Antidepressant treatment outcome depends on the quality of the living environment: a pre-clinical investigation in mice

Igor Branchi, Sara Santarelli, Sara Capoccia, Silvia Poggini, Ivana D'Andrea, Francesca Cirulli, Enrico Alleva, Igor Branchi, Sara Santarelli, Sara Capoccia, Silvia Poggini, Ivana D'Andrea, Francesca Cirulli, Enrico Alleva

Abstract

Antidepressants represent the standard treatment for major depression. However, their efficacy is variable and incomplete. A growing number of studies suggest that the environment plays a major role in determining the efficacy of these drugs, specifically of selective serotonin reuptake inhibitors (SSRI). A recent hypothesis posits that the increase in serotonin levels induced by SSRI may not affect mood per se, but enhances neural plasticity and, consequently, renders the individual more susceptible to the influence of the environment. Thus, SSRI administration in a favorable environment would lead to a reduction of symptoms, while in a stressful environment might lead to a worse prognosis. To test this hypothesis, we treated C57BL/6 adult male mice with chronic fluoxetine while exposing them to either (i) an enriched environment, after exposure to a chronic stress period aimed at inducing a depression-like phenotype, or (ii) a stressful environment. Anhedonia, brain BDNF and circulating corticosterone levels, considered endophenotypes of depression, were investigated. Mice treated with fluoxetine in an enriched condition improved their depression-like phenotype compared to controls, displaying higher saccharin preference, higher brain BDNF levels and reduced corticosterone levels. By contrast, when chronic fluoxetine administration occurred in a stressful condition, mice showed a more distinct worsening of the depression-like profile, displaying a faster decrease of saccharin preference, lower brain BDNF levels and increased corticosterone levels. Our findings suggest that the effect of SSRI on depression-like phenotypes in mice is not determined by the drug per se but is induced by the drug and driven by the environment. These findings may be helpful to explain variable effects of SSRI found in clinical practice and to device strategies aimed at enhancing their efficacy by means of controlling environmental conditions.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Experimental design.
Figure 1. Experimental design.
(A,B) Environmental switch protocols. (A) Fluoxetine treatment in an enriched condition after exposure to stress. (B) Fluoxetine treatment in a stressful condition, after exposure to enrichment. (C,D) Constant environment protocols. (C) Fluoxetine treatment in a stressful condition, after exposure to stress. (D) Fluoxetine treatment in an enriched condition, after exposure to enrichment.
Figure 2. Saccharin preference during fluoxetine treatment…
Figure 2. Saccharin preference during fluoxetine treatment in an enriched condition after exposure to stress.
Stress exposure reduced saccharin preference in both groups of mice. Afterwards, fluoxetine-treated mice showed a significantly higher preference for the saccharin solution compared to control mice. # indicates p = 0.0120 and * indicates p = 0.0464, vs. vehicle group. Data are means ± S.E.M.
Figure 3. BDNF and corticosterone levels in…
Figure 3. BDNF and corticosterone levels in mice in an enriched condition after exposure to stress.
Following the 21 days of recovery, fluoxetine mice showed a significant increase of hippocampal and hypothalamic BDNF levels as well as significantly more marked decrease in corticosterone levels compared to controls. * and ** indicate, respectively, p<0.05 and 0.01 vs. vehicle group. Delta (Δ) values were calculated comparing data obtained on the day before treatment and on last day of treatment. Data are means ± S.E.M.
Figure 4. Anhedonic profile during fluoxetine treatment…
Figure 4. Anhedonic profile during fluoxetine treatment in a stressful condition after exposure to enrichment.
When social stress was imposed during treatment, fluoxetine-treated mice showed a faster and more marked reduction of preference for the saccharin solution compared to control mice. # indicates p = 0.0199, * and ** indicate, respectively, p<0.05 and 0.01 vs. vehicle group. Data are means ± S.E.M.
Figure 5. BDNF and corticosterone levels in…
Figure 5. BDNF and corticosterone levels in mice in a stressful conditions after exposure to enrichment.
Following social stress mice treated with fluoxetine showed reduced BDNF levels in both hippocampus and hypothalamus compared to control mice. Plasmatic corticosterone levels resulted increased in fluoxetine mice as shown by the statistically significant difference between levels before and after the treatment period. ** indicates p<0.01 vs. vehicle group. Delta (Δ) values were calculated comparing data obtained on the day before treatment and on last day of treatment. Data are means ± S.E.M.
Figure 6. Results of experiments investigating the…
Figure 6. Results of experiments investigating the effects of fluoxetine treatment in a constant environment.
(A,B,C,D) Fluoxetine treatment in a stressful condition, after exposure to stress. (A) Saccharin preference: exposure to stress before treatment significantly reduced saccharin preference. The following fluoxetine treatment administered in stressful conditions did not modify the anhedonic response. (B) Hippocampal and (C) hypothalamic BDNF levels: no difference between the two groups has been found. (D) Corticosterone levels. Fluoxetine mice showed a significant increase in corticosterone levels compared to vehicle. (E,F,G,H) Fluoxetine treatment in an enriched condition, after exposure to enrichment. (E) Saccharin preference: no difference between the two groups has been found. (F) Hippocampal and (G) hypothalamic BDNF levels: no difference between the two groups has been found. (H) Corticosterone levels: fluoxetine mice showed a significant decrease in corticosterone levels compared to vehicle. * indicates p<0.05 vs. vehicle group. # indicates p<0.01 vs. baseline level. Delta (Δ) values were calculated comparing data obtained on the day before treatment and on last day of treatment. Data are means ± S.E.M.

References

    1. WHO (2008) The global burden of disease: 2004 update: World Health Organization.
    1. Greenberg PE, Kessler RC, Birnbaum HG, Leong SA, Lowe SW, et al. (2003) The economic burden of depression in the United States: how did it change between 1990 and 2000? J Clin Psychiatry 64: 1465–1475.
    1. Balak N, Elmaci I (2007) Costs of disorders of the brain in Europe. Eur J Neurol 14: e9.
    1. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, et al. (2006) Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry 163: 28–40.
    1. Angell M (2008) Industry-sponsored clinical research: a broken system. JAMA 300: 1069–1071.
    1. Fournier JC, DeRubeis RJ, Hollon SD, Dimidjian S, Amsterdam JD, et al. (2010) Antidepressant drug effects and depression severity: a patient-level meta-analysis. JAMA 303: 47–53.
    1. Turner EH, Matthews AM, Linardatos E, Tell RA, Rosenthal R (2008) Selective publication of antidepressant trials and its influence on apparent efficacy. N Engl J Med 358: 252–260.
    1. Kirsch I, Deacon BJ, Huedo-Medina TB, Scoboria A, Moore TJ, et al. (2008) Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration. PLoS Med 5: e45.
    1. Moncrieff J, Kirsch I (2005) Efficacy of antidepressants in adults. BMJ 331: 155–157.
    1. Moncrieff J, Wessely S, Hardy R (2005) Active Placebos Versus Antidepressants for Depression. The Cochrane Library 4
    1. Kirsch I (2010) The Emperor's New Drugs: Exploding the Antidepressant Myth. New York: Basic Books.
    1. Fountoulakis KN, Moller HJ (2011) Efficacy of antidepressants: a re-analysis and re-interpretation of the Kirsch data. Int J Neuropsychopharmacol 14: 405–412.
    1. McAllister-Williams RH (2008) Do antidepressants work? A commentary on "Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration" by Kirsch et al. Evid Based Ment Health 11: 66–68.
    1. Branchi I (2011) The double edged sword of neural plasticity: increasing serotonin levels leads to both greater vulnerability to depression and improved capacity to recover. Psychoneuroendocrinology 36: 339–351.
    1. Hackman DA, Farah MJ, Meaney MJ (2010) Socioeconomic status and the brain: mechanistic insights from human and animal research. Nat Rev Neurosci 11: 651–659.
    1. Belsky J, Jonassaint C, Pluess M, Stanton M, Brummett B, et al. (2009) Vulnerability genes or plasticity genes? Mol Psychiatry 14: 746–754.
    1. Homberg JR (2012) Genetic sensitivity to the environment, across lifetime. Behav Brain Sci 35: 368.
    1. Kiser D, Steemers B, Branchi I, Homberg JR (2011) The reciprocal interaction between serotonin and social behaviour. Neurosci Biobehav Rev
    1. Maya Vetencourt JF, Sale A, Viegi A, Baroncelli L, De Pasquale R, et al. (2008) The antidepressant fluoxetine restores plasticity in the adult visual cortex. Science 320: 385–388.
    1. Karpova NN, Pickenhagen A, Lindholm J, Tiraboschi E, Kulesskaya N, et al. (2011) Fear erasure in mice requires synergy between antidepressant drugs and extinction training. Science 334: 1731–1734.
    1. Brummett BH, Boyle SH, Siegler IC, Kuhn CM, Ashley-Koch A, et al. (2008) Effects of environmental stress and gender on associations among symptoms of depression and the serotonin transporter gene linked polymorphic region (5-HTTLPR). Behav Genet 38: 34–43.
    1. Eley TC, Sugden K, Corsico A, Gregory AM, Sham P, et al. (2004) Gene-environment interaction analysis of serotonin system markers with adolescent depression. Mol Psychiatry 9: 908–915.
    1. Cohen A, Houck PR, Szanto K, Dew MA, Gilman SE, et al. (2006) Social inequalities in response to antidepressant treatment in older adults. Arch Gen Psychiatry 63: 50–56.
    1. Cryan JF, Holmes A (2005) The ascent of mouse: advances in modelling human depression and anxiety. Nat Rev Drug Discov 4: 775–790.
    1. Cirulli F, Alleva E (2009) The NGF saga: from animal models of psychosocial stress to stress-related psychopathology. Front Neuroendocrinol 30: 379–395.
    1. Sen S, Duman R, Sanacora G (2008) Serum brain-derived neurotrophic factor, depression, and antidepressant medications: meta-analyses and implications. Biol Psychiatry 64: 527–532.
    1. Hellweg R, Ziegenhorn A, Heuser I, Deuschle M (2008) Serum concentrations of nerve growth factor and brain-derived neurotrophic factor in depressed patients before and after antidepressant treatment. Pharmacopsychiatry 41: 66–71.
    1. Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N, et al. (2003) Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biol Psychiatry 54: 70–75.
    1. Nibuya M, Nestler EJ, Duman RS (1996) Chronic antidepressant administration increases the expression of cAMP response element binding protein (CREB) in rat hippocampus. J Neurosci 16: 2365–2372.
    1. Chourbaji S, Brandwein C, Gass P (2011) Altering BDNF expression by genetics and/or environment: impact for emotional and depression-like behaviour in laboratory mice. Neurosci Biobehav Rev 35: 599–611.
    1. Chourbaji S, Hortnagl H, Molteni R, Riva MA, Gass P, et al. (2012) The impact of environmental enrichment on sex-specific neurochemical circuitries - effects on brain-derived neurotrophic factor and the serotonergic system. Neuroscience 220: 267–276.
    1. Nemeroff CB, Widerlov E, Bissette G, Walleus H, Karlsson I, et al. (1984) Elevated concentrations of CSF corticotropin-releasing factor-like immunoreactivity in depressed patients. Science 226: 1342–1344.
    1. Holsboer F (2000) The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 23: 477–501.
    1. Ising M, Horstmann S, Kloiber S, Lucae S, Binder EB, et al. (2007) Combined dexamethasone/corticotropin releasing hormone test predicts treatment response in major depression - a potential biomarker? Biol Psychiatry 62: 47–54.
    1. Chourbaji S, Brandwein C, Vogt MA, Dormann C, Gass P (2008) Evaluation of effects of previous exposure to an acute stressor before testing for depression-like behaviours in mice. Stress 11: 170–175.
    1. Branchi I, D'Andrea I, Cirulli F, Lipp HP, Alleva E (2010) Shaping brain development: mouse communal nesting blunts adult neuroendocrine and behavioral response to social stress and modifies chronic antidepressant treatment outcome. Psychoneuroendocrinology 35: 743–751.
    1. Knapska E, Walasek G, Nikolaev E, Neuhausser-Wespy F, Lipp HP, et al. (2006) Differential involvement of the central amygdala in appetitive versus aversive learning. Learn Mem 13: 192–200.
    1. Mechan AO, Wyss A, Rieger H, Mohajeri MH (2009) A comparison of learning and memory characteristics of young and middle-aged wild-type mice in the IntelliCage. J Neurosci Methods 180: 43–51.
    1. Dulawa SC, Holick KA, Gundersen B, Hen R (2004) Effects of chronic fluoxetine in animal models of anxiety and depression. Neuropsychopharmacology 29: 1321–1330.
    1. Kramer PD (1994) Listening to Prozac: Psychiatrist Explores Antidepressant Drugs and the Remaking of the Self London: Fourth Estate Ltd.
    1. Willner P (1997) Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. Psychopharmacology (Berl) 134: 319–329.
    1. Strekalova T, Spanagel R, Bartsch D, Henn FA, Gass P (2004) Stress-induced anhedonia in mice is associated with deficits in forced swimming and exploration. Neuropsychopharmacology 29: 2007–2017.
    1. Bessa JM, Ferreira D, Melo I, Marques F, Cerqueira JJ, et al. (2009) The mood-improving actions of antidepressants do not depend on neurogenesis but are associated with neuronal remodeling. Mol Psychiatry 14: 764–773, 739.
    1. Rygula R, Abumaria N, Flugge G, Hiemke C, Fuchs E, et al. (2006) Citalopram counteracts depressive-like symptoms evoked by chronic social stress in rats. Behav Pharmacol 17: 19–29.
    1. Brenes JC, Fornaguera J (2009) The effect of chronic fluoxetine on social isolation-induced changes on sucrose consumption, immobility behavior, and on serotonin and dopamine function in hippocampus and ventral striatum. Behav Brain Res 198: 199–205.
    1. Prendergast MA, Yells DP, Balogh SE, Paige SR, Hendricks SE (2002) Fluoxetine differentially suppresses sucrose solution consumption in free-fed and food-deprived rats--reversal by amantadine. Med Sci Monit 8: BR385–390.
    1. Sammut S, Bethus I, Goodall G, Muscat R (2002) Antidepressant reversal of interferon-alpha-induced anhedonia. Physiol Behav 75: 765–772.
    1. Tonissaar M, Mallo T, Eller M, Haidkind R, Koiv K, et al. (2008) Rat behavior after chronic variable stress and partial lesioning of 5-HT-ergic neurotransmission: effects of citalopram. Prog Neuropsychopharmacol Biol Psychiatry 32: 164–177.
    1. Conti AC, Cryan JF, Dalvi A, Lucki I, Blendy JA (2002) cAMP response element-binding protein is essential for the upregulation of brain-derived neurotrophic factor transcription, but not the behavioral or endocrine responses to antidepressant drugs. J Neurosci 22: 3262–3268.
    1. Dias BG, Banerjee SB, Duman RS, Vaidya VA (2003) Differential regulation of brain derived neurotrophic factor transcripts by antidepressant treatments in the adult rat brain. Neuropharmacology 45: 553–563.
    1. Altieri M, Marini F, Arban R, Vitulli G, Jansson BO (2004) Expression analysis of brain-derived neurotrophic factor (BDNF) mRNA isoforms after chronic and acute antidepressant treatment. Brain Res 1000: 148–155.
    1. Jacobsen JP, Mork A (2004) The effect of escitalopram, desipramine, electroconvulsive seizures and lithium on brain-derived neurotrophic factor mRNA and protein expression in the rat brain and the correlation to 5-HT and 5-HIAA levels. Brain Res 1024: 183–192.
    1. Kozisek ME, Middlemas D, Bylund DB (2008) Brain-derived neurotrophic factor and its receptor tropomyosin-related kinase B in the mechanism of action of antidepressant therapies. Pharmacol Ther 117: 30–51.
    1. Miro X, Perez-Torres S, Artigas F, Puigdomenech P, Palacios JM, et al. (2002) Regulation of cAMP phosphodiesterase mRNAs expression in rat brain by acute and chronic fluoxetine treatment. An in situ hybridization study. Neuropharmacology 43: 1148–1157.
    1. Zetterstrom TS, Pei Q, Madhav TR, Coppell AL, Lewis L, et al. (1999) Manipulations of brain 5-HT levels affect gene expression for BDNF in rat brain. Neuropharmacology 38: 1063–1073.
    1. Alboni S, Benatti C, Capone G, Corsini D, Caggia F, et al. (2010) Time-dependent effects of escitalopram on brain derived neurotrophic factor (BDNF) and neuroplasticity related targets in the central nervous system of rats. Eur J Pharmacol 643: 180–187.
    1. Goekint M, Roelands B, Heyman E, Njemini R, Meeusen R (2011) Influence of citalopram and environmental temperature on exercise-induced changes in BDNF. Neurosci Lett 494: 150–154.
    1. Pariante CM, Thomas SA, Lovestone S, Makoff A, Kerwin RW (2004) Do antidepressants regulate how cortisol affects the brain? Psychoneuroendocrinology 29: 423–447.
    1. Uys JD, Muller CJ, Marais L, Harvey BH, Stein DJ, et al. (2006) Early life trauma decreases glucocorticoid receptors in rat dentate gyrus upon adult re-stress: reversal by escitalopram. Neuroscience 137: 619–625.
    1. Anacker C, Zunszain PA, Carvalho LA, Pariante CM (2011) The glucocorticoid receptor: pivot of depression and of antidepressant treatment? Psychoneuroendocrinology 36: 415–425.
    1. Shen Q, Lal R, Luellen BA, Earnheart JC, Andrews AM, et al. (2010) gamma-Aminobutyric acid-type A receptor deficits cause hypothalamic-pituitary-adrenal axis hyperactivity and antidepressant drug sensitivity reminiscent of melancholic forms of depression. Biol Psychiatry 68: 512–520.
    1. Weber CC, Eckert GP, Muller WE (2006) Effects of antidepressants on the brain/plasma distribution of corticosterone. Neuropsychopharmacology 31: 2443–2448.
    1. Berry A, Bellisario V, Capoccia S, Tirassa P, Calza A, et al. (2012) Social deprivation stress is a triggering factor for the emergence of anxiety- and depression-like behaviours and leads to reduced brain BDNF levels in C57BL/6J mice. Psychoneuroendocrinology 37: 762–772.
    1. Jacobsen JP, Mork A (2006) Chronic corticosterone decreases brain-derived neurotrophic factor (BDNF) mRNA and protein in the hippocampus, but not in the frontal cortex, of the rat. Brain Res 1110: 221–225.
    1. Ridder S, Chourbaji S, Hellweg R, Urani A, Zacher C, et al. (2005) Mice with genetically altered glucocorticoid receptor expression show altered sensitivity for stress-induced depressive reactions. J Neurosci 25: 6243–6250.
    1. Cohen A, Gilman SE, Houck PR, Szanto K, Reynolds CF 3rd (2009) Socioeconomic status and anxiety as predictors of antidepressant treatment response and suicidal ideation in older adults. Soc Psychiatry Psychiatr Epidemiol 44: 272–277.
    1. WHO (2010) Equity, social determinants and public health programmes.
    1. Feifel D (2008) More depressing news on antidepressants: should we panic? Psychiatry (Edgmont) 5: 35–36.
    1. Olfson M, Marcus SC (2009) National patterns in antidepressant medication treatment. Arch Gen Psychiatry 66: 848–856.
    1. Kirsch I, Scoboria A, Moore TJ (2002) Antidepressants and placebos: secrets, revelations, and unanswered questions. Prev Treat 5: article 33.
    1. Moncrieff J (2003) A comparison of antidepressant trials using active and inert placebos. Int J Methods Psychiatr Res 12: 117–127.
    1. Wiles N, Thomas L, Abel A, Ridgway N, Turner N et al... (2012) Cognitive behavioural therapy as an adjunct to pharmacotherapy for primary care based patients with treatment resistant depression: results of the CoBalT randomised controlled trial. Lancet.

Source: PubMed

3
订阅