Collapse
Affiliation(s)
- Anita J Bechtholt
- Department of Psychiatry, 2204 Translational Research Labs, University of Pennsylvania, 125 South 31st Street, Philadelphia, PA 19104-3403, USA
| | | | | |
Collapse
|
491
|
David DJ, Klemenhagen KC, Holick KA, Saxe MD, Mendez I, Santarelli L, Craig DA, Zhong H, Swanson CJ, Hegde LG, Ping XI, Dong D, Marzabadi MR, Gerald CP, Hen R. Efficacy of the MCHR1 Antagonist N-[3-(1-{[4-(3,4-Difluorophenoxy)phenyl]methyl}(4-piperidyl))-4-methylphenyl]-2-methylpropanamide (SNAP 94847) in Mouse Models of Anxiety and Depression following Acute and Chronic Administration Is Independent of Hippocampal Neurogenesis. J Pharmacol Exp Ther 2007; 321:237-48. [PMID: 17237257 DOI: 10.1124/jpet.106.109678] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide that plays a role in the modulation of food intake and mood. In rodents, the actions of MCH are mediated via the MCHR1 receptor. The goal of this study was to investigate the effects of acute (1 h) and chronic (28 days) p.o. dosing of a novel MCHR1 antagonist, N-[3-(1-{[4-(3,4-difluorophenoxy)-phenyl]methyl}(4-piperidyl))-4-methylphenyl]-2-methylpropanamide (SNAP 94847), in three mouse models predictive of antidepressant/anxiolytic-like activity: novelty suppressed feeding (NSF) in 129S6/SvEvTac mice and light/dark paradigm (L/D) and forced swim test (FST) in BALB/cJ mice. A significant increase in the time spent in the light compartment of the L/D box was observed in response to acute and chronic treatment with SNAP 94847. An anxiolytic/antidepressant-like effect was found in the NSF test after acute and chronic treatment, whereas no effect was observed in the FST. Because neurogenesis in the dentate gyrus has been shown to be a requirement for the effects of antidepressants in the NSF test, we investigated whether neurogenesis was required for the effect of SNAP 94847. We showed that chronic treatment with SNAP 94847 stimulated proliferation of progenitors in the dentate gyrus. The efficacy of SNAP 94847 in the NSF test, however, was unaltered in mice in which neurogenesis was suppressed by X-irradiation. These results indicate that SNAP 94847 has a unique anxiolytic-like profile after both acute and chronic administration and that its mechanism of action is distinct from that of selective serotonin reuptake inhibitors and tricyclic antidepressants.
Collapse
Affiliation(s)
- D J David
- Center for Neurobiology and Behavior, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
492
|
Abstract
Major depressive disorder (MDD) is a debilitating and complex psychiatric disorder that involves multiple neural circuits and genetic and non-genetic risk factors. In the quest for elucidating the neurobiological basis of MDD, hippocampal neurogenesis has emerged as a candidate substrate, both for the etiology as well as treatment of MDD. This chapter critiques the advances made in the study of hippocampal neurogenesis as they relate to the neurogenic hypothesis of MDD. While an involvement of neurogenesis in the etiology of depression remains highly speculative, preclinical studies have revealed a novel and previously unrecognized role for hippocampal neurogenesis in mediating some of the behavioral effects of antidepressants. The implications of these findings are discussed to reevaluate the role of hippocampal neurogenesis in MDD.
Collapse
Affiliation(s)
- Amar Sahay
- Department of Neuroscience, Columbia University, New York, NY 10032, USA.
| | | | | |
Collapse
|
493
|
Lee HJ, Rao JS, Ertley RN, Chang L, Rapoport SI, Bazinet RP. Chronic fluoxetine increases cytosolic phospholipase A(2) activity and arachidonic acid turnover in brain phospholipids of the unanesthetized rat. Psychopharmacology (Berl) 2007; 190:103-15. [PMID: 17093977 DOI: 10.1007/s00213-006-0582-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 08/28/2006] [Indexed: 01/08/2023]
Abstract
RATIONALE Fluoxetine is used to treat unipolar depression and is thought to act by increasing the concentration of serotonin (5-HT) in the synaptic cleft, leading to increased serotonin signaling. The 5-HT(2A/2C) receptor subtypes are coupled to a phospholipase A(2) (PLA(2)). We hypothesized that chronic fluoxetine would increase the brain activity of PLA(2) and the turnover rate of arachidonic acid (AA) in phospholipids of the unanesthetized rat. MATERIALS AND METHODS To test this hypothesis, rats were administered fluoxetine (10 mg/kg) or vehicle intraperitoneally daily for 21 days. In the unanesthetized rat, [1-(14)C]AA was infused intravenously and arterial blood plasma was sampled until the animal was killed at 5 min and its brain was subjected to chemical, radiotracer, or enzyme analysis. RESULTS Using equations from our fatty acid model, we found that chronic fluoxetine compared with vehicle increased the turnover rate of AA within several brain phospholipids by 75-86%. The activity and protein levels of brain cytosolic PLA(2) (cPLA(2)) but not of secretory or calcium-independent PLA(2) were increased in rats administered fluoxetine. In a separate group of animals that received chronic fluoxetine followed by a 3-day saline washout, the turnover of AA and activity and protein levels of cPLA(2) were not significantly different from controls. The protein levels of cyclooxygenases 1 and 2 as well as the concentration of prostaglandin E(2) in rats chronically administered fluoxetine did not differ significantly from controls. CONCLUSION The results support the hypothesis that fluoxetine increases the cPLA(2)-mediated turnover of AA within brain phospholipids.
Collapse
Affiliation(s)
- Ho-Joo Lee
- Brain Physiology and Metabolism Section National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
494
|
Dziedzicka-Wasylewska M, Faron-Górecka A, Kuśmider M, Drozdowska E, Rogóz Z, Siwanowicz J, Caron MG, Bönisch H. Effect of antidepressant drugs in mice lacking the norepinephrine transporter. Neuropsychopharmacology 2006; 31:2424-32. [PMID: 16554743 DOI: 10.1038/sj.npp.1301064] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
One of the main theories concerning the mechanism of action of antidepressant drugs (ADs) is based on the notion that the neurochemical background of depression involves an impairment of central noradrenergic transmission with a concomitant decrease of the norepinephrine (NE) in the synaptic gap. Many ADs increase synaptic NE availability by inhibition of the reuptake of NE. Using mice lacking NE transporter (NET-/-) we examined their baseline phenotype as well as the response in the forced swim test (FST) and in the tail suspension test (TST) upon treatment with ADs that display different pharmacological profiles. In both tests, the NET-/- mice behaved like wild-type (WT) mice acutely treated with ADs. Autoradiographic studies showed decreased binding of the beta-adrenergic ligand [3H]CGP12177 in the cerebral cortex of NET-/- mice, indicating the changes at the level of beta-adrenergic receptors similar to those obtained with ADs treatment. The binding of [3H]prazosin to alpha1-adrenergic receptors in the cerebral cortex of NET-/- mice was also decreased, most probably as an adaptive response to the sustained elevation of extracellular NE levels observed in these mice. A pronounced NET knockout-induced shortening of the immobility time in the TST (by ca 50%) compared to WT mice was not reduced any further by NET-inhibiting ADs such as reboxetine, desipramine, and imipramine. Citalopram, which is devoid of affinity for the NET, exerted a significant reduction of immobility time in the NET-/- mice. In the FST, reboxetine, desipramine, imipramine, and citalopram administered acutely did not reduce any further the immobility time shortened by NET knockout itself (ca 25%); however, antidepressant-like action of repeatedly (7 days) administered desipramine was observed in NET-/- mice, indicating that the chronic presence of this drug may also affect other neurochemical targets involved in the behavioral reactions monitored by this test. From the present study, it may be concluded that mice lacking the NET may represent a good model of some aspects of depression-resistant behavior, paralleled with alterations in the expression of adrenergic receptors, which result as an adaptation to elevated levels of extracellular NE.
Collapse
|
495
|
Chen ZY, Jing D, Bath KG, Ieraci A, Khan T, Siao CJ, Herrera DG, Toth M, Yang C, McEwen BS, Hempstead BL, Lee FS. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science 2006; 314:140-3. [PMID: 17023662 PMCID: PMC1880880 DOI: 10.1126/science.1129663] [Citation(s) in RCA: 1018] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A common single-nucleotide polymorphism in the brain-derived neurotrophic factor (BDNF) gene, a methionine (Met) substitution for valine (Val) at codon 66 (Val66Met), is associated with alterations in brain anatomy and memory, but its relevance to clinical disorders is unclear. We generated a variant BDNF mouse (BDNF(Met/Met)) that reproduces the phenotypic hallmarks in humans with the variant allele. BDNF(Met) was expressed in brain at normal levels, but its secretion from neurons was defective. When placed in stressful settings, BDNF(Met/Met) mice exhibited increased anxiety-related behaviors that were not normalized by the antidepressant, fluoxetine. A variant BDNF may thus play a key role in genetic predispositions to anxiety and depressive disorders.
Collapse
Affiliation(s)
- Zhe-Yu Chen
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
- School of Medicine, Shandong University, Jinan, Shandong 250012, People’s Republic of China
| | - Deqiang Jing
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Kevin G. Bath
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Alessandro Ieraci
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Tanvir Khan
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Chia-Jen Siao
- Division of Hematology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Daniel G. Herrera
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Miklos Toth
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Chingwen Yang
- Gene Targeting Facility, The Rockefeller University, New York, NY 10021, USA
| | - Bruce S. McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10021, USA
| | - Barbara L. Hempstead
- Division of Hematology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Francis S. Lee
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA
| |
Collapse
|
496
|
Jacobson LH, Cryan JF. Feeling strained? Influence of genetic background on depression-related behavior in mice: a review. Behav Genet 2006; 37:171-213. [PMID: 17029009 DOI: 10.1007/s10519-006-9106-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Accepted: 08/11/2006] [Indexed: 02/03/2023]
Abstract
Depression is a growing pandemic in developed societies. The use of inbred mouse strains in pre-clinical psychiatric research has proven to be a valuable resource. Firstly, they provide the background for genetic manipulations that aid in the discovery of molecular pathways that may be involved in major depression. Further, inbred mouse strains are also being used in the determination of genetic and environmental influences that may pre-dispose or trigger depression-related behavior. This review aims to highlight the utility of inbred mouse strains in depression research, while providing an overview of the current state of research into behavioral differences between strains in paradigms commonly used in the field. Neurochemical differences that may underlie strain differences are examined, and some caveats and cautions associated with the use of inbred strains are highlighted.
Collapse
Affiliation(s)
- L H Jacobson
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4002, Basel, Switzerland
| | | |
Collapse
|
497
|
Kim EJ, Kim WR, Chi SE, Lee KH, Park EH, Chae JH, Park SK, Kim HT, Choi JS. Repetitive transcranial magnetic stimulation protects hippocampal plasticity in an animal model of depression. Neurosci Lett 2006; 405:79-83. [PMID: 16839687 DOI: 10.1016/j.neulet.2006.06.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Revised: 04/26/2006] [Accepted: 06/10/2006] [Indexed: 11/21/2022]
Abstract
Despite its therapeutic success in treating mood-related disorders, little is known about the mechanism by which repetitive transcranial magnetic stimulation (rTMS) alters physiological responses of neurons. Using the forced swim test (FST) in rats as a model of depression, we tested the protective effect of rTMS on synaptic plasticity, specifically, on the induction of hippocampal long-term potentiation (LTP). Male Sprague-Dawley rats were subjected to FST to induce immobility, a behavioral symptom of depression. They were subsequently treated with one of the three conditions: rTMS (rTMS: 1000 stimuli at 10Hz), sham rTMS (SHAM: acoustic stimulation only), or an antidepressant drug, fluoxetine (FLX: 10mg/kg, i.p.) for 7 days. There was a significant difference in immobility time between rTMS and SHAM groups after 7 days of treatment, but not after a single day. Following the second swim test on day 7, they were anesthetized and LTP was induced in vivo in the perforant path-dentate gyrus synapses. Another group (NAIVE) that had received no prior treatment was used as a control for LTP. The SHAM or FLX group exhibited little signs of LTP induction. On the contrary, the rTMS and NAIVE group showed a significant increase in field excitatory postsynaptic potentials after LTP induction. These results show that rTMS has an antidepressant-like effect after a relatively short period of treatment, and this effect might be mediated by a cellular process that can potentially reverse the impaired synaptic efficacy caused by the forced swim procedure.
Collapse
Affiliation(s)
- Eun Joo Kim
- Department of Psychology, Korea University, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
498
|
Dranovsky A, Hen R. Hippocampal neurogenesis: regulation by stress and antidepressants. Biol Psychiatry 2006; 59:1136-43. [PMID: 16797263 PMCID: PMC7537828 DOI: 10.1016/j.biopsych.2006.03.082] [Citation(s) in RCA: 472] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Revised: 03/24/2006] [Accepted: 04/06/2006] [Indexed: 11/20/2022]
Abstract
Accumulating evidence implicates hippocampal neurogenesis in the pathophysiology of depression. Psychosocial stress reduces neurogenesis in rodents, whereas chronic treatment with antidepressants increases neurogenesis and blocks the effects of stress. The effects of stress and antidepressant treatment on hippocampal neurogenesis parallel behavioral changes in animal models. Moreover, ablating hippocampal neurogenesis renders antidepressants inactive in behavioral paradigms used to model antidepressant response and anxiety-like behavior in mice. In humans, monoamine-modulating antidepressants demonstrate clinical efficacy in treating depression and anxiety, which are often precipitated by psychosocial stress. This review examines the mounting evidence that stress and antidepressant treatment regulate neurogenesis in animals. Special attention is paid to the cellular and molecular mechanisms by which this regulation takes place. An analysis of current animal models used to study response to stress and antidepressants indicates the importance of modeling chronic treatment, which reflects both changes in neurogenesis and clinical response. Exploring responses of hippocampal neurogenesis to experimental challenges in appropriate animal models should delineate the role of adult-born neurons in hippocampal physiology. Focusing on neurogenic response to experimental paradigms of stress and antidepressant treatment is particularly interesting for understanding the pathophysiology of major depressive disorder.
Collapse
Affiliation(s)
- Alex Dranovsky
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, New York 10032, USA.
| | | |
Collapse
|
499
|
Rao JS, Ertley RN, Lee HJ, Rapoport SI, Bazinet RP. Chronic fluoxetine upregulates activity, protein and mRNA levels of cytosolic phospholipase A2 in rat frontal cortex. THE PHARMACOGENOMICS JOURNAL 2006; 6:413-20. [PMID: 16636684 DOI: 10.1038/sj.tpj.6500391] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic lithium and carbamazepine, which are effective against mania in bipolar disorder, decrease the activity of cytosolic phospholipase A(2) (cPLA(2)) and the turnover rate of arachidonic acid in phospholipids in rat brain. Assuming that stages of bipolar disorder are related to brain arachidonic acid metabolism, we hypothesized that drugs effective in depression would increase cPLA(2) activity. To test this hypothesis, adult male CDF-344 rats were administered fluoxetine (10 mg/kg intraperitoneally (i.p.) or saline (control) (i.p.) chronically for 21 days. Frontal cortex cPLA(2) protein, phosphorylated cPLA(2), activity and mRNA levels were increased after chronic fluoxetine. Transcription factors (activator protein-1, activator protein-2, glucocorticoid response element, polyoma enhancer element-3 and nuclear factor-kappa B) that are known to regulate cPLA(2) gene expression were not significantly changed by chronic fluoxetine, but nuclear AU-rich element/poly(U)-binding/degradation factor-1 RNA-stabilizing protein was increased significantly. The results suggest that chronic fluoxetine increases brain cPLA(2) gene expression post-transcriptionally by increasing cPLA(2) mRNA stabilization. Chronic fluoxetine's effect on cPLA(2) expression was opposite to the effect reported with chronic lithium or carbamazepine administration, and may be part of fluoxetine's mode of action.
Collapse
Affiliation(s)
- J S Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
500
|
Hackler EA, Airey DC, Shannon CC, Sodhi MS, Sanders-Bush E. 5-HT(2C) receptor RNA editing in the amygdala of C57BL/6J, DBA/2J, and BALB/cJ mice. Neurosci Res 2006; 55:96-104. [PMID: 16580757 DOI: 10.1016/j.neures.2006.02.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2005] [Revised: 02/02/2006] [Accepted: 02/07/2006] [Indexed: 12/26/2022]
Abstract
Post-transcriptional RNA editing of the G-protein coupled 5-hydroxytryptamine-2C (5-HT(2C)) receptor predicts an array of 24 receptor isoforms, some of which are characterized by reduced constitutive activity and potency to initiate intracellular signaling. The amygdala is integral to anxiety, fear, and related psychiatric diseases. Activation of 5-HT(2C) receptors within the amygdala is anxiogenic. Here, we describe the RNA editing profiles from amygdala of two inbred mouse strains (BALB/cJ and DBA/2J) known to be more anxious than a third (C57BL/6J). We confirmed the strain anxiety differences using light<-->dark exploration, and we discovered that BALB/cJ and DBA/2J are each characterized by a higher functioning RNA editing profile than C57BL/6J. BALB/cJ and DBA/2J exhibit a roughly two-fold reduction in C site editing, and a corresponding two-fold reduction in the edited isoform VSV. C57BL/6J is characterized by a relative decrease in the unedited highly functional isoform INI. We estimated the heritability of editing at the C site to be approximately 40%. By sequencing genomic DNA, we found complete conservation between C57BL/6J, BALB/cJ, DBA/2J and 37 other inbred strains for the RNA edited region of Htr2c, suggesting Htr2c DNA sequence does not influence variation in Htr2c RNA editing between inbred strains of mice. We did, however, discover that serotonin turnover is reduced in BALB/cJ and DBA/2J, consistent with emerging evidence that synaptic serotonin levels regulate RNA editing. These results encourage further study of the causes and consequences of 5-HT(2C) receptor RNA editing in the amygdala of mice.
Collapse
Affiliation(s)
- Elizabeth A Hackler
- Department of Pharmacology, Vanderbilt University Medical Center, 465 21st Avenue South, Medical Research Building III, Room 8140, USA
| | | | | | | | | |
Collapse
|