Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2008 Dec;6(4):311–321. doi: 10.2174/157015908787386096

Tianeptine: An Antidepressant with Memory-Protective Properties

Phillip R Zoladz 1,2,4, Collin R Park 1,2,4, Carmen Muñoz 5, Monika Fleshner 6, David M Diamond 1,2,3,4,*
PMCID: PMC2701287  PMID: 19587852

Abstract

The development of effective pharmacotherapy for major depression is important because it is such a widespread and debilitating mental disorder. Here, we have reviewed preclinical and clinical studies on tianeptine, an atypical antidepressant which ameliorates the adverse effects of stress on brain and memory. In animal studies, tianeptine has been shown to prevent stress-induced morphological sequelae in the hippocampus and amygdala, as well as to prevent stress from impairing synaptic plasticity in the prefrontal cortex and hippocampus. Tianeptine also has memory-protective characteristics, as it blocks the adverse effects of stress on hippocampus-dependent learning and memory. We have further extended the findings on stress, memory and tianeptine here with two novel observations: 1) stress impairs spatial memory in adrenalectomized (ADX), thereby corticosterone-depleted, rats; and 2) the stress-induced impairment of memory in ADX rats is blocked by tianeptine. These findings are consistent with previous research which indicates that tianeptine produces anti-stress and memory-protective properties without altering the response of the hypothalamic-pituitary-adrenal axis to stress. We conclude with a discussion of findings which indicate that tianeptine accomplishes its anti-stress effects by normalizing stress-induced increases in glutamate in the hippocampus and amygdala. This finding is potentially relevant to recent research which indicates that abnormalities in glutamatergic neurotransmission are involved in the pathogenesis of depression. Ultimately, tianeptine’s prevention of depression-induced sequelae in the brain is likely to be a primary factor in its effectiveness as a pharmacological treatment for depression.

Key Words: Depression, tianeptine, stress, memory, synaptic plasticity, animal models.

INTRODUCTION

Depression is a widespread, recurrent mental disorder that has detrimental effects on individuals, as well as society, at large [44,189]. Although considerable progress has been made in characterizing the neurobiological sequelae that result from this disorder, the factors that are responsible for depression’s development and progression are not well understood. Research indicates that there is a heritable component to depression, and, more recently, investigators have identified candidate genes that appear to increase one’s susceptibility for the disorder [98,184]. This area of research has provided insight into the etiology of depression with the finding that gene polymorphisms interact with environmental factors, such as stressful events, to increase the likelihood that a person will develop major depression [15,17,62,82, 112,206].

For the past few decades, the prevailing view has been that depression results from abnormally low levels of monoamine neurotransmitter substances (e.g., serotonin, norepinephrine, dopamine), which is commonly known as the monoamine hypothesis [12,22,171]. Support for this hypothesis was based on the incidental finding that efficacious antidepressants, such as monoamine oxidase inhibitors and tricyclics, increased monoamine neurotransmitter levels [127]. Therefore, the primary focus of pharmacotherapy for depression has been to prescribe agents which are known to increase levels of the neurotransmitter serotonin, and today, selective serotonin reuptake inhibitors (SSRIs), such as fluoxetine, paroxetine and sertraline, are the most prescribed pharmacological treatments for this disorder [8,195].

Recent research suggests that increasing the levels of monoamines provides only an indirect contribution to antidepressant actions [186]. Moreover, some findings are inconsistent with the monoamine hypothesis of depression, thereby suggesting that the neurochemical basis of the disorder is more complicated than previously considered. For instance, traditional antidepressants ameliorate depressive symptoms only in a subset of patients, despite their low levels of monoamines [12], and are largely ineffective for people with severe forms of depression [113,137]. In these latter cases, clinicians may resort to electroconvulsive shock therapy (ECT), which has proven to be one of the most effective treatments for severe, pharmacologically-resistant forms of depression [136,147]. Despite its effectiveness, however, ECT’s mechanism of action remains largely unknown.

An alternative and well-established treatment for depression is tianeptine, an antidepressant which does not share pharmacological properties with TCAs, MAOIs or SSRIs [12,18,95,145,191]. Early studies suggested that tianeptine enhanced the uptake of serotonin [28,78,124,200], but more recent work indicates that tianeptine’s actions as an antidepressant are independent of modulating serotonin levels [145,193]. Instead, tianeptine’s primary mode of action is to influence the expression of synaptic plasticity [18,36,55, 77,120,121] via the modulation of glutamatergic neurotransmission [88,118,158,159]. Tianeptine’s effectiveness in treating depression is of clinical, as well as conceptual, significance. That is, the contrast in mechanistic actions between SSRIs and tianeptine, combined with the observation that both types of agents can treat depression, serves as a challenge to the heuristic value of the monoamine hypothesis of depression [66,135].

CHRONIC STRESS AND STRUCTURAL PLASTICITY IN THE HIPPOCAMPUS, PREFRONTAL CORTEX AND AMYGDALA

In recent years, researchers have suggested that depression is manifested through alterations in neuroplasticity, which involves structural and functional changes in how the brain processes information [55,77,191]. Investigators have contended that the emotional and cognitive components of depression manifest themselves as changes in neurochemical levels that ultimately produce significant alterations in brain morphology and, consequently, function [55]. In depressed patients, studies have described structural and functional alterations in three brain regions that are highly involved in emotional and cognitive processing: the hippocampus, prefrontal cortex and amygdala [174].

In general, studies have reported significant reductions of hippocampal and prefrontal cortex volumes in depressed patients [74,111,194]. The hippocampus is a medial temporal lobe structure which is important for declarative memory in humans [49,182] and spatial working memory in rodents [20,21,79,128,129,207]. The prefrontal cortex is located in the anterior portion of the frontal lobe and plays an important role in complex cognitive processes, such as planning, decision-making and behavioral flexibility [13]. Depressed individuals exhibit impaired performance on hippocampus- and prefrontal cortex-dependent cognitive tasks, which corresponds with reduced or abnormal activity in each of these brain regions when depressed patients engage in such tasks [47,126]. In contrast to the hippocampus and prefrontal cortex, amygdala volumes of depressed patients are larger than those of healthy individuals following the first episode of depression [54]. However, with recurrent episodes, amygdala volumes in depressed patients tend to be smaller than those of controls [175]. Nevertheless, most work has reported that activity of the amygdala is increased in depressed individuals [43] and with successful treatment, significantly declines [178].

It is well-established that stress significantly increases one’s likelihood of developing depression [81,132]. Extensive preclinical research has shown that chronic stress produces behavioral alterations that are analogous to those observed in depressed patients (e.g., anhedonia, learned helplessness, cognitive impairments) [4,56,114]. Thus, researchers have utilized animal models of stress effects on brain and behavior to potentially develop a better understanding of the neurobiological sequelae of this disorder. Animal models have shown that chronic stress significantly reduces the length, spine density and arborization of dendrites on neurons located in the prefrontal cortex [26,33,99,152,153] and hippocampus [31,87,97,106,108,122,198,203], while increasing each one of these parameters on neurons in the amygdala [197,198]. Not surprisingly, then, these chronic stress regimens have been shown to produce significant impairments of hippocampus-dependent (e.g., spatial learning) [14,92,104,139,181,185,211] and prefrontal cortex-dependent (e.g., attention set-shifting, reversal learning) memory [26,99], while enhancing performance on tasks that are dependent upon the amygdala (e.g., fear conditioning) [32, 167]. Additionally, the same chronic stress that leads to hypertrophy of cells in the amygdala increases the expression of anxiety-like behaviors in rats tested in the elevated plus maze [197,198].

It is important to note that the effects of chronic stress on hippocampal [31,181] and prefrontal cortex [151] morphology have been found to be reversible – that is, the dendrites re-grew when the stress was discontinued. This was not the case, however, for the effects of chronic stress on amygdala morphology or the amygdala-mediated expression of anxiety-like behavior [199]. Additional work showed that the effects of chronic stress on these brain regions were mediated by an interaction between glucocorticoids and NMDA receptor activity. Thus, chronic administration of corticosterone mimicked the effects of chronic stress on hippocampal [109,181,208] and prefrontal cortex morphology [204], and the stress-induced dendritic retraction observed in the hippocampus was blocked by steroid synthesis inhibitors [107], as well as NMDA receptor antagonists [107] and agents that significantly reduced extracellular levels of glutamate (e.g., phenytoin) [108,201]. These findings resonate with research in depressed patients, which indicates that these individuals have an overactive HPA axis [57,138] and abnormal brain glutamatergic levels [80,94,166].

TIANEPTINE PREVENTS CHRONIC STRESS-INDUCED STRUCTURAL AND FUNCTIONAL CHANGES IN THE HIPPOCAMPUS, PREFRONTAL CORTEX AND AMYGDALA

Daily administration of tianeptine blocks the chronic stress-induced reduction of hippocampal volume [34], as well as the retraction of CA3 dendrites in the hippocampus [31,105,202]. In contrast, the SSRIs fluoxetine and fluvoxamine were ineffective in preventing the stress-induced changes in CA3 morphology [105], suggesting that the effects of tianeptine and SSRIs may be mediated, at least in part, by different cellular and molecular mechanisms. Additional work has shown that tianeptine also prevents the effects of chronic stress on hippocampus-dependent learning and memory [30,104,212]. Investigators have yet to determine whether or not tianeptine prevents the effects of chronic stress on prefrontal cortex morphology. Considering tianeptine’s ability to block the effects of chronic stress on hippocampal structure and function, it is likely that tianeptine would exert positive effects on the prefrontal cortex, as well. Tianeptine also blocks the effects of chronic stress on hypertrophy of amygdala dendritic arbors, as well as the concurrent enhancement of anxiety-like behavior accompanying chronic stress [144]. These findings may be relevant to other work reporting that chronic tianeptine treatment reduced the expression of auditory fear conditioning, an amygdala-dependent task [23].

MECHANISMS UNDERLYING TIANEPTINE’S EFFECTS ON CHRONIC STRESS-INDUCED CHANGES IN BRAIN STRUCTURE AND FUNCTION

The hippocampus is one of only two brain regions in the adult mammalian brain that produces new neurons, a process known as neurogenesis [48]. Although the functional role of neurogenesis has remained a highly debated topic, studies have provided evidence linking hippocampal neurogenesis with hippocampus-dependent learning [59,177]. In addition, several researchers have hypothesized that the pathogenesis of depression involves impaired hippocampal neurogenesis [42,46,65,69,169]. Accordingly, in animal models, chronic stress significantly reduces hippocampal neurogenesis [60, 63,64,143,165,172] and increases apoptotic cell death in the hippocampus and temporal cortex [63,103,210]. Clinically effective antidepressants, including tianeptine, prevent the effects of chronic stress on hippocampal neurogenesis [34, 42,172]. Tianeptine has also been reported to block the chronic stress-induced increase in apoptotic cell death in the temporal cortex [102], which may be related to its prevention of the chronic stress-induced reduction of cerebral metabolites associated with neuronal viability (e.g., N-acetyl-aspartate) [34].

Neurotrophic factors are significant regulators of cell survival and proliferation, thus making them vitally important for the process of neurogenesis [68]. Some of the most extensively characterized neurotrophic factors include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and neurotrophin-4 (NT-4). Numerous studies have shown that acute and chronic stress significantly reduce neurotrophic factor levels [172,179,180, 190], but most studies have focused on the stress-induced reduction of BDNF levels in the hippocampus [3,10,90,131, 146,155,161,162,165,170]. This effect has become the center of attention, at least in part, because several studies have reported significantly reduced levels of serum and hippocampal BDNF in depressed patients [6,75,176]. BDNF knock-out mice have been reported to exhibit morphological changes in the hippocampus that are comparable to those observed following exposure to chronic restraint stress [157]. Interestingly, investigators have shown that the efficacy of antidepressants in ameliorating behavioral symptoms of depression, in depressed patients and animal models of stress, depends on their ability to increase BDNF levels [6,25,29].

Tianeptine’s ability to prevent the effects of chronic stress on neurogenesis may involve blocking the stress-induced reduction of neurotrophic factor levels in the hippocampus [3]. Another study, although reporting no effect of stress or tianeptine on hippocampal BDNF, found that chronic tianeptine treatment significantly increased BDNF levels in the rat amygdala, independent of whether or not the rats were exposed to stress [157]. According to Reagan and colleagues, the amygdala may be the site of initiation of chronic stress-induced morphological changes in other brain regions, such as the hippocampus and prefrontal cortex [157,159]. In support of this hypothesis, clinical studies on depressed patients have reported that morphological changes in the amygdala precede those that are observed in the hippocampus [117]. Therefore, tianeptine’s effectiveness as antidepressant treatment may result from its stabilization of neurotrophin levels in the amygdala. Future studies should be conducted to examine this hypothesis.

STRESS, TIANEPTINE AND SYNAPTIC PLASTICITY IN THE HIPPOCAMPUS, PREFRONTAL CORTEX AND AMYGDALA

Researchers have also hypothesized that depression involves a disruption of long-term synaptic plasticity [36,121]. To indirectly address this issue, investigators have examined the effects of stress on long-term potentiation (LTP), a physiological model of learning and memory involving an enhancement of synaptic efficacy following high-frequency stimulation of afferent fibers [156]. Extensive work has shown that stress impairs the induction of LTP in the hippocampus and prefrontal cortex, while facilitating its induction in the amygdala [37,39,41,85]. The stress-induced modulation of synaptic plasticity has been shown to be mediated by interactions among glucocorticoids [89,110,115], glutamatergic NMDA receptors [84,116,148] and amygdala-induced modulation of hippocampal plasticity [1,2].

Tianeptine has been shown to block the stress-induced impairment of LTP in the hippocampus and prefrontal cortex, without interfering with the stress-induced enhancement of LTP in the basolateral amygdala (BLA) [72,164,173,196]. Tianeptine blocked the inhibitory effects of stress on hippocampal LTP and primed burst potentiation (PBP), a low-threshold form of LTP, when it was administered before or after the stress experience [173,196]. Other antidepressants, including some SSRIs, have also been reported to block the effects of stress on LTP in the hippocampus and prefrontal cortex, although these effects have been less significant and more transitory in nature [164].

MEMORY-PROTECTIVE EFFECTS OF TIANEPTINE

Tianeptine administration, under non-stress conditions, has been shown to increase the magnitude of synaptic plasticity (LTP and PB potentiation) in the hippocampal CA1 region [173,196]. This finding suggests that tianeptine should enhance learning and memory. Indeed, studies have shown that tianeptine enhances spontaneous alternation behavior, as well as performance on discrimination tasks in the T-maze and radial arm maze [70,123]. In contrast, the SSRI fluoxetine impaired performance on the radial arm maze discrimination task [70], a finding that is relevant to other work reporting that fluoxetine impairs the induction of LTP in hippocampal slices [173]. Recent work from our laboratory has shown that the acute administration of tianeptine immediately before training in the radial-arm water maze (RAWM) enhanced long-term (24 hr) spatial memory [130]. The doses of tianeptine used in this experiment (1-10 mg/kg) are the same doses that have been shown to enhance hippocampal LTP and PB potentiation [173,196], and to block the effects of chronic stress on hippocampal morphology and hippocampus-dependent learning and memory.

Extensive research has shown that acute stress impairs hippocampus-dependent learning and memory in humans and rodents [37,39,41,83,85]. We recently reported that tianeptine, but not the anxiolytic propranolol, blocked the predator stress-induced impairment of rat spatial memory in the RAWM [24]. Tianeptine prevented the effects of stress on memory without altering the stress-induced increase in glucocorticoids, which suggests that tianeptine’s memory-protective effects are independent of the stress-induced activation of the HPA axis. Moreover, the findings are consistent with in vivo electrophysiological studies reporting that tianeptine blocked the effects of stress on hippocampal LTP without affecting stress-induced increases in corticosterone levels in rats [173].

To extend this observation, we have tested whether tianeptine could prevent the stress-induced impairment of spatial memory in adrenalectomized (ADX) rats. If tianeptine’s mechanism of action is independent of the stress-induced increase in adrenal hormones, then tianeptine should prevent the effects of stress on hippocampus-dependent spatial memory in ADX rats. In this experiment, rats (250-275 g; Harlan Laboratories; Indianapolis, IN) underwent ADX or sham surgery, following previously-described methods [52,133]. The drinking water of ADX rats was composed of 0.9% saline with 25 mg/l of corticosterone to prevent any adverse effects of corticosterone depletion on their physiology. One week following surgery, the rats were injected intraperitoneally with tianeptine (10 mg/kg) or vehicle (0.9% saline, 1 ml/kg) and then, 30 min later, they were given 12 trials to learn the location of a hidden escape platform, which was placed at the end of one of six arms, in the RAWM [24, 38,140,142,211]. Arm entry errors (i.e., entries into arms that did not contain the hidden platform) served as an indicator of a rat’s memory for the hidden platform. Following training, the rats spent a 30 min delay period in their home cages (No Stress) or confined to a small plexiglas chamber near a cat (Stress) [24,142,168]. The 30 min delay period was terminated with a single memory test trial in the RAWM, which was followed by the collection of a 0.5 ml sample of tail blood for subsequent analysis of corticosterone levels [24, 142,168].

As can be seen in the memory test trial data on the left side of Fig. (1), the control (i.e., non-stressed) rats demonstrated excellent memory for the location of the hidden platform, independent of whether or not they had undergone ADX surgery. This finding indicates that adrenal hormones are not necessary for spatial learning in the water maze and successful retrieval of short-term (30 min) hippocampus-dependent memory. As shown on the right side of Fig. (1), water maze training increased serum corticosterone levels in sham-operated control rats, relative to control rats that had undergone the ADX procedure. We found that vehicle-treated, adrenal-intact and ADX rats that were exposed to the cat during the 30 min delay period displayed significantly impaired performance on the memory test trial. This finding supports the notion that increased levels of glucocorticoids do not underlie the rapid impairing effects of stress on hippocampus-dependent memory [140]. Most importantly, acute administration of tianeptine prior to water maze training prevented the stress-induced impairment of spatial memory in both ADX and sham-operated animals, without having any significant effect on serum corticosterone levels.

Fig. (1).

Fig. (1)

Pre-training administration of tianeptine blocked the effects of predator stress on spatial memory in adrenalectomized (ADX) and adrenal-intact (Sham) rats without affecting corticosterone levels. Arm entry errors from the 12 acquisition trials in the RAWM (data not shown) were analyzed with a mixed-model ANOVA. This analysis revealed significant main effects of trials, F(11,693) = 19.59, and drug, F(1,63) = 4.28, and a significant Surgery x Stress x Drug interaction, F(1,63) = 4.18 (p’s < 0.05). While all groups made significantly fewer arm entry errors as trials progressed, tianeptine led to significantly more arm entry errors than vehicle in all groups except for the stress-exposed ADX group. All other main effects and interactions were not significant. Arm entry errors from the 30 min memory test trial (left) were analyzed with a one-way ANOVA. This analysis revealed significant main effects of stress, F(1,63) = 20.64, and drug, F(1,63) = 9.22, as well as a significant Stress x Drug interaction, F(1,63) = 18.22 (p’s < 0.01). Vehicle-treated rats exposed to predator stress during the 30 min delay period made significantly more arm entry errors than control (i.e., unstressed) rats. The administration of tianeptine prior to training blocked this effect in both ADX and sham-operated animals. Serum corticosterone levels (right) were analyzed with a one-way ANOVA. This analysis revealed significant main effects of surgery, F(1,41) = 129.71, and stress, F(1,41) = 49.75, and a significant Surgery x Stress interaction, F(1,41) = 46.08 (p’s < 0.0001). Water maze training significantly increased corticosterone levels in sham-operated control rats, relative to ADX controls. Predator stress significantly increased corticosterone levels in sham-operated, but not ADX, rats, an effect that was independent of tianeptine treatment. For the water maze data, the dashed line at 2.5 errors indicates chance level of performance [40]. * = p < 0.05 relative to no stress groups and tianeptine-treated stress groups; β = p < 0.05 relative to stress groups and ADX-no stress groups; # = p < 0.05 relative to no stress groups and ADX-stress groups.

In conjunction with our prior work [24], these findings provide convincing evidence that tianeptine’s memory-protective effects are not accomplished via the modulation of stress-induced increases in glucocorticoid levels. This series of electrophysiological and behavioral experiments supports the hypothesis that tianeptine enables hippocampus-dependent information to be stored more efficiently, thereby protecting its retrieval from being disrupted by stress.

MECHANISMS UNDERLYING TIANEPTINE’S ANTIDEPRESSANT AND MEMORY-PROTECTIVE PROPERTIES

Although it initially appeared that tianeptine’s antidepressant action was attributable to its effects on serotonin reuptake [78,96], recent work indicates that its therapeutic effects may be more associated with its modulation of the glutamatergic system [18,77]. Glutamate is the primary excitatory neurotransmitter of the central nervous system, and one of its roles is to regulate calcium influx by acting on postsynaptic AMPA and NMDA receptors [160]. Studies have shown that depressed patients exhibit elevated glutamate levels in plasma, CSF and post-mortem brain samples, which supports current views implicating the dysregulation of glutamate transmission in the pathogenesis of depression [80,94,166].

Extensive work has implicated hyperactivity of the glutamatergic system in the deleterious effects of stress on brain structure and function. Experiments conducted primarily on the hippocampus have shown that stress significantly increases glutamate levels [7,100,101,125,159], inhibits glutamate uptake [209], increases the expression and binding of glutamate receptors [11,93,119] and increases calcium currents [73]. Accordingly, researchers have shown that administration of NMDA receptor antagonists blocks the effects of stress on behavioral, morphological and electrophysiological measures of hippocampal function [84,107,141].

Tianeptine appears to protect the hippocampus and prefrontal cortex from the deleterious effects of stress by normalizing the stress-induced modulation of glutamatergic activity. For instance, tianeptine blocked the stress-induced increase in NMDA channel currents, as well as the ratio of NMDA:non-NMDA receptor currents, in the CA3 region of the hippocampus [88]. Tianeptine also inhibited the acute stress-induced increase in extracellular levels of glutamate in the basolateral amygdala (BLA), while having no effect on the stress-induced increase in these levels in the central nuclei of the amygdala (CeA) [159]. Interestingly, as mentioned above, tianeptine had no effect on the stress-induced enhancement of LTP in the BLA [196]. This finding suggests that the stress-induced enhancement of LTP in the BLA may involve NMDA-independent forms of synaptic plasticity, such as voltage-gated calcium channel-dependent LTP [91].

In contrast to tianeptine, administration of the SSRI fluoxetine increased baseline and stress-induced levels of glutamate in the BLA and CeA [159]. This finding may explain why SSRIs are anxiogenic early in the treatment phase and exert therapeutic antidepressant and anxiolytic effects only after a substantial delay [23,58]. Moreover, investigators have shown that acute administration of the SSRI citalopram enhanced the acquisition of auditory fear conditioning, while chronic treatment with citalopram impaired the acquisition and expression of conditioned fear [23]. Acute treatment with tianeptine, in contrast, had no effect on auditory fear conditioning, but when given chronically, exerted effects comparable to those of citalopram. Thus, tianeptine demonstrates long-lasting anxiolytic and antidepressant effects that are similar to SSRIs, without the adverse acute effects typically found with these agents.

Tianeptine’s effect on glutamatergic activity in amygdala may play an important role in its ability to reverse the effects of chronic stress on amygdala morphology and the expression of anxiety-like behaviors. In addition to its glutamatergic modulation, tianeptine reduces the expression of corticotropin-releasing hormone (CRH) mRNA in the amygdala and the bed nucleus of the stria terminalis (BNST), a brain region that is highly innervated by amygdala fibers [86]. CRH neurotransmission in both of these regions has been implicated in the expression of anxiety-like behaviors, and several studies have reported significantly elevated CSF CRH levels in depressed patients [67,76,183]. If the amygdala is the site of the initiation of chronic stress-induced functional changes in other brain regions, such as the hippocampus and prefrontal cortex, then tianeptine’s ability to stabilize amygdala activity could underlie its widespread anti-stress effects.

Chronic stress has been shown to increase expression of the glutamate transporter, GLT-1, which is important for removing excess glutamate from synaptic regions [158]. This effect was specifically observed in the CA3 region of the hippocampus, the primary area exhibiting significant morphological alterations following chronic stress. Researchers have postulated that the up-regulation of GLT-1 levels in this region is a compensatory response to chronic elevations of extracellular glutamate levels. Importantly, tianeptine has been shown to block the stress-induced increase in hippocampal GLT-1 levels. In theory, tianeptine accomplishes this feat by normalizing stress-induced glutamate levels in the hippocampus, thereby removing the stimulus (i.e., excessive glutamate) which necessitates increased expression of GLT-1.

Despite its ability to normalize the stress-induced increase in NMDA receptor currents, tianeptine also increases basal excitatory synaptic transmission in hippocampal circuits, predominantly via enhancing AMPA EPSCs [88]. In addition to NMDA receptors, AMPA receptors play an important role in excitatory synaptic transmission and the induction of long-term synaptic plasticity [154]. Recent work has reported that tianeptine modulates the phosphorylation of AMPA receptor subunits in the hippocampus [186]. Other antidepressants, such as SSRIs and tricyclics, have been shown to increase phosphorylation of the Ser845 site on the glutamate receptor subunit 1 (GluR1) of hippocampal AMPA receptors [45,187]. Investigators found that chronic, but not acute, tianeptine treatment significantly increased phosphorylation of the Ser831 and Ser845 sites on the GluR1 of AMPA receptors in the CA3 region of the hippocampus [186]. Typically, phosphorylation of the Ser831 and Ser845 sites of AMPA receptors occurs via protein kinase A (PKA) and calcium/calmodulin-dependent protein kinase II (CaMKII) or protein kinase C (PKC), respectively, and potentiates AMPA currents in the hippocampus [9,163]. Thus, the tianeptine-mediated increase in the phosphorylation of the serine sites on the GluR1 of AMPA receptors could explain the finding of a tianeptine-induced enhancement of AMPA EPSCs in the study of Kole et al. [88], which may also be relevant toward understanding tianeptine’s effectiveness as an antidepressant.

Recent work has also reported that tianeptine has anticonvulsant properties. Uzbay and colleagues found that tianeptine reduced the intensity [27] and delayed the onset [192] of pentylenetetrazole-induced seizures in rodents. The latter effect was blocked by the administration of caffeine, a nonspecific adenosine receptor antagonist, and 8-cyclopentyl-1,3-dipropylxanthine, an A1 receptor-specific antagonist. However, administration of the A2 receptor-specific antagonist, 8-(3-chlorostyryl) caffeine, had no effect on the tianeptine-induced delay of seizure onset, suggesting that tianeptine’s anticonvulsant properties are dependent upon activation of A1 adenosine receptors. Since previous work has shown that activation of A1 adenosine receptors has anxiolytic effects [53,71,149,150], this specific category of adenosinergic receptors could be responsible, at least in part, for tianeptine’s anxiolytic effects in rodents [23,50,51,144] and in the depressed population [35,205].

SUMMARY AND CONCLUSIONS

Depression is a common mental disorder for which effective pharmacological treatments are lacking. Investigators have utilized animal models of depression to develop a better understanding of the neurobiological basis of this disorder, which could ultimately produce improved treatment options for the patient. We have reviewed the findings of preclinical research demonstrating that tianeptine prevents the deleterious effects of stress on physiology and behavior. Tianeptine prevents chronic stress-induced morphological changes in the hippocampus and amygdala and blocks the effects of acute stress on synaptic plasticity in the hippocampus and prefrontal cortex. We have also reviewed findings demonstrating that tianeptine has memory-protective properties, in which tianeptine-treated rats exhibited intact hippocampus-dependent memory despite their being exposed to powerful fear-provoking stressors. Tianeptine’s prevention of the adverse effects of stress on brain and behavior is likely to contribute to its effectiveness as a treatment for people suffering from major depressive disorder. Although the antidepressant effects of tianeptine in people have been obtained through chronic administration, studies on the acute effects of tianeptine provide researchers with important information regarding tianeptine’s mechanism of action and ways in which its use may be expanded in humans.

Tianeptine’s actions do not appear to involve the modulation of stress-induced changes in HPA activity. We previously reported that tianeptine blocked the stress-induced impairment of spatial memory without affecting the stress-induced increase in glucocorticoid levels. Here, we have found that tianeptine prevented the acute stress-induced impairment of spatial memory in adrenalectomized rats, thereby demonstrating conclusively that elevated levels of glucocorticoids are not necessary for acute stress to affect memory, nor are they involved in tianeptine’s protective actions on memory. More recent work has suggested that tianeptine’s antidepressant effects may be attributable to its normalization of the stress-induced alterations of glutamatergic neurotransmission [18,77]. This finding resonates with accumulating evidence that has implicated abnormal glutamate activity in the pathogenesis of depression. Other research has shown that tianeptine has anticonvulsant properties, which are dependent upon adenosine receptor activation. Given the involvement of adenosine receptors in anxiolytic effects on behavior, tianeptine’s antidepressant effects could also involve modulation of adenosinergic neurotransmitter systems.

In summary, tianeptine is a well-described antidepressant with effective actions against stress-induced sequelae of the nervous system. It is as effective as SSRIs in treating depression, produces fewer adverse side effects and reduces anxious symptoms associated with depression without the need for concomitant anxiolytic therapy [5,16,19,61,188]. It is therefore relevant to note that tianeptine ameliorates symptoms in people with post-traumatic stress disorder (PTSD) [134] and in recent work has been shown to block the effects of intense stress on behavior and cardiovascular systems in an animal model of PTSD [212]. Thus, the well-described antidepressant and memory protective properties of tianeptine indicate that, in addition to its effectiveness as a treatment in mood disorders, it potentially has broader applications, as in the treatment of anxiety.

ACKNOWLEDGEMENTS

The work reported in this manuscript was supported by a VA Merit Review Award and a grant from I.R.I. Servier. We thank Adam Campbell for his technical assistance in the surgical and behavioral components of this work.

REFERENCES

  • 1.Akirav I, Richter-Levin G. Biphasic modulation of hippocampal plasticity by behavioral stress and basolateral amygdala stimulation in the rat. J. Neurosci. 1999;19:10530–10535. doi: 10.1523/JNEUROSCI.19-23-10530.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Akirav I, Richter-Levin G. Mechanisms of amygdala modulation of hippocampal plasticity. J. Neurosci. 2002;22:9912–9921. doi: 10.1523/JNEUROSCI.22-22-09912.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Alfonso J, Frick LR, Silberman DM, Palumbo ML, Genaro AM, Frasch AC. Regulation of hippocampal gene expression is conserved in two species subjected to different stressors and antidepressant treatments. Biol. Psychiatry. 2006;59:244–251. doi: 10.1016/j.biopsych.2005.06.036. [DOI] [PubMed] [Google Scholar]
  • 4.Anisman H, Matheson K. Stress, depression, and anhedonia: caveats concerning animal models. Neurosci. Biobehav. Rev. 2005;29:525–546. doi: 10.1016/j.neubiorev.2005.03.007. [DOI] [PubMed] [Google Scholar]
  • 5.Atmaca M, Kuloglu M, Tezcan E, Buyukbayram A. Switching to tianeptine in patients with antidepressant-induced sexual dysfunction. Hum. Psychopharmacol. Clin. Exp. 2003;18:277–280. doi: 10.1002/hup.479. [DOI] [PubMed] [Google Scholar]
  • 6.Aydemir C, Yalcin ES, Aksaray S, Kisa C, Yildirim SG, Uzbay T, Goka E. Brain-derived neurotrophic factor (BDNF) changes in the serum of depressed women. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2006;30:1256–1260. doi: 10.1016/j.pnpbp.2006.03.025. [DOI] [PubMed] [Google Scholar]
  • 7.Bagley J, Moghaddam B. Temporal dynamics of glutamate efflux in the prefrontal cortex and in the hippocampus following repeated stress: effects of pretreatment with saline or diazepam. Neuroscience. 1997;77:65–73. doi: 10.1016/s0306-4522(96)00435-6. [DOI] [PubMed] [Google Scholar]
  • 8.Barrett B, Byford S, Knapp M. Evidence of cost-effective treatments for depression: a systematic review. J. Affect. Disord. 2005;84:1–13. doi: 10.1016/j.jad.2004.10.003. [DOI] [PubMed] [Google Scholar]
  • 9.Barria A, Muller D, Derkach V, Griffith LC, Soderling TR. Regulatory phosphorylation of AMPA-type glutamate receptors by CaM-KII during long-term potentiation. Science. 1997;276:2042–2045. doi: 10.1126/science.276.5321.2042. [DOI] [PubMed] [Google Scholar]
  • 10.Barrientos RM, Sprunger DB, Campeau S, Higgins EA, Watkins LR, Rudy JW, Maier SF. Brain-derived neurotrophic factor mRNA downregulation produced by social isolation is blocked by intrahippocampal interleukin-1 receptor antagonist. Neuroscienc. 2003;121:847–853. doi: 10.1016/s0306-4522(03)00564-5. [DOI] [PubMed] [Google Scholar]
  • 11.Bartanusz V, Aubry JM, Pagliusi S, Jezova D, Baffi J, Kiss JZ. Stress-induced changes in messenger RNA levels of N-methyl-D-aspartate and AMPA receptor subunits in selected regions of the rat hippocampus and hypothalamus. Neuroscience. 1995;66:247–252. doi: 10.1016/0306-4522(95)00084-v. [DOI] [PubMed] [Google Scholar]
  • 12.Berton O, Nestler EJ. New approaches to antidepressant drug discovery: beyond monoamines. Nat. Rev. Neurosci. 2006;7:137–151. doi: 10.1038/nrn1846. [DOI] [PubMed] [Google Scholar]
  • 13.Blumenfeld RS, Ranganath C. Prefrontal cortex and long-term memory encoding: an integrative review of findings from neuropsychology and neuroimaging. Neuroscientist. 2007;13:280–291. doi: 10.1177/1073858407299290. [DOI] [PubMed] [Google Scholar]
  • 14.Bodnoff SR, Humphreys AG, Lehman JC, Diamond DM, Rose GM, Meaney MJ. Enduring effects of chronic corticosterone treatment on spatial learning, synaptic plasticity, and hippocampal neuropathology in young and mid-aged rats. J. Neurosci. 1995;15:61–69. doi: 10.1523/JNEUROSCI.15-01-00061.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bondy B. Common genetic factors for depression and cardiovascular disease. Dialogues Clin. Neurosci. 2007;9:19–28. doi: 10.31887/DCNS.2007.9.1/bbondy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bonierbale M, Lancon C, Tignol J. The ELIXIR study: evaluation of sexual dysfunction in 4557 depressed patients in France. Curr. Med. Res. Opin. 2003;19:114–124. doi: 10.1185/030079902125001461. [DOI] [PubMed] [Google Scholar]
  • 17.Bradley RG, Binder EB, Epstein MP, Tang Y, Nair HP, Liu W, Gillespie CF, Berg T, Evces M, Newport DJ, Stowe ZN, Heim CM, Nemeroff CB, Schwartz A, Cubells JF, Ressler KJ. Influence of child abuse on adult depression: moderation by the corticotropin-releasing hormone receptor gene. Arch. Gen. Psychiatry. 2008;65:190–200. doi: 10.1001/archgenpsychiatry.2007.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Brink CB, Harvey BH, Brand L. Tianeptine: a novel atypical antidepressant that may provide new insights into the biomolecular basis of depression. Recent Patents CNS Drug Discov. 2006;1:29–41. doi: 10.2174/157488906775245327. [DOI] [PubMed] [Google Scholar]
  • 19.Brion S, Audrain S, De Bodinat C. [Major depressive episodes in patients over 70 years of age. Evaluation of the efficiency and acceptability of tianeptine and mianserin] Presse Med. 1996;25:461–468. [PubMed] [Google Scholar]
  • 20.Broadbent NJ, Squire LR, Clark RE. Spatial memory, recognition memory, and the hippocampus. Proc. Natl. Acad. Sci. USA. 2004;101:14515–14520. doi: 10.1073/pnas.0406344101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Broadbent NJ, Squire LR, Clark RE. Reversible hippocampal lesions disrupt water maze performance during both recent and remote memory tests. Learn. Mem. 2006;13:187–191. doi: 10.1101/lm.134706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bunney WE, Jr., Davis JM. Norepinephrine in depressive reactions. A review. Arch. Gen. Psychiatry. 1965;13:483–494. doi: 10.1001/archpsyc.1965.01730060001001. [DOI] [PubMed] [Google Scholar]
  • 23.Burghardt NS, Sullivan GM, McEwen BS, Gorman JM, LeDoux JE. The selective serotonin reuptake inhibitor citalopram increases fear after acute treatment but reduces fear with chronic treatment: A comparison with tianeptine. Biol. Psychiatry. 2004;55:1171–1178. doi: 10.1016/j.biopsych.2004.02.029. [DOI] [PubMed] [Google Scholar]
  • 24.Campbell AM, Park CR, Zoladz PR, Munoz C, Fleshner M, Diamond DM. Pre-training administration of tianeptine, but not propranolol, protects hippocampus-dependent memory from being impaired by predator stress. Eur. Neuropsychopharmacol. 2008;18:87–98. doi: 10.1016/j.euroneuro.2007.04.004. [DOI] [PubMed] [Google Scholar]
  • 25.Castren E. Neurotrophic effects of antidepressant drugs. Curr. Opin. Pharmacol. 2004;4:58–64. doi: 10.1016/j.coph.2003.10.004. [DOI] [PubMed] [Google Scholar]
  • 26.Cerqueira JJ, Mailliet F, Almeida OF, Jay TM, Sousa N. The prefrontal cortex as a key target of the maladaptive response to stress. J. Neurosci. 2007;27:2781–2787. doi: 10.1523/JNEUROSCI.4372-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ceyhan M, Kayir H, Uzbay IT. Investigation of the effects of tianeptine and fluoxetine on pentylenetetrazole-induced seizures in rats. J. Psychiatr. Res. 2005;39:191–196. doi: 10.1016/j.jpsychires.2004.06.002. [DOI] [PubMed] [Google Scholar]
  • 28.Chamba G, Lemoine P, Flachaire E, Ferry N, Quincy C, Sassard J, Ferber C, Mocaer E, Kamoun A, Renaud B. Increased serotonin platelet uptake after tianeptine administration in depressed patients. Biol. Psychiatry. 1991;30:609–617. doi: 10.1016/0006-3223(91)90030-p. [DOI] [PubMed] [Google Scholar]
  • 29.Chen B, Dowlatshahi D, MacQueen GM, Wang JF, Young LT. Increased hippocampal BDNF immunoreactivity in subjects treated with antidepressant medication. Biol. Psychiatry. 2001;50:260–265. doi: 10.1016/s0006-3223(01)01083-6. [DOI] [PubMed] [Google Scholar]
  • 30.Conrad CD, Galea LA, Kuroda Y, McEwen BS. Chronic stress impairs rat spatial memory on the Y maze, and this effect is blocked by tianeptine pretreatment. Behav. Neurosci. 1996;110:1321–1334. doi: 10.1037//0735-7044.110.6.1321. [DOI] [PubMed] [Google Scholar]
  • 31.Conrad CD, LeDoux JE, Magarinos AM, McEwen BS. Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy. Behav. Neurosci. 1999;113:902–913. doi: 10.1037//0735-7044.113.5.902. [DOI] [PubMed] [Google Scholar]
  • 32.Conrad CD, Magarinos AM, LeDoux JE, McEwen BS. Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy. Behav. Neurosci. 1999;113:902–913. doi: 10.1037//0735-7044.113.5.902. [DOI] [PubMed] [Google Scholar]
  • 33.Cook SC, Wellman CL. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. J. Neurobiol. 2004;60:236–248. doi: 10.1002/neu.20025. [DOI] [PubMed] [Google Scholar]
  • 34.Czeh B, Michaelis T, Watanabe T, Frahm J, de Biurrun G, Van Kampen M, Bartolomucci A, Fuchs E. Stress-induced changes in cerebral metabolites, hippocampal volume, and cell proliferation are prevented by antidepressant treatment with tianeptine. Proc. Natl. Acad. Sci. USA. 2001;98:12796–12801. doi: 10.1073/pnas.211427898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Defrance R, Marey C, Kamoun A. Antidepressant and anxiolytic activities of tianeptine: an overview of clinical trials. Clin. Neuropharmacol. 1988;11(Suppl 2):S74–S82. [PubMed] [Google Scholar]
  • 36.Diamond DM, Campbell A, Park CR, Vouimba RM. Preclinical research on stress, memory, and the brain in the development of pharmacotherapy for depression. Eur. Neuropsychopharmacol. 2004;14(Suppl 5):S491–S495. doi: 10.1016/j.euroneuro.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 37.Diamond DM, Campbell AM, Park CR, Halonen J, Zoladz PR. The temporal dynamics model of emotional memory processing: a synthesis on the neurobiological basis of stress-induced amnesia, flashbulb and traumatic memories, and the Yerkes-Dodson Law. Neural. Plast. 2007;2007:60803. doi: 10.1155/2007/60803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Diamond DM, Campbell AM, Park CR, Woodson JC, Conrad CD, Bachstetter AD, Mervis RF. Influence of predator stress on the consolidation versus retrieval of long-term spatial memory and hippocampal spinogenesis. Hippocampus. 2006;16:571–576. doi: 10.1002/hipo.20188. [DOI] [PubMed] [Google Scholar]
  • 39.Diamond DM, Park CR, Campbell AM, Woodson JC. Competitive interactions between endogenous LTD and LTP in the hippocampus underlie the storage of emotional memories and stress-induced amnesia. Hippocampus. 2005;15:1006–1025. doi: 10.1002/hipo.20107. [DOI] [PubMed] [Google Scholar]
  • 40.Diamond DM, Park CR, Heman KL, Rose GM. Exposing rats to a predator impairs spatial working memory in the radial arm water maze. Hippocampus. 1999;9:542–552. doi: 10.1002/(SICI)1098-1063(1999)9:5<542::AID-HIPO8>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
  • 41.Diamond DM, Park CR, Woodson JC. Stress generates emotional memories and retrograde amnesia by inducing an endogenous form of hippocampal LTP. Hippocampus. 2004;14:281–291. doi: 10.1002/hipo.10186. [DOI] [PubMed] [Google Scholar]
  • 42.Dranovsky A, Hen R. Hippocampal neurogenesis: regulation by stress and antidepressants. Biol. Psychiatry. 2006;59:1136–1143. doi: 10.1016/j.biopsych.2006.03.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Drevets WC. Neuroimaging studies of mood disorders. Biol. Psychiatry. 2000;48:813–829. doi: 10.1016/s0006-3223(00)01020-9. [DOI] [PubMed] [Google Scholar]
  • 44.Druss BG, Rask K, Katon WJ. Major depression, depression treatment and quality of primary medical care. Gen. Hosp. Psychiatry. 2008;30:20–25. doi: 10.1016/j.genhosppsych.2007.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Du J, Suzuki K, Wei Y, Wang Y, Blumenthal R, Chen Z, Falke C, Zarate CA Jr, Manji HK. The anticonvulsants lamotrigine, riluzole, and valproate differentially regulate AMPA receptor membrane localization: relationship to clinical effects in mood disorders. Neuropsychopharmacology. 2007;32:793–802. doi: 10.1038/sj.npp.1301178. [DOI] [PubMed] [Google Scholar]
  • 46.Duman RS. Depression: a case of neuronal life and death? Biol. Psychiatry. 2004;56:140–145. doi: 10.1016/j.biopsych.2004.02.033. [DOI] [PubMed] [Google Scholar]
  • 47.Ebmeier K, Rose E, Steele D. Cognitive impairment and fMRI in major depression. Neurotox. Res. 2006;10:87–92. doi: 10.1007/BF03033237. [DOI] [PubMed] [Google Scholar]
  • 48.Ehninger D, Kempermann G. Neurogenesis in the adult hippocampus. Cell Tissue Res. 2008;331:243–250. doi: 10.1007/s00441-007-0478-3. [DOI] [PubMed] [Google Scholar]
  • 49.Eichenbaum H. Hippocampus: cognitive processes and neural representations that underlie declarative memory. Neuron. 2004;44:109–120. doi: 10.1016/j.neuron.2004.08.028. [DOI] [PubMed] [Google Scholar]
  • 50.File SE, Andrews N, Al Farhan M. Anxiogenic responses of rats on withdrawal from chronic ethanol treatment: effects of tianeptine. Alcohol Alcohol. 1993;28:281–286. [PubMed] [Google Scholar]
  • 51.File SE, Mabbutt PS. Effects of tianeptine in animal-models of anxiety and on learning and memory. Drug Dev. Res. 1991;23:47–56. [Google Scholar]
  • 52.Fleshner M, Campisi J, Amiri L, Diamond DM. Cat exposure induces both intra- and extracellular Hsp72: the role of adrenal hormones. Psychoneuroendocrinology. 2004;29:1142–1152. doi: 10.1016/j.psyneuen.2004.01.007. [DOI] [PubMed] [Google Scholar]
  • 53.Florio C, Prezioso A, Papaioannou A, Vertua R. Adenosine A1 receptors modulate anxiety in CD1 mice. Psychopharmacology (Berl) 1998;136:311–319. doi: 10.1007/s002130050572. [DOI] [PubMed] [Google Scholar]
  • 54.Frodl T, Meisenzahl E, Zetzsche T, Bottlender R, Born C, Groll C, Jager M, Leinsinger G, Hahn K, Moller HJ. Enlargement of the amygdala in patients with a first episode of major depression. Biol. Psychiatry. 2002;51:708–714. doi: 10.1016/s0006-3223(01)01359-2. [DOI] [PubMed] [Google Scholar]
  • 55.Fuchs E, Czeh B, Kole MH, Michaelis T, Lucassen PJ. Alterations of neuroplasticity in depression: the hippocampus and beyond. Eur. Neuropsychopharmacol. 2004;14(Suppl 5):S481–S490. doi: 10.1016/j.euroneuro.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 56.Gambarana C, Scheggi S, Tagliamonte A, Tolu P, De Montis MG. Animal models for the study of antidepressant activity. Brain Res. Brain Res. Protoc. 2001;7:11–20. doi: 10.1016/s1385-299x(00)00056-8. [DOI] [PubMed] [Google Scholar]
  • 57.Gillespie CF, Nemeroff CB. Hypercortisolemia and depression. Psychosom. Med. 2005;67(Suppl 1):S26–S28. doi: 10.1097/01.psy.0000163456.22154.d2. [DOI] [PubMed] [Google Scholar]
  • 58.Goldstein BJ, Goodnick PJ. Selective serotonin reuptake inhibitors in the treatment of affective disorders--III. Tolerability, safety and pharmacoeconomics. J. Psychopharmacol. 1998;12:S55–S87. doi: 10.1177/0269881198012003041. [DOI] [PubMed] [Google Scholar]
  • 59.Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ. Learning enhances adult neurogenesis in the hippocampal formation. Nat. Neurosci. 1999;2:260–265. doi: 10.1038/6365. [DOI] [PubMed] [Google Scholar]
  • 60.Gould E, Tanapat P. Stress and hippocampal neurogenesis. Biol. Psychiatry. 1999;46:1472–1479. doi: 10.1016/s0006-3223(99)00247-4. [DOI] [PubMed] [Google Scholar]
  • 61.Guelfi JD, Pichot P, Dreyfus JF. Efficacy of tianeptine in anxious-depressed patients: results of a controlled multicenter trial versus amitriptyline. Neuropsychobiology. 1989;22:41–48. doi: 10.1159/000118590. [DOI] [PubMed] [Google Scholar]
  • 62.Hayden EP, Dougherty LR, Maloney B, Olino TM, Sheikh H, Durbin CE, Nurnberger JI, Jr., Lahiri DK, Klein DN. Early-emerging cognitive vulnerability to depression and the serotonin transporter promoter region polymorphism. J. Affect Disord. 2008;107:227–230. doi: 10.1016/j.jad.2007.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Heine VM, Maslam S, Zareno J, Joels M, Lucassen PJ. Suppressed proliferation and apoptotic changes in the rat dentate gyrus after acute and chronic stress are reversible. Eur. J. Neurosci. 2004;19:131–144. doi: 10.1046/j.1460-9568.2003.03100.x. [DOI] [PubMed] [Google Scholar]
  • 64.Heine VM, Zareno J, Maslam S, Joels M, Lucassen PJ. Chronic stress in the adult dentate gyrus reduces cell proliferation near the vasculature and VEGF and Flk-1 protein expression. Eur. J. Neurosci. 2005;21:1304–1314. doi: 10.1111/j.1460-9568.2005.03951.x. [DOI] [PubMed] [Google Scholar]
  • 65.Henn FA, Vollmayr B. Neurogenesis and depression: etiology or epiphenomenon? Biol. Psychiatry. 2004;56:146–150. doi: 10.1016/j.biopsych.2004.04.011. [DOI] [PubMed] [Google Scholar]
  • 66.Hindmarch I. Expanding the horizons of depression: beyond the monoamine hypothesis. Hum. Psychopharmacol. Clin. Exp. 2001;16:203–218. doi: 10.1002/hup.288. [DOI] [PubMed] [Google Scholar]
  • 67.Holsboer F. The rationale for corticotropin-releasing hormone receptor (CRH-R) antagonists to treat depression and anxiety. J. Psychiatr. Res. 1999;33:181–214. doi: 10.1016/s0022-3956(98)90056-5. [DOI] [PubMed] [Google Scholar]
  • 68.Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu. Rev. Neurosci. 2001;24:677–736. doi: 10.1146/annurev.neuro.24.1.677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Jacobs BL, Praag H, Gage FH. Adult brain neurogenesis and psychiatry: a novel theory of depression. Mol. Psychiatry. 2000;5:262–269. doi: 10.1038/sj.mp.4000712. [DOI] [PubMed] [Google Scholar]
  • 70.Jaffard R, Mocaer E, Poignant JC, Micheau J, Marighetto A, Meunier M, Beracochea D. Effects of tianeptine on spontaneous alternation, simple and concurrent spatial discrimination learning and on alcohol-induced alternation deficits in mice. Behav. Pharmacol. 1991;2:37–46. [PubMed] [Google Scholar]
  • 71.Jain N, Kemp N, Adeyemo O, Buchanan P, Stone TW. Anxiolytic activity of adenosine receptor activation in mice. Br. J. Pharmacol. 1995;116:2127–2133. doi: 10.1111/j.1476-5381.1995.tb16421.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jay TM, Rocher C, Hotte M, Naudon L, Gurden H, Spedding M. Plasticity at hippocampal to prefrontal cortex synapses is impaired by loss of dopamine and stress: Importance for psychiatric diseases. Neurotox. Res. 2004;6:233–244. doi: 10.1007/BF03033225. [DOI] [PubMed] [Google Scholar]
  • 73.Joels M, Velzing E, Nair S, Verkuyl JM, Karst H. Acute stress increases calcium current amplitude in rat hippocampus: temporal changes in physiology and gene expression. Eur. J. Neurosci. 2003;18:1315–1324. doi: 10.1046/j.1460-9568.2003.02845.x. [DOI] [PubMed] [Google Scholar]
  • 74.Kanner AM. Structural MRI changes of the brain in depression. Clin. EEG. Neurosci. 2004;35:46–52. doi: 10.1177/155005940403500111. [DOI] [PubMed] [Google Scholar]
  • 75.Karege F, Perret G, Bondolfi G, Schwald M, Bertschy G, Aubry JM. Decreased serum brain-derived neurotrophic factor levels in major depressed patients. Psychiatry Res. 2002;109:143–148. doi: 10.1016/s0165-1781(02)00005-7. [DOI] [PubMed] [Google Scholar]
  • 76.Kasckow JW, Baker D, Geracioti TD Jr. Corticotropin-releasing hormone in depression and post-traumatic stress disorder. Peptides. 2001;22:845–851. doi: 10.1016/s0196-9781(01)00399-0. [DOI] [PubMed] [Google Scholar]
  • 77.Kasper S, McEwen BS. Neurobiological and clinical effects of the antidepressant tianeptine. CNS Drugs. 2008;22:15–26. doi: 10.2165/00023210-200822010-00002. [DOI] [PubMed] [Google Scholar]
  • 78.Kato G, Weitsch AF. Neurochemical profile of tianeptine, a new antidepressant drug. Clin. Neuropharmacol. 1988;11(Suppl 2):S43–S50. [PubMed] [Google Scholar]
  • 79.Kaut KP, Bunsey MD. The effects of lesions to the rat hippocampus or rhinal cortex on olfactory and spatial memory: retrograde and anterograde findings. Cogn Affect. Behav. Neurosci. 2001;1:270–286. doi: 10.3758/cabn.1.3.270. [DOI] [PubMed] [Google Scholar]
  • 80.Kendell SF, Krystal JH, Sanacora G. GABA and glutamate systems as therapeutic targets in depression and mood disorders. Expert. Opin. Ther. Targets. 2005;9:153–168. doi: 10.1517/14728222.9.1.153. [DOI] [PubMed] [Google Scholar]
  • 81.Kendler KS, Karkowski LM, Prescott CA. Causal relationship between stressful life events and the onset of major depression. Am. J. Psychiatry. 1999;156:837–841. doi: 10.1176/ajp.156.6.837. [DOI] [PubMed] [Google Scholar]
  • 82.Kilpatrick DG, Koenen KC, Ruggiero KJ, Acierno R, Galea S, Resnick HS, Roitzsch J, Boyle J, Gelernter J. The serotonin transporter genotype and social support and moderation of posttraumatic stress disorder and depression in hurricane-exposed adults. Am. J. Psychiatry. 2007;164:1693–1699. doi: 10.1176/appi.ajp.2007.06122007. [DOI] [PubMed] [Google Scholar]
  • 83.Kim JJ, Diamond DM. The stressed hippocampus, synaptic plasticity and lost memories. Nat. Rev. Neurosci. 2002;3:453–462. doi: 10.1038/nrn849. [DOI] [PubMed] [Google Scholar]
  • 84.Kim JJ, Foy MR, Thompson RF. Behavioral stress modifies hippocampal plasticity through N-methyl-D-aspartate receptor activation. Proc. Natl. Acad. Sci. USA. 1996;93:4750–4753. doi: 10.1073/pnas.93.10.4750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kim JJ, Song EY, Kosten TA. Stress effects in the hippocampus: synaptic plasticity and memory. Stress. 2006;9:1–11. doi: 10.1080/10253890600678004. [DOI] [PubMed] [Google Scholar]
  • 86.Kim SJ, Park SH, Choi SH, Moon BH, Lee KJ, Kang SW, Lee MS, Choi SH, Chun BG, Shin KH. Effects of repeated tianeptine treatment on CRF mRNA expression in non-stressed and chronic mild stress-exposed rats. Neuropharmacology. 2006;50:824–833. doi: 10.1016/j.neuropharm.2005.12.003. [DOI] [PubMed] [Google Scholar]
  • 87.Kole MH, Costoli T, Koolhaas JM, Fuchs E. Bidirectional shift in the cornu ammonis 3 pyramidal dendritic organization following brief stress. Neuroscience. 2004;125:337–347. doi: 10.1016/j.neuroscience.2004.02.014. [DOI] [PubMed] [Google Scholar]
  • 88.Kole MH, Swan L, Fuchs E. The antidepressant tianeptine persistently modulates glutamate receptor currents of the hippocampal CA3 commissural associational synapse in chronically stressed rats. Eur. J. Neurosci. 2002;16:807–816. doi: 10.1046/j.1460-9568.2002.02136.x. [DOI] [PubMed] [Google Scholar]
  • 89.Korz V, Frey JU. Stress-related modulation of hippocampal long-term potentiation in rats: Involvement of adrenal steroid receptors. J. Neurosci. 2003;23:7281–7287. doi: 10.1523/JNEUROSCI.23-19-07281.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kozlovsky N, Matar MA, Kaplan Z, Kotler M, Zohar J, Cohen H. Long-term down-regulation of BDNF mRNA in rat hippocampal CA1 subregion correlates with PTSD-like behavioural stress response. Int. J. Neuropsychopharmacol. 2007;10:741–758. doi: 10.1017/S1461145707007560. [DOI] [PubMed] [Google Scholar]
  • 91.Krugers HJ, Alfarez DN, Karst H, Parashkouhi K, van Gemert N, Joels M. Corticosterone shifts different forms of synaptic potentiation in opposite directions. Hippocampus. 2005;15:697–703. doi: 10.1002/hipo.20092. [DOI] [PubMed] [Google Scholar]
  • 92.Krugers HJ, Douma BRK, Andringa G, Bohus B, Korf J, Luiten PGM. Exposure to chronic psychosocial stress and corticosterone in the rat: Effects on spatial discrimination learning and hippocampal protein kinase C gamma immunoreactivity. Hippocampus. 1997;7:427–436. doi: 10.1002/(SICI)1098-1063(1997)7:4<427::AID-HIPO8>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 93.Krugers HJ, Koolhaas JM, Bohus B, Korf J. A single social stress-experience alters glutamate receptor-binding in rat hippocampal CA3 area. Neurosci. Lett. 1993;154:73–77. doi: 10.1016/0304-3940(93)90174-j. [DOI] [PubMed] [Google Scholar]
  • 94.Krystal JH, Sanacora G, Blumberg H, Anand A, Charney DS, Marek G, Epperson CN, Goddard A, Mason GF. Glutamate and GABA systems as targets for novel antidepressant and mood-stabilizing treatments. Mol. Psychiatry. 2002;7(Suppl 1):S71–S80. doi: 10.1038/sj.mp.4001021. [DOI] [PubMed] [Google Scholar]
  • 95.Kucia K, Malecki A, Gabryel B, Trzeciak HI. Effect of antidepressants on the phospholipase A(2) activity in plasma membranes of the rat brain cortex. Pol. J. Pharmacol. 2003;55:5–15. [PubMed] [Google Scholar]
  • 96.Labrid C, Mocaer E, Kamoun A. Neurochemical and pharmacological properties of tianeptine, a novel antidepressant. Br. J. Psychiatry. 1992;160:56–60. [PubMed] [Google Scholar]
  • 97.Lambert KG, Buckelew SK, Staffiso-Sandoz G, Gaffga S, Carpenter W, Fisher J, Kinsley CH. Activity-stress induces atrophy of apical dendrites of hippocampal pyramidal neurons in male rats. Physiol. Behav. 1998;65:43–49. doi: 10.1016/s0031-9384(98)00114-0. [DOI] [PubMed] [Google Scholar]
  • 98.Levinson DF. The genetics of depression: a review. Biol. Psychiatry. 2006;60:84–92. doi: 10.1016/j.biopsych.2005.08.024. [DOI] [PubMed] [Google Scholar]
  • 99.Liston C, Miller MM, Goldwater DS, Radley JJ, Rocher AB, Hof PR, Morrison JH, McEwen BS. Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting. J. Neurosci. 2006;26:7870–7874. doi: 10.1523/JNEUROSCI.1184-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lowy MT, Gault L, Yamamoto BK. Adrenalectomy attenuates stress-induced elevations in extracellular glutamate concentrations in the hippocampus. J. Neurochem. 1993;61:1957–1960. doi: 10.1111/j.1471-4159.1993.tb09839.x. [DOI] [PubMed] [Google Scholar]
  • 101.Lowy MT, Wittenberg L, Yamamoto BK. Effect of acute stress on hippocampal glutamate levels and spectrin proteolysis in young and aged rats. J. Neurochem. 1995;65:268–274. doi: 10.1046/j.1471-4159.1995.65010268.x. [DOI] [PubMed] [Google Scholar]
  • 102.Lucassen PJ, Fuchs E, Czeh B. Antidepressant treatment with tianeptine reduces apoptosis in the hippocampal dentate gyrus and temporal cortex. Biol. Psychiatry. 2004;55:789–796. doi: 10.1016/j.biopsych.2003.12.014. [DOI] [PubMed] [Google Scholar]
  • 103.Lucassen PJ, Vollmann-Honsdorf GK, Gleisberg M, Czeh B, de Kloet ER, Fuchs E. Chronic psychosocial stress differentially affects apoptosis in hippocampal subregions and cortex of the adult tree shrew. Eur. J. Neurosci. 2001;14:161–166. doi: 10.1046/j.0953-816x.2001.01629.x. [DOI] [PubMed] [Google Scholar]
  • 104.Luine V, Villegas M, Martinez C, McEwen BS. Repeated stress causes reversible impairments of spatial memory performance. Brain Res. 1994;639:167–170. doi: 10.1016/0006-8993(94)91778-7. [DOI] [PubMed] [Google Scholar]
  • 105.Magarinos AM, Deslandes A, McEwen BS. Effects of antidepressants and benzodiazepine treatments on the dendritic structure of CA3 pyramidal neurons after chronic stress. Eur. J. Pharmacol. 1999;371:113–122. doi: 10.1016/s0014-2999(99)00163-6. [DOI] [PubMed] [Google Scholar]
  • 106.Magarinos AM, McEwen BS. Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: comparison of stressors. Neuroscience. 1995;69:83–88. doi: 10.1016/0306-4522(95)00256-i. [DOI] [PubMed] [Google Scholar]
  • 107.Magarinos AM, McEwen BS. Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: involvement of glucocorticoid secretion and excitatory amino acid receptors. Neuroscience. 1995;69:89–98. doi: 10.1016/0306-4522(95)00259-l. [DOI] [PubMed] [Google Scholar]
  • 108.Magarinos AM, McEwen BS, Flugge G, Fuchs E. Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews. J. Neurosci. 1996;16:3534–3540. doi: 10.1523/JNEUROSCI.16-10-03534.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Magarinos AM, Orchinik M, McEwen BS. Morphological changes in the hippocampal CA3 region induced by non-invasive glucocorticoid administration: a paradox. Brain Res. 1998;809:314–318. doi: 10.1016/s0006-8993(98)00882-8. [DOI] [PubMed] [Google Scholar]
  • 110.Mailliet F, Qi H, Rocher C, Spedding M, Svenningsson P, Jay TM. Protection of stress-induced impairment of hippocampal/prefrontal LTP through blockade of glucocorticoid receptors: implication of MEK signaling. Exp. Neurol. 2008;211:593–596. doi: 10.1016/j.expneurol.2008.02.030. [DOI] [PubMed] [Google Scholar]
  • 111.Maletic V, Robinson M, Oakes T, Iyengar S, Ball SG, Russell J. Neurobiology of depression: an integrated view of key findings. Int. J. Clin. Pract. 2007;61:2030–2040. doi: 10.1111/j.1742-1241.2007.01602.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Mandelli L, Serretti A, Marino E, Pirovano A, Calati R, Colombo C. Interaction between serotonin transporter gene, catechol-O-methyltransferase gene and stressful life events in mood disorders. Int. J. Neuropsychopharmacol. 2007;10:437–447. doi: 10.1017/S1461145706006882. [DOI] [PubMed] [Google Scholar]
  • 113.Maoz H. Failure of first SSRI for depression--what is the next step? Isr. J. Psychiatry Relat. Sci. 2007;44:327–329. [PubMed] [Google Scholar]
  • 114.McArthur R, Borsini F. Animal models of depression in drug discovery: A historical perspective. Pharmacol. Biochem. Behav. 2006;84:436–452. doi: 10.1016/j.pbb.2006.06.005. [DOI] [PubMed] [Google Scholar]
  • 115.McEwen BS. Corticosteroids and hippocampal plasticity. Ann. N. Y. Acad. Sci. 1994;746:134–142. doi: 10.1111/j.1749-6632.1994.tb39223.x. [DOI] [PubMed] [Google Scholar]
  • 116.McEwen BS. Stress and hippocampal plasticity. Ann. Rev. Neurosci. 1999;22:105–122. doi: 10.1146/annurev.neuro.22.1.105. [DOI] [PubMed] [Google Scholar]
  • 117.McEwen BS. Mood disorders and allostatic load. Biol. Psychiatry. 2003;54:200–207. doi: 10.1016/s0006-3223(03)00177-x. [DOI] [PubMed] [Google Scholar]
  • 118.McEwen BS, Chattarji S. Molecular mechanisms of neuroplasticity and pharmacological implications: the example of tianeptine. Eur. Neuropsychopharmacol. 2004;14:S497–S502. doi: 10.1016/j.euroneuro.2004.09.008. [DOI] [PubMed] [Google Scholar]
  • 119.McEwen BS, Magarinos AM, Reagan LP. Structural plasticity and tianeptine: cellular and molecular targets. Eur. Psychiatry. 2002;17:318S–330S. doi: 10.1016/s0924-9338(02)00650-8. [DOI] [PubMed] [Google Scholar]
  • 120.McEwen BS, Olie JP. Neurobiology of mood, anxiety, and emotions as revealed by studies of a unique antidepressant: tianeptine. Mol. Psychiatry. 2005;10:525–537. doi: 10.1038/sj.mp.4001648. [DOI] [PubMed] [Google Scholar]
  • 121.McEwen BS, Olie JP. Neurobiology of mood, anxiety, and emotions as revealed by studies of a unique antidepressant: tianeptine. Mol. Psychiatry. 2005;10:525–537. doi: 10.1038/sj.mp.4001648. [DOI] [PubMed] [Google Scholar]
  • 122.McKittrick CR, Magarinos AM, Blanchard DC, Blanchard RJ, McEwen BS, Sakai RR. Chronic social stress reduces dendritic arbors in CA3 of hippocampus and decreases binding to serotonin transporter sites. Synapse. 2000;36:85–94. doi: 10.1002/(SICI)1098-2396(200005)36:2<85::AID-SYN1>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 123.Meneses A. Tianeptine: 5-HT uptake sites and 5-HT1-7 receptors modulate memory formation in an autoshaping Pavlovian/instrumental task. Neurosci. Biobehav. Rev. 2002;26:309–319. doi: 10.1016/s0149-7634(02)00005-2. [DOI] [PubMed] [Google Scholar]
  • 124.Mennini T, Mocaer E, Garattini S. Tianeptine, a selective enhancer of serotonin uptake in rat brain. Naunyn Schmiedebergs Arch. Pharmacol. 1987;336:478–482. doi: 10.1007/BF00169302. [DOI] [PubMed] [Google Scholar]
  • 125.Moghaddam B. Stress preferentially increases extraneuronal levels of excitatory amino acids in the prefrontal cortex: comparison to hippocampus and basal ganglia. J. Neurochem. 1993;60:1650–1657. doi: 10.1111/j.1471-4159.1993.tb13387.x. [DOI] [PubMed] [Google Scholar]
  • 126.Mondal S, Sharma VK, Das S, Goswami U, Gandhi A. Neuro-cognitive functions in patients of major depression. Indian J. Physiol. Pharmacol. 2007;51:69–75. [PubMed] [Google Scholar]
  • 127.Morilak DA, Frazer A. Antidepressants and brain monoaminergic systems: a dimensional approach to understanding their behavioural effects in depression and anxiety disorders. Int. J. Neuropsychopharmacol. 2004;7:193–218. doi: 10.1017/S1461145704004080. [DOI] [PubMed] [Google Scholar]
  • 128.Moser MB, Moser EI. Distributed encoding and retrieval of spatial memory in the hippocampus. J. Neurosci. 1998;18:7535–7542. doi: 10.1523/JNEUROSCI.18-18-07535.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Moses SN, Cole C, Ryan JD. Relational memory for object identity and spatial location in rats with lesions of perirhinal cortex, amygdala and hippocampus. Brain Res. Bull. 2005;65:501–512. doi: 10.1016/j.brainresbull.2005.02.029. [DOI] [PubMed] [Google Scholar]
  • 130.Munoz C, Park CR, Campbell AM, Diamond DM. The enhancement of long-term (24 hr) spatial memory by tianeptine, memantine and CPP supports the hypothesis that a reduction of NMDA receptor activity during learning will enhance memory. Soc. Neurosci. Abst. 2005;35:887.10. [Google Scholar]
  • 131.Murakami S, Imbe H, Morikawa Y, Kubo C, Senba E. Chronic stress, as well as acute stress, reduces BDNF mRNA expression in the rat hippocampus but less robustly. Neurosci. Res. 2005;53:129–139. doi: 10.1016/j.neures.2005.06.008. [DOI] [PubMed] [Google Scholar]
  • 132.Nemeroff CB, Vale WW. The neurobiology of depression: inroads to treatment and new drug discovery. J. Clin. Psychiatry. 2005;66(Suppl 7):5–13. [PubMed] [Google Scholar]
  • 133.Nguyen KT, Deak T, Owens SM, Kohno T, Fleshner M, Watkins LR, Maier SF. Exposure to acute stress induces brain interleukin-1beta protein in the rat. J. Neurosci. 1998;18:2239–2246. doi: 10.1523/JNEUROSCI.18-06-02239.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Onder E, Tural U, Aker T. A comparative study of fluoxetine, moclobemide, and tianeptine in the treatment of posttraumatic stress disorder following an earthquake. Eur. Psychiatry. 2006;21:174–179. doi: 10.1016/j.eurpsy.2005.03.007. [DOI] [PubMed] [Google Scholar]
  • 135.Pacher P, Kecskemeti V. Trends in the development of new antidepressants. Is there a light at the end of the tunnel? Curr. Med. Chem. 2004;11:925–943. doi: 10.2174/0929867043455594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Pagnin D, de Queinoz V, Pini S, Cassano GB. Efficacy of ECT in depression: a meta-analytic review. J. ECT. 2004;20:13–20. doi: 10.1097/00124509-200403000-00004. [DOI] [PubMed] [Google Scholar]
  • 137.Papakostas GI, Fava M, Thase ME. Treatment of SSRI-resistant depression: a meta-analysis comparing within- versus across-class switches. Biol. Psychiatry. 2008;63:699–704. doi: 10.1016/j.biopsych.2007.08.010. [DOI] [PubMed] [Google Scholar]
  • 138.Pariante CM, Miller AH. Glucocorticoid receptors in major depression: relevance to pathophysiology and treatment. Biol. Psychiatry. 2001;49:391–404. doi: 10.1016/s0006-3223(00)01088-x. [DOI] [PubMed] [Google Scholar]
  • 139.Park CR, Campbell AM, Diamond DM. Chronic psychosocial stress impairs learning and memory and increases sensitivity to yohimbine in adult rats. Biol. Psychiatry. 2001;50:994–1004. doi: 10.1016/s0006-3223(01)01255-0. [DOI] [PubMed] [Google Scholar]
  • 140.Park CR, Campbell AM, Woodson JC, Smith TP, Fleshner M, Diamond DM. Permissive influence of stress in the expression of a U-shaped relationship between serum corticosterone levels and spatial memory in rats. Dose Response. 2006;4:55–74. doi: 10.2203/dose-response.004.01.005.Park. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Park CR, Fleshner M, Diamond DM. An NMDA antagonist can impair, protect or have no effect on memory depending on training parameters and stress at the time of retrieval. Soc. Neurosci. Abst. 2004;34:776.22. [Google Scholar]
  • 142.Park CR, Zoladz PR, Conrad CD, Fleshner M, Diamond DM. Acute predator stress impairs the consolidation and retrieval of hippocampus-dependent memory in male and female rats. Learn. Mem. 2008;15:271–280. doi: 10.1101/lm.721108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Pham K, Nacher J, Hof PR, McEwen BS. Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus. Eur. J. Neurosci. 2003;17:879–886. doi: 10.1046/j.1460-9568.2003.02513.x. [DOI] [PubMed] [Google Scholar]
  • 144.Pillai AG, Munoz C, Chattarji S. The antidepressant tianeptine prevents the dendritic hypertrophy in the amygdala and increase in anxiety induced by chronic stress in the rat. Soc. Neurosci. Abst. 2004;34:762.1. [Google Scholar]
  • 145.Pitra P, Tokarski K, Grzegorzewska M, Hess G. Effects of repetitive administration of tianeptine, zinc hydroaspartate and electroconvulsive shock on the reactivity of 5-HT(7) receptors in rat hippocampus. Pharmacol. Rep. 2007;59:627–635. [PubMed] [Google Scholar]
  • 146.Pizarro JM, Lumley LA, Medina W, Robison CL, Chang WE, Alagappan A, Bah MJ, Dawood MY, Shah JD, Mark B, Kendall N, Smith MA, Saviolakis GA, Meyerhoff JL. Acute social defeat reduces neurotrophin expression in brain cortical and subcortical areas in mice. Brain Res. 2004;1025:10–20. doi: 10.1016/j.brainres.2004.06.085. [DOI] [PubMed] [Google Scholar]
  • 147.Pluijms EM, Birkenhager TK, Mulder PG, van den Broek WW. Influence of episode duration of major depressive disorder on response to electroconvulsive therapy. J Affect Disord. 2006;90:233–237. doi: 10.1016/j.jad.2005.11.010. [DOI] [PubMed] [Google Scholar]
  • 148.Popoli M, Gennarelli M, Racagni G. Modulation of synaptic plasticity by stress and antidepressants. Bipolar. Disord. 2002;4:166–182. doi: 10.1034/j.1399-5618.2002.01159.x. [DOI] [PubMed] [Google Scholar]
  • 149.Prediger RD, Batista LC, Takahashi RN. Adenosine A1 receptors modulate the anxiolytic-like effect of ethanol in the elevated plus-maze in mice. Eur. J. Pharmacol. 2004;499:147–154. doi: 10.1016/j.ejphar.2004.07.106. [DOI] [PubMed] [Google Scholar]
  • 150.Prediger RD, da Silva GE, Batista LC, Bittencourt AL, Takahashi RN. Activation of adenosine A1 receptors reduces anxiety-like behavior during acute ethanol withdrawal (hangover) in mice. Neuropsychopharmacology. 2006;31:2210–2220. doi: 10.1038/sj.npp.1301001. [DOI] [PubMed] [Google Scholar]
  • 151.Radley JJ, Rocher AB, Janssen WG, Hof PR, McEwen BS, Morrison JH. Reversibility of apical dendritic retraction in the rat medial prefrontal cortex following repeated stress. Exp. Neurol. 2005;196:199–203. doi: 10.1016/j.expneurol.2005.07.008. [DOI] [PubMed] [Google Scholar]
  • 152.Radley JJ, Rocher AB, Miller M, Janssen WG, Liston C, Hof PR, McEwen BS, Morrison JH. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cereb. Cortex. 2006;16:313–320. doi: 10.1093/cercor/bhi104. [DOI] [PubMed] [Google Scholar]
  • 153.Radley JJ, Sisti HM, Hao J, Rocher AB, McCall T, Hof PR, McEwen BS, Morrison JH. Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex. Neuroscience. 2004;125:1–6. doi: 10.1016/j.neuroscience.2004.01.006. [DOI] [PubMed] [Google Scholar]
  • 154.Rao VR, Finkbeiner S. NMDA and AMPA receptors: old channels, new tricks. Trends Neurosci. 2007;30:284–291. doi: 10.1016/j.tins.2007.03.012. [DOI] [PubMed] [Google Scholar]
  • 155.Rasmusson AM, Shi L, Duman R. Downregulation of BDNF mRNA in the hippocampal dentate gyrus after re-exposure to cues previously associated with footshock. Neuropsychopharmacology. 2002;27:133–142. doi: 10.1016/S0893-133X(02)00286-5. [DOI] [PubMed] [Google Scholar]
  • 156.Raymond CR. LTP forms 1, 2 and 3: different mechanisms for the "long" in long-term potentiation. Trends Neurosci. 2007;30:167–175. doi: 10.1016/j.tins.2007.01.007. [DOI] [PubMed] [Google Scholar]
  • 157.Reagan LP, Hendry RM, Reznikov LR, Piroli GG, Wood GE, McEwen BS, Grillo CA. Tianeptine increases brain-derived neurotrophic factor expression in the rat amygdala. Eur. J. Pharmacol. 2007;565:68–75. doi: 10.1016/j.ejphar.2007.02.023. [DOI] [PubMed] [Google Scholar]
  • 158.Reagan LP, Rosell DR, Wood GE, Spedding M, Munoz C, Rothstein J, McEwen BS. Chronic restraint stress up-regulates GLT-1 mRNA and protein expression in the rat hippocampus: reversal by tianeptine. Proc. Natl. Acad. Sci. USA. 2004;101:2179–2184. doi: 10.1073/pnas.0307294101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Reznikov LR, Grillo CA, Piroli GG, Pasumarthi RK, Reagan LP, Fadel J. Acute stress-mediated increases in extracellular glutamate levels in the rat amygdala: differential effects of antidepressant treatment. Eur. J. Neurosci. 2007;25:3109–3114. doi: 10.1111/j.1460-9568.2007.05560.x. [DOI] [PubMed] [Google Scholar]
  • 160.Riedel G, Platt B, Micheau J. Glutamate receptor function in learning and memory. Behav. Brain Res. 2003;140:1–47. doi: 10.1016/s0166-4328(02)00272-3. [DOI] [PubMed] [Google Scholar]
  • 161.Roceri M, Cirulli F, Pessina C, Peretto P, Racagni G, Riva MA. Postnatal repeated maternal deprivation produces age-dependent changes of brain-derived neurotrophic factor expression in selected rat brain regions. Biol. Psychiatry. 2004;55:708–714. doi: 10.1016/j.biopsych.2003.12.011. [DOI] [PubMed] [Google Scholar]
  • 162.Roceri M, Hendriks W, Racagni G, Ellenbroek BA, Riva MA. Early maternal deprivation reduces the expression of BDNF and NMDA receptor subunits in rat hippocampus. Mol. Psychiatry. 2002;7:609–616. doi: 10.1038/sj.mp.4001036. [DOI] [PubMed] [Google Scholar]
  • 163.Roche KW, O'Brien RJ, Mammen AL, Bernhardt J, Huganir RL. Characterization of multiple phosphorylation sites on the AMPA receptor GluR1 subunit. Neuron. 1996;16:1179–1188. doi: 10.1016/s0896-6273(00)80144-0. [DOI] [PubMed] [Google Scholar]
  • 164.Rocher C, Spedding M, Munoz C, Jay TM. Acute stress-induced changes in hippocampal/prefrontal circuits in rats: effects of antidepressants. Cereb. Cortex. 2004;14:224–229. doi: 10.1093/cercor/bhg122. [DOI] [PubMed] [Google Scholar]
  • 165.Rosenbrock H, Koros E, Bloching A, Podhorna J, Borsini F. Effect of chronic intermittent restraint stress on hippocampal expression of marker proteins for synaptic plasticity and progenitor cell proliferation in rats. Brain Res. 2005;1040:55–63. doi: 10.1016/j.brainres.2005.01.065. [DOI] [PubMed] [Google Scholar]
  • 166.Sanacora G, Gueorguieva R, Epperson CN, Wu YT, Appel M, Rothman DL, Krystal JH, Mason GF. Subtype-specific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Arch. Gen. Psychiatry. 2004;61:705–713. doi: 10.1001/archpsyc.61.7.705. [DOI] [PubMed] [Google Scholar]
  • 167.Sandi C, Merino JJ, Cordero MI, Touyarot K, Venero C. Effects of chronic stress on contextual fear conditioning and the hippocampal expression of the neural cell adhesion molecule, its polysialylation, and L1. Neuroscience. 2001;102:329–339. doi: 10.1016/s0306-4522(00)00484-x. [DOI] [PubMed] [Google Scholar]
  • 168.Sandi C, Woodson JC, Haynes VF, Park CR, Touyarot K, Lopez-Fernandez MA, Venero C, Diamond DM. Acute stress-induced impairment of spatial memory is associated with decreased expression of neural cell adhesion molecule in the hippocampus and prefrontal cortex. Biol. Psychiatry. 2005;57:856–864. doi: 10.1016/j.biopsych.2004.12.034. [DOI] [PubMed] [Google Scholar]
  • 169.Sapolsky RM. Is impaired neurogenesis relevant to the affective symptoms of depression? Biol Psychiatry. 2004;56:137–139. doi: 10.1016/j.biopsych.2004.04.012. [DOI] [PubMed] [Google Scholar]
  • 170.Scaccianoce S, Del Bianco P, Caricasole A, Nicoletti F, Catalani A. Relationship between learning, stress and hippocampal brain-derived neurotrophic factor. Neuroscience. 2003;121:825–828. doi: 10.1016/s0306-4522(03)00514-1. [DOI] [PubMed] [Google Scholar]
  • 171.Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am. J. Psychiatry. 1965;122:509–522. doi: 10.1176/ajp.122.5.509. [DOI] [PubMed] [Google Scholar]
  • 172.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. doi: 10.1097/FBP.0b013e3282ee2aa8. [DOI] [PubMed] [Google Scholar]
  • 173.Shakesby AC, Anwyl R, Rowan MJ. Overcoming the effects of stress on synaptic plasticity in the intact hippocampus: rapid actions of serotonergic and antidepressant agents. J. Neurosci. 2002;22:3638–3644. doi: 10.1523/JNEUROSCI.22-09-03638.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sheline YI. 3D MRI studies of neuroanatomic changes in unipolar major depression: the role of stress and medical comorbidity. Biol. Psychiatry. 2000;48:791–800. doi: 10.1016/s0006-3223(00)00994-x. [DOI] [PubMed] [Google Scholar]
  • 175.Sheline YI, Gado MH, Price JL. Amygdala core nuclei volumes are decreased in recurrent major depression. NeuroReport. 1998;9:2023–2028. doi: 10.1097/00001756-199806220-00021. [DOI] [PubMed] [Google Scholar]
  • 176.Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N, Kumakiri C, Nakazato M, Watanabe H, Shinoda N, Okada S, Iyo M. Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biol. Psychiatry. 2003;54:70–75. doi: 10.1016/s0006-3223(03)00181-1. [DOI] [PubMed] [Google Scholar]
  • 177.Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, Gould E. Neurogenesis in the adult is involved in the formation of trace memories. Nature. 2001;410:372–376. doi: 10.1038/35066584. [DOI] [PubMed] [Google Scholar]
  • 178.Siegle GJ, Carter CS, Thase ME. Use of FMRI to predict recovery from unipolar depression with cognitive behavior therapy. Am. J. Psychiatry. 2006;163:735–738. doi: 10.1176/ajp.2006.163.4.735. [DOI] [PubMed] [Google Scholar]
  • 179.Smith MA, Makino S, Kvetnansky R, Post RM. Effects of stress on neurotrophic factor expression in the rat brain. Ann. N. Y Acad. Sci. 1995;771:234–239. doi: 10.1111/j.1749-6632.1995.tb44684.x. [DOI] [PubMed] [Google Scholar]
  • 180.Smith MA, Makino S, Kvetnansky R, Post RM. Stress and glucocorticoids affect the expression of brain-derived neurotrophic factor and neurotrophin-3 mRNAs in the hippocampus. J. Neurosci. 1995;15:1768–1777. doi: 10.1523/JNEUROSCI.15-03-01768.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Sousa N, Lukoyanov NV, Madeira MD, Almeida OF, Paula-Barbosa MM. Reorganization of the morphology of hippocampal neurites and synapses after stress-induced damage correlates with behavioral improvement. Neuroscience. 2000;97:253–266. doi: 10.1016/s0306-4522(00)00050-6. [DOI] [PubMed] [Google Scholar]
  • 182.Squire LR, Stark CE, Clark RE. The medial temporal lobe. Annu. Rev. Neurosci. 2004;27:279–306. doi: 10.1146/annurev.neuro.27.070203.144130. [DOI] [PubMed] [Google Scholar]
  • 183.Strohle A, Holsboer F. Stress responsive neurohormones in depression and anxiety. Pharmacopsychiatry. 2003;36 (Suppl 3):S207–S214. doi: 10.1055/s-2003-45132. [DOI] [PubMed] [Google Scholar]
  • 184.Sullivan PF, Neale MC, Kendler KS. Genetic epidemiology of major depression: review and meta-analysis. Am. J Psychiatry. 2000;157:1552–1562. doi: 10.1176/appi.ajp.157.10.1552. [DOI] [PubMed] [Google Scholar]
  • 185.Sunanda Rao BSS, Raju TR. Chronic restraint stress impairs acquisition and retention of spatial memory task in rats. Curr. Sci. 2000;79:1581–1584. [Google Scholar]
  • 186.Svenningsson P, Bateup H, Qi H, Takamiya K, Huganir RL, Spedding M, Roth BL, McEwen BS, Greengard P. Involvement of AMPA receptor phosphorylation in antidepressant actions with special reference to tianeptine. Eur. J. Neurosci. 2007;26:3509–3517. doi: 10.1111/j.1460-9568.2007.05952.x. [DOI] [PubMed] [Google Scholar]
  • 187.Svenningsson P, Tzavara ET, Witkin JM, Fienberg AA, Nomikos GG, Greengard P. Involvement of striatal and extrastriatal DARPP-32 in biochemical and behavioral effects of fluoxetine (Prozac) Proc. Natl. Acad. Sci. USA. 2002;99:3182–3187. doi: 10.1073/pnas.052712799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Szadoczky E, Furedi J. Efficacy and acceptability of tianeptine and sertraline in the acute treatment phase of depression. Encephale. 2002;28:343–348. [PubMed] [Google Scholar]
  • 189.Thakur M, Blazer DG. Depression in long-term care. J Am. Med. Dir. Assoc. 2008;9:82–87. doi: 10.1016/j.jamda.2007.09.007. [DOI] [PubMed] [Google Scholar]
  • 190.Ueyama T, Kawai Y, Nemoto K, Sekimoto M, Tone S, Senba E. Immobilization stress reduced the expression of neurotrophins and their receptors in the rat brain. Neurosci. Res. 1997;28:103–110. doi: 10.1016/s0168-0102(97)00030-8. [DOI] [PubMed] [Google Scholar]
  • 191.Uzbay TI. Tianeptine: Potential influences on neuroplasticity and novel pharmacological effects. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2008;32:915–924. doi: 10.1016/j.pnpbp.2007.08.007. [DOI] [PubMed] [Google Scholar]
  • 192.Uzbay TI, Kayir H, Ceyhan M. Effects of tianeptine on onset time of pentylenetetrazole-induced seizures in mice: possible role of adenosine A1 receptors. Neuropsychopharmacology. 2007;32:412–416. doi: 10.1038/sj.npp.1301143. [DOI] [PubMed] [Google Scholar]
  • 193.Uzbekov MG, Misionzhnik EY, Maximova NM, Vertogradova OP. Biochemical profile in patients with anxious depression under the treatment with serotonergic antidepressants with different mechanisms of action. Hum. Psychopharmacol. 2006;21:109–115. doi: 10.1002/hup.749. [DOI] [PubMed] [Google Scholar]
  • 194.Vasic N, Walter H, Hose A, Wolf RC. Gray matter reduction associated with psychopathology and cognitive dysfunction in unipolar depression: A voxel-based morphometry study. J Affect Disord. 2008;109:107–116. doi: 10.1016/j.jad.2007.11.011. [DOI] [PubMed] [Google Scholar]
  • 195.Vaswani M, Linda FK, Ramesh S. Role of selective serotonin reuptake inhibitors in psychiatric disorders: a comprehensive review. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2003;27:85–102. doi: 10.1016/s0278-5846(02)00338-x. [DOI] [PubMed] [Google Scholar]
  • 196.Vouimba RM, Munoz C, Diamond DM. Differential effects of predator stress and the antidepressant tianeptine on physiological plasticity in the hippocampus and basolateral amygdala. Stress. 2006;9:29–40. doi: 10.1080/10253890600610973. [DOI] [PubMed] [Google Scholar]
  • 197.Vyas A, Jadhav S, Chattarji S. Prolonged behavioral stress enhances synaptic connectivity in the basolateral amygdala. Neuroscience. 2006;143:387–393. doi: 10.1016/j.neuroscience.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 198.Vyas A, Mitra R, Shankaranarayana Rao BS, Chattarji S. Chronic stress induces contrasting patterns of dendritic remodeling in hippocampal and amygdaloid neurons. J. Neurosci. 2002;22:6810–6818. doi: 10.1523/JNEUROSCI.22-15-06810.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Vyas A, Pillai AG, Chattarji S. Recovery after chronic stress fails to reverse amygdaloid neuronal hypertrophy and enhanced anxiety-like behavior. Neuroscience. 2004;128:667–673. doi: 10.1016/j.neuroscience.2004.07.013. [DOI] [PubMed] [Google Scholar]
  • 200.Wagstaff AJ, Ormrod D, Spencer CM. Tianeptine: a review of its use in depressive disorders. CNS Drugs. 2001;15:231–259. doi: 10.2165/00023210-200115030-00006. [DOI] [PubMed] [Google Scholar]
  • 201.Watanabe Y, Gould E, Cameron HA, Daniels DC, McEwen BS. Phenytoin prevents stress- and corticosterone-induced atrophy of CA3 pyramidal neurons. Hippocampus. 1992;2:431–435. doi: 10.1002/hipo.450020410. [DOI] [PubMed] [Google Scholar]
  • 202.Watanabe Y, Gould E, Daniels DC, Cameron H, McEwen BS. Tianeptine attenuates stress-induced morphological changes in the hippocampus. Eur. J. Pharmacol. 1992;222:157–162. doi: 10.1016/0014-2999(92)90830-w. [DOI] [PubMed] [Google Scholar]
  • 203.Watanabe Y, Gould E, McEwen BS. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Res. 1992;588:341–345. doi: 10.1016/0006-8993(92)91597-8. [DOI] [PubMed] [Google Scholar]
  • 204.Wellman CL. Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration. J. Neurobiol . 2001.;49:245–253. doi: 10.1002/neu.1079. [DOI] [PubMed] [Google Scholar]
  • 205.Wilde MI, Benfield P. Tianeptine. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in depression and coexisting anxiety and depression [published erratum appears in Drugs 1995; Jul 50(1): 156]. [Review] Drugs. 1995;49:411–439. doi: 10.2165/00003495-199549030-00007. [DOI] [PubMed] [Google Scholar]
  • 206.Wilhelm K, Siegel JE, Finch AW, Hadzi-Pavlovic D, Mitchell PB, Parker G, Schofield PR. The long and the short of it: associations between 5-HTT genotypes and coping with stress. Psychosom. Med. 2007;69:614–620. doi: 10.1097/PSY.0b013e31814cec64. [DOI] [PubMed] [Google Scholar]
  • 207.Winocur G, Moscovitch M, Caruana DA, Binns MA. Retrograde amnesia in rats with lesions to the hippocampus on a test of spatial memory. Neuropsychologia. 2005;43:1580–1590. doi: 10.1016/j.neuropsychologia.2005.01.013. [DOI] [PubMed] [Google Scholar]
  • 208.Woolley CS, Gould E, McEwen BS. Exposure to excess glucocorticoids alters dendritic morphology of adult hippocampal pyramidal neurons. Brain Res. 1990;531:225–231. doi: 10.1016/0006-8993(90)90778-a. [DOI] [PubMed] [Google Scholar]
  • 209.Yang CH, Huang CC, Hsu KS. Behavioral stress enhances hippocampal CA1 long-term depression through the blockade of the glutamate uptake. J. Neurosci. 2005;25:4288–4293. doi: 10.1523/JNEUROSCI.0406-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Zhao H, Xu H, Xu X, Young D. Predatory stress induces hippocampal cell death by apoptosis in rats. Neurosci. Lett. 2007;421:115–120. doi: 10.1016/j.neulet.2007.04.084. [DOI] [PubMed] [Google Scholar]
  • 211.Zoladz PR, Conrad CD, Fleshner M, Diamond DM. Acute episodes of predator exposure in conjunction with chronic social instability as an animal model of post-traumatic stress disorder. Stress. 2008;11:259–281. doi: 10.1080/10253890701768613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Zoladz PR, Halonen J, Munoz C, Diamond DM. Daily tianeptine treatment initiated after stress onset blocks the effects of chronic psychosocial stress on physiology and behavior in an animal model of PTSD. Soc. Neurosci. Abst. 2007;37:171–17. [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES