Elsevier

Neuropharmacology

Volume 75, December 2013, Pages 426-436
Neuropharmacology

Chronic stress-induced changes in the rat brain: Role of sex differences and effects of long-term tianeptine treatment

https://doi.org/10.1016/j.neuropharm.2013.08.018Get rights and content

Highlights

  • Sex-related dimorphisms were identified on FOS immunoreactivity, CREB phosphorylation and HPA activation.

  • Chronic stress impaired neuroplasticity and HPA axis regulation in both sexes.

  • Tianeptine attenuated stress-induced impairments in a sex-specific manner.

  • Tianeptine modulated prefrontocortical activity in females and HPA axis activation in males.

Abstract

Growing evidence suggests neuroplasticity changes are pivotal in both the occurrence and treatment of affective disorders. Abnormal expression and/or phosphorylation of numerous plasticity-related proteins have been observed in depression, while prolonged antidepressant treatment has been associated with the attenuation of stress-mediated effects on dendritic remodeling and adult hippocampal neurogenesis in experimental animals. This study explores the neurobiological adaptations induced by chronic stress and/or long-term tianeptine treatment. Male and female rats were studied to determine the potential contributory role of sex differences on stress-induced pathology and antidepressant-mediated actions. Our results confirm chronic stress-induced HPA axis disturbance and neuroplasticity impairment in both sexes (i.e. reduced CREB phosphorylation and hippocampal BrdU labeling). Commonly ensuing neurobiological alterations were accompanied by unique sex-specific adaptations. When the antidepressant tianeptine was administered, HPA axis hyperactivity was attenuated and specific neuronal defects were ameliorated in both sexes. These findings provide novel insight into sex-related influences on the neurobiological substrates mediating chronic stress-induced actions on neuroplasticity and the mechanisms underlying tianeptine-mediated therapeutic effects.

Introduction

Affective disorders, such as anxiety and depression, are among today's most common and disabling illnesses. Despite the great burden on afflicted individuals and society, the exact neurobiological mechanisms underlying disease pathophysiology as well as treatment remain scarcely understood. Numerous factors account for this lack of progress including poor understanding of sex-dependent adaptations to chronic stress and/or long-term pharmacotherapy. Curiously, although women are twice as likely as men to suffer from stress-related psychiatric disorders and constitute the majority of antidepressant recipients (Kessler et al., 1993), basic research has focused predominantly on male animals (Palanza, 2001).

Stress represents a major contributor to neuronal dysfunctions in brain structures important for emotional processing and stress response regulation (Price and Drevets, 2010). In the hippocampus, chronic stress has been linked to profound structural and functional changes in humans and experimental animals (Leuner and Gould, 2010) including reduced hippocampal volume (Lee et al., 2009, MacQueen and Frodl, 2011), dendritic remodeling of pyramidal neurons (Magarinos et al., 1997) and suppression of adult neurogenesis (Kuipers et al., 2006, Dagyte et al., 2009). Exposure to stress is not unequivocally predictive however of subsequent hippocampal pathology as stress-induced effects on neuroplasticity vary substantially depending upon stressor type, duration and severity. Since repeated exposure to severe physical (tailshock) and psychological (resident-intruder) stressors have failed to affect hippocampal neurogenesis (Hanson et al., 2011), this challenges the hypothesis that any stressor of sufficient intensity and duration has a negative impact upon neuroplasticity. Furthermore, another factor well-known to affect stress-mediated consequences is sex (McLaughlin et al., 2009). Chronic stress has been shown to impair neurogenesis in a sex-dependent manner, by reducing neurogenic rates in one sex without affecting the other (Westenbroek et al., 2004, Kuipers et al., 2006, Shors et al., 2007, Oomen et al., 2009). Moreover it seems to do so by differentially targeting the regulation of neurogenesis on the cellular level as it inhibits cell proliferation and increases neural stem cell quiescence in males and reduces newborn neuron survival in females (Barha et al., 2011, Hillerer et al., 2013).

In contrast to stress, antidepressants exert their therapeutic actions by stimulating synaptic plasticity and enhancing adult neurogenesis. Increased expression and phosphorylation of CREB, a transcription factor with a well-established role in learning-related synaptic plasticity (Carlezon et al., 2005), have been observed in response to prolonged administration of serotonin and/or noradrenaline reuptake inhibitors (Nibuya et al., 1996, Thome et al., 2000, Kuipers et al., 2006). Long-term pharmacotherapy has also been shown to attenuate stress-evoked dendritic remodeling (Magarinos et al., 1999) and impairments of hippocampal neurogenesis (Malberg et al., 2000, Czeh et al., 2001). In this regard, the antidepressant tianeptine has proven particularly effective in ameliorating stress-induced morphological sequelae in the hippocampus, prefrontal cortex and amygdala (Zoladz et al., 2008). More importantly, although structurally related to tricyclic agents, tianeptine illustrates unique pharmacological properties that challenge the hypothesis of direct monoaminergic modulation to promote antidepressant action and therefore, provides new insights into the neurobiological basis of depression and its treatment (Brink et al., 2006, McEwen et al., 2010). Tianeptine triggers a cascade of neuronal adaptations which include increased phosphorylation of different glutamate receptors subtypes (NMDA and AMPA). As a major excitatory neurotransmitter, glutamate controls neuronal excitability and neuroplasticity in most brain circuits and glutamatergic dysfunctions have been documented in depression (Paul and Skolnick, 2003, Zarate et al., 2003). Evidence suggests tianeptine ameliorates depressive symptoms and attenuates neuroplasticity impairments by modulating the glutamatergic system in key brain structures (McEwen et al., 2010).

This study explores the cellular and molecular changes associated with chronic stress and/or long-term tianeptine treatment in male and female Wistar rats. Although sex differences have been identified in responses to stress and pharmacotherapy, few studies have systematically evaluated the complex interplay between sex, stress and antidepressants in animal disease models. Here, patterns of c-fos, phosphorylated CREB and BrdU immunoreactivity were examined in specific cortical and limbic regions known to undergo structural changes in depression and marked neuroplastic adaptations in response to antidepressants (Duman and Monteggia, 2006). Visualization of FOS induction provides a valuable tool for functional mapping of neural circuits activated by stress and antidepressants (Veening et al., 1998, Martinez et al., 2002, Kovacs, 2008), while changes in phosphorylated CREB and BrdU labeling provide insight into the effects of drug treatment and aversive experience on neuroplasticity. The findings presented here provide evidence to support sex-dependent mechanisms underlying the deleterious consequences of chronic stress and the beneficial influences of tianeptine.

Section snippets

Animals

Use was made of 10-week old male (n = 24) and female (n = 25) Wistar rats. Animals were individually housed (cages 45 × 28 × 20 cm) with ad libitum access to food and tap water. They were maintained on a 12-h light/dark cycle (07:00 lights on), weighed and handled daily (09:00) and allowed 10 days to acclimatize to their new environmental conditions prior to onset of the experiment. This study was designed to minimize animal numbers and suffering, and performed in accordance with the European

Body weight gain

Throughout this experiment, body weight gain increased steadily in both sexes (time: F21,861 = 175.334, P < 0.001) with males gaining weight faster than females (time × sex: F21,861 = 35.612, P < 0.001) (Fig. 1a,b). In line with previous data, repeated footshock stress attenuated this time-dependent increase in body weight (time × stress: F21,861 = 7.631, P < 0.001), an effect which was sex-dependent (time × stress × sex: F21,861 = 6.356, P < 0.001) given the marked reduction of weight gain in

Discussion

The risk of developing affective disorders, particularly anxiety and depression, has a strong sex bias, marked by a greater incidence in women. The high female-to-male ratio, especially during the reproductive years, is one of the most replicated findings in epidemiology (Leibenluft, 1999) and various factors contribute to this differential prevalence. Gender-related dimorphisms naturally occur in brain anatomy, function and neurochemistry (Jazin and Cahill, 2010). Women are also more prone to

Conclusions

In this study, sex differences were observed in prefrontocortical FOS-ir patterns, HPA axis activity, hippocampal and amygdalar neuroplasticity following repeated stress and long-term tianeptine treatment. These structures represent key components of the stress response system. Females demonstrated enhanced prefrontocortical activation and HPA axis hyperactivity, suggesting sex-specific adaptations underlying negative emotional processing. These may represent susceptibility traits to account

Acknowledgments

This study was financially supported by University of Groningen. The technical assistance of T. Koch and F. Postema was greatly appreciated. The authors would like to thank Prof. Dr. I.P. Kema for the corticosterone analysis. We also would like to acknowledge the Bergen Medical Research Foundation (BMFS) for its support through the “Young Investigator Recruitment” grant. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References (72)

  • I. Elakovic et al.

    Gender-specific response of brain corticosteroid receptors to stress and fluoxetine

    Brain Res.

    (2011)
  • N.D. Hanson et al.

    Several stressors fail to reduce adult hippocampal neurogenesis

    Psychoneuroendocrinology

    (2011)
  • P.E. Holtzheimer et al.

    Stuck in a rut: rethinking depression and its treatment

    Trends Neurosci.

    (2011)
  • P. Hughes et al.

    Basal expression of Fos, Fos-related, Jun, and Krox 24 proteins in rat hippocampus

    Brain Res. Mol. Brain Res.

    (1992)
  • R.C. Kessler et al.

    Sex and depression in the National Comorbidity Survey. I: lifetime prevalence, chronicity and recurrence

    J. Affect. Disord.

    (1993)
  • S.D. Kuipers et al.

    Unique patterns of FOS, phospho-CREB and BrdU immunoreactivity in the female rat brain following chronic stress and citalopram treatment

    Neuropharmacology

    (2006)
  • A.M. Magarinos et al.

    Effects of antidepressants and benzodiazepine treatments on the dendritic structure of CA3 pyramidal neurons after chronic stress

    Eur. J. Pharmacol.

    (1999)
  • B.S. McEwen et al.

    Prevention of stress-induced morphological and cognitive consequences

    Eur. Neuropsychopharmacol.

    (1997)
  • P.H. Roseboom et al.

    Predator threat induces behavioral inhibition, pituitary-adrenal activation and changes in amygdala CRF-binding protein gene expression

    Psychoneuroendocrinology

    (2007)
  • T.J. Shors et al.

    Neurogenesis and helplessness are mediated by controllability in males but not in females

    Biol. Psychiatry

    (2007)
  • M.B. Solomon et al.

    Sex differences in psychopathology: of gonads, adrenals and mental illness

    Physiol. Behav.

    (2009)
  • J. Stamp et al.

    Corticosterone modulates autonomic responses and adaptation of central immediate-early gene expression to repeated restraint stress

    Neuroscience

    (2001)
  • M. Szymanska et al.

    The effect of antidepressant drugs on the HPA axis activity, glucocorticoid receptor level and FKBP51 concentration in prenatally stressed rats

    Psychoneuroendocrinology

    (2009)
  • A. Trentani et al.

    Immunohistochemical changes induced by repeated footshock stress: revelations of gender-based differences

    Neurobiol. Dis.

    (2003)
  • J.G. Veening et al.

    Patterns of c-fos expression induced by fluvoxamine are different after acute vs. chronic oral administration

    Eur. Neuropsychopharmacol.

    (1998)
  • C. Westenbroek et al.

    Gender-specific effects of social housing in rats after chronic mild stress exposure

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2003)
  • C. Westenbroek et al.

    Chronic stress and social housing differentially affect neurogenesis in male and female rats

    Brain Res. Bull.

    (2004)
  • C.K. Barha et al.

    Chronic restraint stress in adolescence differentially influences hypothalamic-pituitary-adrenal axis function and adult hippocampal neurogenesis in male and female rats

    Hippocampus

    (2011)
  • C.B. Brink et al.

    Tianeptine: a novel atypical antidepressant that may provide new insights into the biomolecular basis of depression

    Recent Pat. CNS. Drug Discov.

    (2006)
  • B. Czeh et al.

    Stress-induced changes in cerebral metabolites, hippocampal volume, and cell proliferation are prevented by antidepressant treatment with tianeptine

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • G. Dagyte et al.

    The novel antidepressant agomelatine normalizes hippocampal neuronal activity and promotes neurogenesis in chronically stressed rats

    CNS. Neurosci. Ther.

    (2010)
  • G. Dagyte et al.

    Chronic stress and antidepressant agomelatine induce region-specific changes in synapsin I expression in the rat brain

    J. Neurosci. Res.

    (2011)
  • J.N. Flak et al.

    Identification of chronic stress-activated regions reveals a potential recruited circuit in rat brain

    Eur. J. Neurosci.

    (2012)
  • G. Guidotti et al.

    Glucocorticoid receptor and FKBP5 expression is altered following exposure to chronic stress: modulation by antidepressant treatment

    Neuropsychopharmacology

    (2013)
  • R.W. Guillery

    On counting and counting errors

    J. Comp. Neurol.

    (2002)
  • K.M. Hillerer et al.

    Sex-dependent regulation of hippocampal neurogenesis under basal and chronic stress conditions in rats

    Hippocampus

    (2013 Mar 18)
  • Cited by (22)

    • Effect of ZBD-2 on chronic pain, depressive-like behaviors, and recovery of motor function following spinal cord injury in mice

      2017, Behavioural Brain Research
      Citation Excerpt :

      In the present study, we observed gliocyte activation in the lumbar spinal dorsal horn following SCI, although treatment with ZBD-2 markedly suppressed chronic post-traumatic gliocyte activation at lesion sites and limited the development of hyperesthesia after SCI. In addition, previous studies have indicated that the CREB-BDNF signaling system can be affected by chronic stress and is a potential target for antidepressant treatment [54]. In fact, BDNF may be a crucial factor in mediating neuronal survival, differentiation, function, and plasticity.

    • Sex differences in drug addiction and response to exercise intervention: From human to animal studies

      2016, Frontiers in Neuroendocrinology
      Citation Excerpt :

      For example, estradiol could promote pCREB levels (Cheong et al., 2012). In particularly, fluctuating levels of estrogen along with the menstrual cycle affects CREB-BDNF signaling in the PFC and hippocampus, which have been implicated in the regulation of substance use disorders (Luine and Frankfurt, 2013; Carbone and Handa, 2013; Kuipers et al., 2013). In summary, the above results demonstrate that exercise-induced changes of CREB-BDNF signaling are likely to improve some drug addiction-induced deleterious behaviors, and CREB-BDNF signaling is different in males and females that exercise or are addicted to drugs.

    • Hippocampal signaling pathways are involved in stress-induced impairment of memory formation in rats

      2015, Brain Research
      Citation Excerpt :

      On the other hand, numerous studies have attempted to evaluate the effect of stress on the level of P-CREB in the brain. It has been shown that exposure to acute or chronic foot shock stress decreased the CREB and P-CREB levels in the hippocampus and the amygdala (Lin et al., 1998; Kuipers et al., 2013), while exposure to swimming stress increased P-CREB in these sites (Bilang-Bleuel et al., 2002; Shen et al., 2004a, 2004b). Moreover, exposure to acute immobilization stress as well as increase in the level of stress hormone can reduce the hippocampal level of the brain-derived neutrophic factor (BDNF) in rats (Adlard and Cotman, 2004).

    • Changes in hippocampal neurogenesis throughout early development

      2015, Neurobiology of Aging
      Citation Excerpt :

      Cell clusters were examined under the 40× objective. Total numbers of BrdU+, Sox1+, and DCX+ cells in the SGZ, GCL, and dentate gyrus (sum of cell in the SGZ and GCL) were estimated by multiplying the total number of cells every sixth section by 6 and reported as mean ± SE (Kuipers et al., 2009, 2013). Although this method does not account for split nuclei and a potential slight overcount, the thickness of the sections combined with the small object width and large inter-group differences, render the influence of this error as minimal and likely of no consequence to the conclusions.

    View all citing articles on Scopus
    1

    These authors contributed equally to the preparation of the manuscript.

    View full text