Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2018 Mar;16(3):234–270. doi: 10.2174/1570159X15666170818095105

Neurobiological Mechanisms of Stress Resilience and Implications for the Aged Population

Charlène Faye 1,*, Josephine C McGowan 2,*, Christine A Denny 3,4, Denis J David 1
PMCID: PMC5843978  PMID: 28820053

Abstract

Background:

Stress is a common reaction to an environmental adversity, but a dysregulation of the stress response can lead to psychiatric illnesses such as major depressive disorder (MDD), post-traumatic stress disorder (PTSD), and anxi-ety disorders. Yet, not all individuals exposed to stress will develop psychiatric disorders; those with enhanced stress resili-ence mechanisms have the ability to adapt successfully to stress without developing persistent psychopathology. Notably, the potential to enhance stress resilience in at-risk populations may prevent the onset of stress-induced psychiatric disorders. This novel idea has prompted a number of studies probing the mechanisms of stress resilience and how it can be manipulat-ed.

Methods:

Here, we review the neurobiological factors underlying stress resilience, with particular focus on the serotoninergic (5-HT), glutamatergic, and γ-Aminobutyric acid (GABA) systems, as well as the hy-pothalamic-pituitary axis (HPA) in rodents and in humans. Finally, we discuss stress resiliency in the context of aging, as the likelihood of mood disorders increases in older adults.

Results:

Interestingly, increased resiliency has been shown to slow aging and improved overall health and quality of life. Research in the neurobiology of stress resilience, particularly throughout the aging process, is a nascent, yet, burgeoning field.

Conclusion:

Overall, we consider the possible methods that may be used to induce resilient phenotypes, prophylactically in at-risk populations, such as in military personnel or in older MDD patients. Research in the mechanisms of stress resilience may not only elucidate novel targets for antidepressant treatments, but also provide novel insight about how to prevent these debilitating disorders from developing.

Keywords: Stress, resilience, aging, serotonin, glutamate, γ-aminobutyric acid, hypothalamic–pituitary–adrenal axis

1. INTRODUCTION

Brain disorders and particularly psychiatric disorders, comprising affective disorder [major depression (MDD), and anxiety disorders [panic disorders (PD) with and without agoraphobia, generalized anxiety disorder (GAD), panic bipolar disorder (BP), and cyclo- and dysthymic disorders] or social phobias, PTSD and obsessive-compulsive disorder (OCD)] are among the leading causes of disease and disability in the world. MDD was ranked as the fourth leading contributor to the global burden of disease and is the leading cause of disability worldwide as measured by Years Lived with Disability (YLDs) [1]. Mental disorders amounted to €240 billion and hence constitute 62% of the total annual cost of brain disorders in Europe [2]. In the United States, the total annual cost of depressive disorders was estimated to be $83.1 billion [3] and more than $42 billion for anxiety disorders [4], among the most prevalent mental disorders. Since the 2000s, an increasing prevalence of mental health problems, and consequently an increasing annual financial burden, has spurred an investigation into brain mechanisms of resilience that may protect against the development of psychiatric disorders.

In this review, we will first describe the psychosocial and neurobiological factors that contribute to stress resilience in rodents, nonhuman primates, and humans. The effects of stress throughout aging will then be discussed, as well as the potential risk factors and biological changes that increase susceptibility to mood disorders in older populations. There is an emerging field of research focused on resilience in the context of aging, as several studies have shown that the risk for developing mood disorders, particularly anxiety disorders, is increased with age [5-7]. Current behavioral methods to improve resilience in the aged will be considered, as well as suggestions for novel possibilities of fostering resilient phenotypes in this population and throughout life based on pre-clinical studies. Though these avenues of resilience enhancement are still in their infancy, there is great potential to refine these techniques to be administered to individuals who are most susceptible to stress, particularly as they age, in order to increase resiliency and prevent stress-induced disorders from occurring at any point in one’s lifetime.

2. DEFINITION OF RESILIENCY

An external, unexpected stressor can induce a normal physiological and behavioral stress response. This normal and basic response resulting in an organism’s adaptive capacity is characterized by activation of the hypothalamic-pituitary-adrenal (HPA) axis, with a relative peak of serum cortisol or corticosterone 30 minutes following stress exposure, followed by a quick recovery either in human or rodent, in 60-80 minutes after the stress [8, 9]. But prolonged exposure to stressful events (i.e., over weeks) can lead to chronic stress and persistent sequelae, depending on the ability of each person to adapt to potent stressors. Indeed, individuals perceive stressful events differently, and when the stress response becomes overactive, the recovery mechanisms fail to achieve, leading to increased susceptibility to stress. Yet, some individuals are less vulnerable to stress than others, and are deemed resilient. When faced with adversity, people with low resilience are at risk of mental illnesses such MDD or anxiety disorders. Conversely, people who are able to effectively develop well socially, mentally or physically despite exposure to stress or adversity demonstrate resiliency; elucidating the underlying mechanisms of this resilience may lend insight into methods to prevent mood disorders in susceptible individuals.

3. PSYCHOSOCIAL FACTOR-ASSOCIATED STRESS VULNERABILITY AND RESILIENCY

It is well established that the combination of genetic predisposition, familial heritage, socio-environmental factors, early life stress (ELS), and chronic illness or treatment largely determine vulnerability to psychiatric disorders such as MDD or anxiety disorders (Fig. 1). Whereas stressful life events such as the loss of a loved one are related to the development of MDD and GAD, combat-associated injury and traumatic experiences are associated with the development of PTSD. Since the majority of people who experience such stressful events do not develop psychopathology, increasing interest has been directed at potential resilience factors that may provide mental resistance. Developing novel therapeutics targeted at these factors is one avenue to decrease the incidence of mental illnesses.

The idea that increasing stress resilience could protect against the development of psychiatric disorders is appealing. Currently, five basic psychosocial resilience factors where listed based on enormous body of published research: positive emotions and optimism, cognitive flexibility, religion and spirituality, life meaning, social support and active coping style to be protective psychosocial factors that have been associated with stress resilience [10]. A study using a sample of 200 post-doctoral fellows indicated that resilience moderated the impact of stress on trait depressive and anxiety symptoms [11]. Besides revealing a positive association between positive emotions and resilience, this study also indicated that coping strategies partially mediated the link between positive emotions and resilience [11]. Moreover, findings from a prospective longitudinal study provide 
evidence that individual differences in stress resilience are predicted by both lower levels of negative emotions and higher levels of responsiveness in a positive direction before stressor exposure [12]. Overall, these findings suggest that coping strategies are essential to minimize the impact of stress.

The coping strategies that have been studied so far have focused their efforts on adolescent and youth populations. For instance, the Protection for Adolescent Depression Study (PADS) developed a vulnerability-resilience stress model for mood disorders, based on the principle that vulnerability and resilience in the face of stress inform an accurate prediction of psychopathology [13]. Using this model in a cohort of 283 healthy adolescents and 119 from a mood disorder clinic, they found two productive coping strategies-focusing on the positive and working hard to achieve-which moderate the impact of stress and predict lower levels of light-to-severe depression and suicidal behavior. Interestingly, girls who focused on the positive were more protected than boys against light-to-severe depression. Finally, self-discovery from the spirituality scale was the only protective factor in the community and reached the significance level only for boys in the clinical population. Thus, this study suggests targeting different factors for men and women to increase or restore protection in the presence of chronic stress conditions. The Early Prediction of Adolescent Depression (EPAD), a longitudinal study of parents with a history of recurrent MDD and their high-risk adolescent offspring, provide evidence that higher executive functioning, such as inhibition and mental flexibility, may confer protection against depressive symptoms to the adolescent offspring of depressed parents as compared to adolescents with poorer performance on these measures [14].

4. PERSONAL STRATEGIES FOR BUILDING STRESS RESILIENCE

Since it was reported that resilient individuals showed an increase in protective psychosocial factors, a group psychosocial resilience-training program was developed to promote resilience and psychosocial well-being in adults who are at risk for stress-induced depressive symptoms, but are otherwise generally healthy. The REsilience and Activity for every DaY (READY), targeting positive emotions, problem solving, life purpose and cognitive flexibility, demonstrated favorable intervention effects on measures of cognitive flexibility (acceptance and mindfulness), environmental mastery, positive emotions and personal growth, as well as moderate effects on measures of stress, self-acceptance, valued living and autonomy [15]. Other studies investigating the effectiveness of resilience training showed positive change occurring at post-intervention compared with baseline or control group [16, 17].

Further, regular exercise is frequently associated with general well-being and lower rates of mood disorders, probably in enhancing resilience to stress. Indeed, among young healthy men and women, individuals who exercise regularly showed smaller decline in positive affect after an acute stressor than those who did not reported regular physical exercise [18]. These findings suggest that regular exercisers are more resistant to the emotional effects of acute stress and might protect them against diseases related to chronic stress burden.

Overall, resilience is an active process that involves a set of neural and cellular mechanisms leading to avoid some of the negative consequences of extreme stress in individuals. The social environmental as well as the genetic and the biological backgrounds of an organism fundamentally influence the balance between risk and resilience to stress (Fig. 1). Nevertheless, the identification of biological resiliency factors for mood disorders is an important challenge facing biological psychiatry today and the key leading to the development of new pharmacologic treatments for preventing mental illnesses.

5. NEUROBIOLOGICAL FACTORS IN STRESS RESILIENCE

While there has been extensive research on psychosocial factors promoting susceptibility or resilience to psychiatric illnesses, studies published on genetic or biological mechanisms of human resilience are still in their infancy. The development of animal models is necessary not only to appreciate the various aspects of human pathology, such as physiological or behavioral changes, but also to understand the dynamics of therapeutic effects. It is therefore essential to evaluate mechanisms underlying stress resilience processes in animal models expressing symptoms of anxiety-depression pathologies.

6. RODENTS-ANIMAL MODEL OF STRESS RESILIENCY

Despite the complexity in designing and validating animal models in psychiatric research, the animal models [chronic mild stress (CMS), social defeat (SD), learned helplessness (LH), prenatal restraint stress, maternal separation and daily corticosterone administration (CORT)] we have chosen to use in this review were developed and are most often used to model affective disorders relevant to humans, as well as to screen a large panel of molecules with putative anxiolytic- and antidepressant-like activities. These models showed predictive validity in terms of depression or anxiety. However, even though they are inbred strains, a remarkable inter-individual variation in developing anxio-depressive symptoms has been noted. Recognizing the existence of this variation led to the classification of susceptible versus resilient animals that have varying degrees of depressive or anxiety. Work in animal models has suggested that stress resilience is mediated through active neurobiological processes, and has identified a number of protective psychosocial factors.

7. THE SEROTONERGIC SYSTEM

7.1. The Tryptophan Hydroxylase (TPH)

Tryptophan (TRP) is the precursor of peripheral and central serotonin (5-HT) synthesis. Thus, low TRP leads to low 5-HT levels, which contributes to mood state impairments and induces stress vulnerability (reviewed in [19]). Indeed, TRP-depleting drink administration-induced 5-HT depletion made healthy volunteers more susceptible to the effects of uncontrollable stress. These subjects showed more robust negative mood responses, particularly in sadness-related ratings, compared with TRP-supplemented group [20].

5-HT is produced from TRP via the rate-limiting enzyme TPH, which exists in two different isoforms. Whereas TPH1 mRNA is mainly expressed in peripheral tissues and in the pineal gland, the TPH2 isoform gene, recently discovered, is predominantly expressed in the brain. Although TPH2 is mainly responsible for the synthesis of brain 5-HT, studies to establish an association between the TPH2 gene and psychiatric disorders or stress resilience are still under investigation. One study demonstrated that in male Tph2 knockout (KO) mice, which completely lacked brain 5-HT synthesis while 5-HT raphe neurons remained functional, the 5-HT deficiency resulted in reducing anxiety-like behavior in the elevated plus maze (EPM) and open field (OF) [21] (Table 1). Discrepant results were reported for gene-by-environment interactions (GxE). A 3-week CMS was deleterious in Tph2 KO mice resulting in anxiety-like behavior in the EPM, but surprisingly, 5-HT dysfunction provided protective effect against chronic mild stress (CMS) in the OF [21] (Table 1).

From a clinical perspective, it appears counterintuitive that 5-HT deficiency results in anxiolytic effects, but similar results were found in resilient rats exposed to ELS [22]. Indeed, when MS15 rats, which are normally used as a model of decreased stress sensitivity, were subjected to a 10-day social defeat (SD) experience, the mean of Tph2 mRNA expression throughout the dorsal raphe nucleus (DRN) was decreased and conversely, MS180 rats, regarded as a model of vulnerability to depressive-like symptoms, elicited a two-fold increase in Tph2 mRNA as compared to MS15 rats [22] (Table 1). Interestingly, effects of ELS on Tph2 mRNA expression were only apparent in rats exposed to SD during adulthood [22]. These data suggested that MS15 resilient rats are resistant to SD-induced changes in Tph2 mRNA expression. However, it is important to note that this study lacks a complete behavioral analysis and that the mRNA results should be correlated with the measurement of social interaction in defeated rats. Moreover, these data contradict those obtained in highly stress-resilient (HSR) cynomolgus monkeys submitted to a 5-day exposure of a combined moderate stress paradigm. It has been shown that the HSR group had significantly higher average Tph2-positive cells than the stress sensitive (SS) group, suggesting that an upregulation of serotonin-related genes is protective against stress (Table 1). Unfortunately, no comparison with control animals was made in this study. In conclusion, TPH impairment leading to decreased brain 5-HT synthesis subsequently compromises one’s ability to cope with uncontrollable stress.

7.2. The Serotonin Transporter (SERT)

The 5-HT transporter (SERT) gene (SLC6A4), another actor of the neurotransmission homeostasis, may play a modulating role in rendering individuals vulnerable or resilient to stress. A common polymorphism, resulting in insertion or deletion, called, respectively, long (l) and short (s) forms have been identified in the SERT gene-linked polymorphic region (5-HTTLPR) of the SLC6A4. It has been argued that individuals carrying two copies of the long-allele (l/l) are relatively protected against mood disorder development [23, 24], and have increased cognitive emotional control and higher inhibition of negative information [25]. Individuals with 1 or 2 copies of the short-allele (s/l or s/s) are less resilient, and exhibited increased depressive symptoms following stressful life experiences [26-28]. Yet, these alleles are fundamentally neutral because in the absence of stress, no direct association between the SERT gene and depression was observed among members of the Dunedin Multidisciplinary Health and Development Study [26]. Thus, instead of viewing the s-allele as a risk-allele for vulnerability and the l-allele as a protective-allele for resiliency, functional polymorphisms in the 5-HTTLPR was only predisposing to mood disorders when interacting with environmental stress factors. For example, a study in U.S. Army soldiers with no prior war zone exposition who were scheduled to deploy to Iraq confirmed that s-allele carriers, interacted with levels of stressful exposure to predict the development of PTSD, anxiety, and depressive symptoms [29].

In order to model the human allelic variation in SERT function and for a better understanding of its behavioral consequences, heterozygous SERT-deficient mice were generated. In the absence of stress exposure, SERT KO offspring displayed lower levels of anxiety in the elevated zero maze (EZM), and in addition, developed a resilient phenotype to prenatal restraint stress (PS), compared to wild-type (WT) offspring [30] (Table 1). However, while, SERT KO offspring showed antidepressant-like behavior in the forced swim test (FST) compared to WT offspring, distance swum was decreased in restrained SERT KO offspring [30]. In this way, a decrease in SERT expression may protect against ELS. Indeed, SERT expression seems to be related to the levels of the vulnerability or resiliency of the animals. After isolating CMS-resilient animals, Couch and colleagues reported that unlike resilient mice, mRNA-encoding SERT was upregulated in vulnerable mice in the prefrontal cortex (PFC), striatum, and hippocampus, but not in the DRN [31] (Table 1). Similarly, Zurawek and collaborators found significantly decreased SERT protein level in the ventral tegmental area (VTA), but not in the DRN or PFC of CMS-resilient animals as compared to anhedonic and control rats [32] (Table 1).

With the idea still being that female macaques with different stress sensitivity exhibit different adaptive biological mechanisms from those set up in rodent in response to stress, the Bethea study also looked at the SERT-positive cell number in the DRN of HSR and SS female monkeys, but no difference was found [33]. Nevertheless, these results should be interpreted with caution, since this same team has earlier shown that although SERT mRNA are significantly lowered in non-stressed SS monkeys compared with non-stressed HSR monkeys in the caudal regions of the raphe [34], the ratio of the HSR/SS groups for the SERT gene was similar in the presence or absence of stress, implying that there is no adjustment in the 5-HT system after 5 days of moderate stress [33].

7.3. The Serotonin Receptors

5-HT released from serotoninergic neurons in the DRN throughout the brain modulates the acute stress response via its impact on a diverse group of serotonin receptors (for review, see [35]). Among the serotoninergic receptors, the 5-HT1A receptor (5-HTR1A), located either on the presynaptic (autoreceptor) or postsynaptic (heteroreceptor) terminals of

5-HT neurons, is one of the main actors responsible for the therapeutic delay of SSRIs. For this reason, 5-HTR1A has been a matter of interest for its role in the etiology of both anxiety and depression. Plenty of human genetic and imaging studies demonstrated that differences in regulation and binding potential of 5-HTR1A are associated with depression and anxiety (reviewed in [36]). While many studies in humans report an association of the G variant of the C(-1019)G 5-HTR1A promoter gene polymorphism with higher expression of 5-HTR1A and higher susceptibility to depression, no studies have focused on the effect of carrying the C(-1019) allele on vulnerability or resilience to mood disorders. Recently, Lemonde and colleagues postulated that carrying the C(-1019) allele, unlike the G(-1019), leads to a marked decrease in 5-HT1A mRNA, protein, and binding sites in the DRN, which could confer behavioral resilience to stressful situations [37]. Similarly, to provide a mechanistic model of one of the predicted consequences of the recently identified human Htr1a C(-1019)G polymorphism, a new strategy to manipulate 5-HT1A autoreceptors in raphe nuclei without affecting 5-HT1A heteroreceptors, generating mice with higher (1A-High) or lower (1A-Low) autoreceptor levels, has been performed [38]. Adult 5-HT1A-Low mice with 5-HT1A autoreceptor deficiency in DRN neurons as compared to 5-HT1A-High mice, exhibited reduced susceptibility to daily mild stressors (gavage) in the tail suspension test (TST) but not in the FST [38] (Table 1). However, in basal state, 1A-Low mice displayed less behavioral despair in the FST, suggesting an active coping mechanism to stress over time [38] (Table 1). Overall, these data suggest that reduced 5-HTR1A expression correlates with stress resiliency, even if opposite results in female monkeys were reported with higher 5-HT1A-positive cell numbers in the DRN of HSR individuals [33, 34] (Table 1).

5-HT2A receptors (5-HTR2A) were also implicated in pathophysiology of emotional disorders, since brain imaging studies revealed a decrease in 5-HTR2A binding in cortical [39-41] and hippocampal [42, 43] regions of depressed patients, and an increase in 5-HTR2A binding in the caudate nucleus of unmedicated patients with OCD [44]. Regarding the role of 5-HTR2A in stress resilience specifically, the data remain poor. A 3-fold increase in 5-HTR2A expression in the PFC of CMS-resilient mice relative to control mice have been reported, even though this change was also observed in the PFC of CMS-vulnerable mice, with no significant difference between resilient and vulnerable groups [31]. In addition, higher 5-HTR2A binding was also observed in the cortex of CMS rats [45]. The difference reported between CMS-resilient and CMS-vulnerable mice was an increase in 5-HTR2A expression in the striatum of susceptible compared to control mice, whereas resilient mice did not show any change [31]. However, Farhang and collaborators did not find any difference in levels of 5-HTR2A in either cerebral hemispheres of resilient rats to CMS [46], indicating that the connection between these receptors and stress resilience is not well-established.

7.4. The Serotonin Antidepressants in Preventing Stress 
Resilience

Antidepressants are typically used to treat existing depressive symptoms, but chronic antidepressant treatment also protects against subsequent depressive episodes [47]. Clinical practices recommend a 6 to 24 month antidepressant maintenance therapy to prevent recurrence, once an acute episode and the continuation treatment phase are over [48]. Placebo-controlled, double blind and randomized studies designed in patients with unipolar major depression demonstrated the efficacy of fluoxetine (20 mg/d) for the prevention of new depressive episodes, since treated patients were symptom-free for a longer period of time than patients assigned to placebo [49, 50]. In the same way, one-year treatment with paroxetine (20-30 mg/d) was effective in preventing the reappearance of depression for one year in depressed patient with more than two previous episodes in the preceding four years [51]. Citalopram (20 or 40 mg/d) treatment for 24 weeks in patients with MDD provided evidence for preventing relapse as compared to placebo [52]. Additionally, a 52-week treatment with sertraline in depressed patients conferred significantly greater prophylaxis against depression relapse and recurrence than did placebo [53]. In order to evaluate the efficacy of the different SSRIs in relapse prevention and prophylaxis of depression, Peselow and colleagues conducted a 3-year follow-up of MDD patients who had initially responded to SSRIs. Escitalopram and fluoxetine had the highest numerical prophylactic efficacy with 36% and 33.3% of the patients remained recurrence-free, respectively, although these differences were not statistically significant [54]. These results confirm the potential benefit of long-term pharmacotherapy for treating depressive illness.

However, one critical question emerges from these studies on the potential protectiveness of SSRIs: would these drugs confer similar prophylaxis if administered to healthy patients who are at-risk for developing mental illnesses such MDD or anxiety disorders? For example, fluoxetine (7.5 mg/kg/d, 21 days) pretreatment before SD seemed to prevent the defeat-induced anxiety-like behavior in Lewis rats, a highly stress-sensitive strain [55] (Table 1). However, these results must be interpreted with caution, since no comparison was made between defeated rats pretreated with vehicle and defeated rats pretreated with fluoxetine. Moreover, chronic fluoxetine (20 mg/kg/d, 14 days) treatment in C57BL/6J non-stressed mice has been shown to suppress depression-related behaviors in the TST, but impair anxiety and stress-coping ability in the EPM and in the OF [56]. Recently, it was demonstrated that administration of fluoxetine (18 mg/kg/d, 3 weeks) before the start of SD in 129S6/SvEvTac mice did not consistently protect against stress-induced depressive- and anxiety-like behavior, respectively, in the FST and in the EPM [57] (Table 1). However, fluoxetine pretreatment (18 mg/kg/d, 3 weeks) in C57BL/6NTac mice prevented the CORT-induced immobility in the FST, but did not protect against CORT-induced depressive- and anxiolytic-like behavior in the other paradigms [57] (Table 1). These latest results may be related to the antidepressant-like effects of fluoxetine more than a prophylactic effect, since the FST has originally been developed to screen drugs based on acute administration before the swim stress. Systemic administration of escitalopram (4 mg/kg/d, 14 days), concurrent with a CMS procedure in 129S1/SvImJ mice, prior to testing in the hole-board test (HBT) appeared to increase the time spent in the center of the hole-board, which is assumed to be aversive to rodents, but reduced the number of head dipping compared with saline-treated mice, which may reflect anxiogenic behavior [58] (Table 1). Moreover, escitalopram failed to reverse the depressive-like symptoms induced by CMS; rather, it increased immobility in the FST in treated stressed mice compared to non-treated stressed mice [58] (Table 1). In conclusion, prophylactic SSRI administration, at least in rodents as a protective strategy for stress-induced anxiety/depression-like behavior, is not fully efficient, but as we will describe later, other drugs may be more useful.

8. THE GLUTAMATERGIC SYSTEM

8.1. The N-methyl-D-aspartate (NMDA) Receptor

In the early 1990s, the glutamatergic pathway emerged as one of the most promising approaches for developing new antidepressant therapies, since an acute N-methyl-D-aspartate (NMDA) receptor blockade has been shown to improve behavioral deficits induced by inescapable stressors in NIH-Swiss and C57BL/6J mice [59]. Several years later, postmortem and magnetic resonance spectroscopy (MRS) studies reported region-dependent findings concerning glutamate or glutamine levels in MDD and BP patients, with higher levels in the frontal [60] and occipital [61] cortices and lower levels in the prefrontal [62, 63] and the anterior cingulate cortices [64, 65]. The literature so far provides contradictory results for glutamate levels in the hippocampus [66-68]. In rodents, exposure to stress produced an increase in glutamate release in regions mediating emotional behavior, as measured by intracerebral microdialysis [69]. In regards to specific glutamate receptors, some but not all studies have found a reduced NMDA binding affinity and expression in different brain regions of patients with MDD (for review, see [70]). Moreover, animal studies confirmed that dysfunctional subunits of the NMDA receptor may contribute to the pathophysiology of mood disorders, as mice lacking NR2A [71], NR2B in principal cortical neurons [72], or NR2D [73] NMDA receptor subunits exhibited strong anxiolytic- and antidepressant-like phenotypes relative to littermate control or heterozygous mice (Table 2). Nevertheless, a recent study invalidated these observations for NR2D knockout (KO) mice, which displayed anhedonic- and depressive-like behaviors in the sucrose preference test (SPT) and TST [74] (Table 2).

Overall, consistent with a promisingly significant role of NMDA receptors in mood disorders, a number of clinical trials demonstrated that targeting NMDA receptors with antagonists may be efficacious for treating mood disorders, as exemplified by the rapid and persistent antidepressant effect induced by ketamine, an NMDA receptor antagonist, following a single injection at sub-anesthetic doses in MDD or BP with or without treatment-resistance patients [75-79]. Clinical antidepressant efficacy of ketamine is clear, but studies in other psychiatric illnesses, like anxiety disorders, are more limited, although ketamine seems to persistently improve anxiety-like symptoms in anxious patients [80, 81], reduce PTSD symptom severity [82], or decrease OCD symptoms [83]. While ketamine given after a stressor did not protect against stress-induced depressive-like behavior for some preclinical studies [57, 84], it is an excellent candidate for plausible resilience-enhancing pharmaceuticals. Indeed, in mice, a single injection of ketamine (30 mg/kg) one week before an SD stress procedure protected mice against depressive-like behavior in the FST but did not prevent anxiety-like behavior in the EPM [57] (Table 2). Furthermore, a single injection of ketamine (30 mg/kg) in mice 1 week before LH training protected against LH-induced coping deficits to deal with inescapable shocks [57] (Table 2). In the CORT model, a single injection of ketamine (90 mg/kg) before a 3-week CORT treatment was protective against depressive-like behaviors in the splash test (ST) and the novelty suppressed feeding (NSF), but not against anxiety-like phenotype in the EPM [57] (Table 2). Finally, in male Sprague Dawley rats, administration of ketamine (10 mg/kg), 2 weeks, 1 week, or 2 hours before inescapable tail shocks (IS) blocked the IS-induced decrease in time spent in social juvenile exploration at any of time intervals between ketamine and testing [85] (Table 2). However, ketamine (30 mg/kg) buffered fear expression only when administered 1 week before contextual fear conditioning (CFC), but not 1 month, 24 hours, or 1 hour prior to CFC in 129S6/SvEvTac mice [86] (Table 2). Interestingly, a single injection of ketamine 1 hour following a 3-shock reinstatement decreased subsequent fear expression but did not prevent fear reinstatement when administered 1 week prior to 1- or 3-shock reinstatement [86]. Together, these results suggest a long-lasting protective effect of ketamine prior to stressful events that persist up to 3 weeks, but that the timing of administering ketamine relative to a stressor is critical for its effectiveness.

8.2. The α-Amino-3-Hydroxy-5-Methyl-4-Isoxa-Zolepro-pionic Acid (AMPA) Receptors

In addition to NMDA receptors, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors have also been proposed to be a major factor in the pathophysiology and treatment of mood disorders, but their role has been largely neglected. AMPA receptor activation is required for the antidepressant effects of ketamine, since blockage of AMPA receptor with NBQX antagonist pretreatment attenuated ketamine-induced antidepressant-like behavior in the FST [87]. Autoradiography in post-mortem brains of mood disorder-suffering subjects found increased AMPA binding density in the anterior cingulate cortex (ACC) of MDD patients [88] whereas GluR1 and GluR3 subunits of AMPA receptors were downregulated in the perihinal cortex of MDD and BP individuals [89]. In preclinical studies (summarized in [90]), acute or chronic stress exposure in animal models resulted in conflicting data concerning changes in AMPA receptor gene expression, depending on brain areas, especially for the GluR1 subunit. Numerous studies agree that the non-specific deletion of the GluR1-containing AMPA receptors in forebrain neurons reduced immobility in the FST and in the TST, increased time into the open arms in the EPM, and increased entries in the center in the OF as well as in the light compartment in the light dark test (LDT), suggesting that GluR1 deletion protects against acute stressors [91, 92] (Table 2). However, others studies reported depressive-related behavior in GluR1 KO mice with true coping deficits in the LH paradigm, not caused by an altered pain sensitivity [93] (Table 2). Thus, the protective effects of GluR1 deletion might only be due to exaggerated increases in locomotor activity during exposure to a novel environment

reported by all the studies. Interestingly, restricted ablation of GluR1 in glutamatergic neurons did not result in behavioral changes associated with mood disorders [94], whereas KO mice lacking GluR1 specifically in 5-HT neurons of the DRN, showed increased anxiety-like behavior [95] (Table 2). This discrepancy between global and 5-HT DRN neuron-specific GluR1 KO might be related to a reduction in THP2 expression and activity in the DRN as well as reduction in tissue levels of 5-HT in GluR1 KO mice [95].

Hence, data concerning the association between the GluR1 subunit and stress resilience or vulnerability are not clear for now, but other subunits of AMPA receptors might be involved in protective action against adverse experience. For instance, mice resilient to a 10-day SD showed increased GluR2 mRNA expression compared to control mice, while susceptible mice displayed a decrease in GluR2 levels in 
the nucleus accumbens (NAc) [96] (Table 2). Moreover, in adult male mice, overexpression of ΔFosB, a transcription factor targeting GluR2-containing AMPA receptors, specifically in the NAc reduced the propensity to develop social avoidance in mice subjected to chronic SD. Conversely, blockade of ΔFosB function promoted stress susceptibility [96]. Thus, induction of GluR2 in resilient mice appeared to reflect a direct effect of ΔFosB on the GluR2 gene, as ΔFosB binding to the GluR2 promoter is increased in these mice [96]. Yet, mice lacking the GluR2 subunit showed reduced anxiety in the EPM, but the markedly impaired motor competence of these mice precludes a definitive conclusion [97] (Table 2).

Despite contradictory outcomes from GluR1 and GluR2 studies, AMPA potentiating agents might be a promising new approach for increasing stress resilience and preventing stress-induced depressive-like states. For example, AMPA potentiator agents administered 1 hour before an acute stressor (FST and TST paradigms) induced antidepressant-like effects in both rats and mice [98] (Table 2). They also prevented against chronic stress-induced increases in corticosterone levels and stress-induced increases in latency to feed in NSF as compared with vehicle-treated mice [99]. Consequently, there is some evidence pointing to AMPA receptors as one potential molecular cause for individual stress vulnerability but also as a novel therapeutic targets for inducing resiliency for stress-related disorders.

8.3. The Kainate Receptors

Kainate receptors, a third glutamate ionotropic receptor, have also been implicated in anxiety and depression physiopathology, but still remain poorly explored. Among all the kainate receptor subunits (GluK1-5, previously called GluR5-7 and KA1-2), only the GluK5 subunit mRNA was decreased in the PFC of individuals with MDD or BP [100]. Also, decreased GluK2 mRNA expression in the entorhinal cortex has been found to correlate with MDD [89] as well as treatment-emergent suicidal ideation during treatment with escitalopram [101]. Moreover, genetic variations in the intronic region of the GluK2 gene suggested a potential association with somatic anxiety [102]. Further, a GluK3 gene polymorphism has been shown to be associated with recurrent MDD [103], while 2 single nucleotide polymorphisms (SNPs) in the GluK4 gene is associated with a protective effect against BP [104].

As previously mentioned, clinical evidences suggest that GluK2 may be a potential candidate gene in mood disorder susceptibility [89, 101]. Preclinical data are consistent with them because mice with genetically deleted GluK2 receptors exhibited less anxiety-like behavior in the OF and EPM and displayed an antidepressant-like phenotype in the FST, though these effects may be confounded by an increase of spontaneous locomotor activity [105] (Table 2). Finally, GluK4 KO mice showed a reduction in anxiety-like behavior in the EZM, the marble-burying test, and the NSF paradigm, as well as an antidepressant-like phenotype in the FST and the SPT, without effects on locomotor activity [106] (Table 2). In conclusion, progress remains to be made regarding involvement of kainate receptors in stress vulnerability or resiliency.

8.4. The Glutamatergic Metabotropic Receptors (mGluRs)

mGluRs are subclassified into three groups, based on sequence homology, G-protein coupling and ligand selectivity. In Group I, mGluR1 is associated with schizophrenia, while mGluR5 may play an important role in mood disorders [107]. In postmortem tissue, magnetic resonance spectroscopy (MRS) and positron emission tomography (PET) studies found reduced mGluR5 expression in MDD patients, whereas mGluR5 binding was increased in OCD patients. Moreover, in Flinders Sensitive Line (FSL) rats, proposed to be a model of retarded depression, lower densities of mGluR5 in the brain limbic structures were observed as compared to control Flinders Resistant Line (FRL) rats [108]. In two other models of anxiety and depression in male rats, elevated glucocorticoids and prenatal restraint stress, mGluR5 protein expression was reduced in the hippocampus [109, 110]. Finally, segregation of chronically stressed mice into susceptible and resilient subpopulations showed that susceptible mice exhibited less mGluR5 in the NAc than either resilient or control mice [111] (Table 2). Moreover, after a 3-day exposure to LH, SD, or restraint stress, mGluR5 KO mice exhibited enhanced susceptibility to stress-induced depression, social avoidance, and anhedonia [111] (Table 2). However, both social avoidance and anhedonia in mGluR5 KO was reversed by lentiviral rescue of mGluR5 in the NAc [111]. Hence, mGluR5-deficient mice failed to deploy effective defenses against chronic stress-induced depression, although Li and collaborators found a reduction of despair behavior in the FST [112] (Table 2). Due to the outcomes variation, the role of mGluR5 in stress resilience remains uncertain.

In Group II, clustering mGluR2 and mGluR3, the clinical data first suggested a strong increase in mGluR2/3 level in the postmortem PFC of MDD subjects [113]. However, these data were later refuted by Muguruza’s group which reported no changes in expression and density in the PFC of MDD subjects [114]. Based on the preclinical data, a decrease in mGluR2/3 gene expression in the hippocampus and an increase in the frontal cortex was a hallmark of rats exhibiting depressive-like behavior [115, 116]. Indeed, vulnerable and resilient mice to unpredictable chronic stress (UCMS) had a decreased hippocampal mGluR2 expression, but only resilient mice exhibited 82% lower mGluR2 expression within the PFC [117] (Table 2). No consistent difference in anxiety-like behavior was found between mice lacking mGluR2 and/or mGluR3 and their respective controls [118] (Table 2). Yet, mGluR2 is essential for promoting resilience to stress, since mGluR2 KO mice subjected to 28-day UCMS resulted in higher immobility in the FST, as well as a severe deterioration of the coat-state rating scale [117] (Table 2).

In Group III, mGluR4, mGluR6, mGluR7 and mGluR8 have been suggested to be involved in mood disorder physiopathology. Due to the lack of specific ligands for these receptor subtypes, various studies have been conducted in genetically modified mice. Thus, mGluR7 KO mice exhibited an antidepressant-like phenotype and anxiolytic-like behavior [119] (Table 2). In contrast, naïve 12-week-old mGluR8 KO mice displayed higher anxiety-related state in the EPM [120] (Table 2). Interestingly, when mGluR8 KO mice were tested in more stressful conditions or immediately after a 30-minute restraint stress in the EPM, no difference was observed between WT and KO mice, suggesting that the anxiety-like state of WT mice, but not KO mice, increases under more stressful conditions (Table 2). In conclusion, Group III receptors might play a role in acute stress resilience, but their protective potential against chronic stress still need further consideration.

With the exception of the clear long-lasting protective effect of ketamine, many studies have reported discrepant results depending on the target glutamatergic receptors. However, it is clear that the glutamatergic pathway is involved in stress resilience, though further research is needed for understanding how it can modulate stress vulnerability or resiliency.

9. THE ΓAMINOBUTYRIC ACID (GABA) SYSTEM

GABAergic neurotransmission is currently considered a key player in the control of fear and anxiety, but the role of the glutamate-GABA balance is becoming increasingly relevant in the development and treatment of stress-induced mood disorders. Abnormalities in the benzodiazepine (BZD) site, localized on the GABAA receptor, have been associated with the etiology and modulation of anxiety from imaging studies (see, e.g., the review of [121]). The GABAergic hypothesis in anxiety disorders has been well established, particularly in regards to successful efficacy of BZD in treating anxious patients. A recent meta-analysis of the existing MRS GABA studies across psychiatric disorders described lower GABA levels in MDD patients, but not in remitted MDD patients compared with healthy controls, and no significant difference in individuals with BP, panic disorder, or PTSD [122]. However, road traffic accident victims who developed PTSD had lower GABA-plasma levels 6 weeks after their accident. Interestingly, 75% of victims who had plasma GABA levels above 0.20 mmol/ml immediately after the accident and met criteria for PTSD 6 weeks after their 
accident were remitted at the 1-year follow-up, indicating that higher levels of GABA after a traumatic event may protect against chronic PTSD [123]. Seven-week CMS non-anhedonic rats developed a significant increase in cholecystokinin- and neuropeptide Y-positive GABAergic neurons density in the orbitofrontal cortex, as compared to anhedonic rats [124] (Table 3). Thus, alterations in GABAergic transmission may represent some aspects of the pathophysiology of depression (reviewed in [125]).

9.1. The GABAergic Receptors

The development of more selective agents has led to the identification of two distinct classes of GABA receptors: GABAA and GABAB. The pentameric GABAA receptor has been shown to have a critical role in the etiology of anxiety disorders, as patients with panic disorders or GAD show a reduction in BZD binding throughout the brain (for review, see [121]). A genetic association study revealed that common polymorphisms in the GABAA receptor α2, α3, α6, and γ2 genes do not play a major role in liability to anxiety disorders and MDD [126]. Feusner and collaborators reported an association between polymorphisms in the GABAA receptor ß3 subunit and higher symptom severity in anxiety and depression symptoms in a population of PTSD patients [127]. On the preclinical side, exposing juvenile rats to early postnatal stress resulted in an anxiety-like phenotype in adulthood associated with a decrease in the GABAA receptor α1/α2 ratio in both the amygdala and hippocampus, and α1/α3 ratio in the amygdala, suggesting that the α2 and α3 subunits might confer vulnerability to stress. As expected, specific deletion of the GABAA α1 subunit in corticotrophin-releasing hormone neuron enhance anxiety-like phenotype in the EPM and in the OF, impaired extinction of conditioned fear, but did not impact depressive-like behavior [128] (Table 3). It is noteworthy that genetic inactivation of α2-containing GABAA receptors results in a depressive-like phenotype, as indicated by increased immobility in the FST and the TST and a trend for increased latency to feed in the NSF [129] (Table 3). If GABAA α3 KO mice showed an enhance in swimming behavior, there is no evidence for an increase in the SPT [130] (Table 3). However, α2 or α3 deletion, in global brain or in intra-hippocampal areas of adult mice, did not affect baseline anxiety-like behavior in the EPM or in the OF [130-132] (Table 3). Finally, as homozygous GABAA γ2 KO is lethal in the perinatal period, heterozygous γ2 KO mice, with markedly attenuated expression of γ2 gene, was used to assess the protective role of this subunit in mood disorders. It appeared that GABAA γ2 plays a protective role, as γ2 KO mice had enhanced anxiety-related reactivity to natural aversive stimuli in the EPM and the LDT [133, 134] (Table 3). Collectively, the findings on various KO mice GABAA receptors identify α1- and γ2-containing GABAA receptors as key subtypes mediating resilience against acute stressors, but the role of all subunits of GABAA receptors should also be studied in chronic stress conditions, with applications to the stressful life events in humans leading to mental disorders.

GABAB receptors are emerging therapeutic targets for treating stress-related disorders since deficits in cortical GABAB receptors has been observed in MDD and treatment resistant MDD (TRD) patients [135]. A recent study suggested that the GABAB(1) receptor subunit isoforms differentially regulate resilience or vulnerability to either early life maternal separation or 10-day SD stress [136]. Indeed, the authors reported that in the absence of chronic stress, GABAB(1a) KO and GABAB(1b) KO mice displayed antidepressant-like behavior in the FST, but no difference in the SPT [136] (Table 3). Interestingly, following chronic SD, GABAB(1a) KO mice showed a decrease in social interaction and the SPT, while GABAB(1b) KO mice exhibited resilience as compared to WT stressed littermates (Table 3). Similarly, after maternal separation, GABAB(1a) KO mice exhibited increased anhedonia in the SPT, whereas GABAB(1b) KO mice maintained the reduction of immobility in the FST (Table 3). Anxiety levels in adulthood were unaffected either by genotype or chronic stress procedure in the EPM [136]. In conclusion, GABAB(1a) KO mice are more susceptible whereas GABAB(1b) KO mice are more resilient to both stress-induced anhedonia and psychosocial stress-induced social withdrawal.

9.2. The GABA Transporter (GAT)

The GABA transporter (GAT) also many play a major role in GABAergic transmission, as it participates in GABA homeostasis in the synaptic cleft. Because anxious and depressed individuals seem to have a deficit in inhibitory GABAergic activity, GAT may be involved in the pathogenesis of depression and anxiety. Nevertheless, there is little evidence concerning this hypothesis. A case-control association study found a link between two genetic variants located on the 5’ region of the GAT-1 gene and severity of panic attacks in anxiety disorder patients [137]. Furthermore, individuals with an anxiety disorder display a positive clinical response to tiagabine, a selective GAT-1 inhibitor [138]. Preclinical studies in GAT-1 deficient mice support the role of GAT-1 in mood disorders, as they demonstrate an increase in resilience to acute stress in several behavioral paradigms related to antidepressant- and anxiolytic-like activity [139, 140] (Table 3).

9.3. The Glutamic Acid Decarboxylase (GAD)

Another potential mechanism for the observed decrease in GABA levels in psychiatric disorders may involve abnormalities in the GABA synthesizing enzyme, glutamic acid decarboxylase (GAD) 65/67. Indeed, postmortem studies in patients with MDD revealed a significant reduction in GAD65 in the PFC [141] or in GAD65/67 density in the hypothalamic paraventricular nucleus (PVN) [142]. Similarly, in both mice and rat models of depression, GAD67 expression was downregulated in the PFC [143, 144]. Preclinical data from mice lacking GAD65 confirmed the necessity of functional GAD65 for preventing anxiety-like behavior and fear generalization to neutral stimuli and resistance to fear extinction in the cued fear conditioning test [145-147] (Table 3). But, surprisingly, deletion of the GAD65 gene elicited a reduction of stress-induced immobility associated with an enhancement of the swimming activity in the FST [145] (Table 3). Unlike homozygous GAD65 mutants, GAD65 haplodeficient (GAD65+/-) mice, which show a delayed postnatal increase in tissue GABA content in limbic and cortical brain areas, do not display an anxiety- or depressive-like phenotype, but rather exhibited similar emotionality to WT mice [148] (Table 3). In order to investigate the importance of the GAD65-mediated postnatal maturation of the GABA system in the development of PTSD, Muller and collaborators subjected GAD65+/- mice to either juvenile variable stress, isolation stress, or a combination of both stressors before testing their adulthood emotionality in a behavioral test battery [148]. In generalized contextual fear and anxiety, stressed WT mice showed an increased emotionality score following all stress procedures, whereas GAD65+/- mice were susceptible to isolation stress but resilient to variable stress [148] (Table 3). In contrast, in pre-conditioning anxiety in the OF and EPM, GAD65+/- mice were resilient to isolation stress but susceptible to variable stress [148] (Table 3). Altogether, these data suggest that GAD65 haplodeficiency confers resilience to ELS-induced development of contextual fear generalization, which is commonly observed in PTSD patients.

9.4. Benzodiazepines in Promoting Stress Resilience

The fast anxiolytic effects of BZDs are well documented, and their use for prophylactic purposes have also been studied. Indeed, male Sprague-Dawley rats treated for 6 days with diazepam (5 mg/kg) and tested 1 hour after the last injection, displayed an attenuation of FST-induced enhancement of plasma corticosterone levels [149]. Similarly, pretreatment with midazolam (1.5 mg/kg), administered 5 minutes prior to a restraint stress, in male Wistar rats prevented stress-induced enhanced conditioned freezing response through activation of ERK1/2 pathway into basolateral amygdala [150] (Table 3). Moreover, a single injection of midazolam (0.25 mg/kg) in low-anxiety but not in high-anxiety male rats 30 minutes prior to re-exposure to the CFC context reduced freezing levels and prevented fear reinstatement [151] (Table 3). Thus, stimulation of the GABAergic pathway prior to stress exposure or re-exposure may be relevant pharmacotherapy in regards to PTSD prevention.

9.5. THE HYPOTHALAMIC-PITUITARY-ADRENAL AXIS (HPA)

The HPA axis is a key controller of endocrine and behavioral adaptation to stress by facilitating responses to threat. Perception of physically or psychologically stressful situations is followed by activation of the HPA axis, leading to a secretion of the neuropeptide corticotrophin-releasing hormone (CRH) from the PVN, which in turn activates the secretion of adrenocorticotrophic hormone (ACTH) from the pituitary. Finally, ACTH stimulates the adrenal cortex, resulting in the secretion of glucocorticoids. Susceptible individuals exhibit persistently altered stress responses, acting principally upon a dysregulation of the negative feedback by endogenous glucocorticoids through the glucocorticoid receptors (GR), leading to chronic enhanced activity of the HPA axis and hypersecretion of glucocorticoids (for review, see [152, 153]). When glucocorticoids are hypersecreted over extended periods of time, the initially beneficial effects of HPA axis activation reverse into increased risk to develop a mood disorder. This may explain the increased pituitary and adrenal gland volume, as well as the elevated circulating levels of stress hormones, found in patients diagnosed with stress-related emotional disorders. These observations had even led to the development of the corticosterone (CORT) model in rodents, in which long-term exposure to exogenous glucocorticoids induced anxiety- and depressive-like phenotype, reversed by antidepressant administration [154]. Thus, effective therapeutics to improve stress resilience might be possible by targeting different levels of the HPA axis, such as CRH or GR.

9.6. The Corticotrophin-Releasing Hormone (CRH) and Its Receptors

Several studies have established that patients with mood disorders or PTSD present overactivity in CRH transmission. They contain higher CRH-positive neurons and CRH receptor density [152], though the contribution of CRH levels in the cerebrospinal fluid are still under investigation [155]. The preclinical data support this hypothesis, as adult mice susceptible to 10 consecutive days of SD showed increased CRH mRNA levels in the PVN, while no difference was reported in resilient mice [156] (Table 4). Therefore, it is reasonable to expect that reduced CRH transmission may confer resilience to stress and protect against mood disorder development.

CRH predominantly acts through CRH1 receptor to produce anxiety- and depressive-like symptoms; targeting the CRH1 receptor might be relevant for preventing stress-related disorders. One study found that CRH1-deficient mice spent a longer time in the light compartment in the LDT and in the open arms in the EPM as compared to WT mice [157] (Table 4). Interestingly, leaving basal and stress-induced activation of the HPA axis intact after postnatal inactivation of CRH1 receptor in mouse limbic structures, but not in the pituitary, induces an anxiolytic-like phenotype in the LDT and the EPM [158] (Table 4). These data are consistent with the downregulation of CRH1 receptors observed in the locus cœruleus (LC) and amygdala adult male Sprague-Dawley rats resilient to unpredictable and inescapable stress [159] (Table 4). Thus, it is not surprising to observe an anxiolytic/antidepressant-like effect of CRH1 receptor antagonism either in preclinical or in clinical studies (see, e.g., the review of [152]). In addition, a protective polymorphism formed by 3 SNPs located in intron 1 of the CRH1 receptor, which either decreased sensitivity of the CRH1 receptor or increased negative feed-back regulation of its functioning, was identified to lower effects of child abuse on adult depressive symptoms [160].

9.7. Cortisol and Its Receptors

A meta-analysis of 354 studies including 18,374 subjects across both depressed and control groups have reported greater cortisol levels in approximately two-thirds of depressed patients [155]. This observation was confirmed in 
an animal study in which adult male C57BL/6 mice were stratified into resilient or vulnerable subpopulations after a 10-day SD exposure [161]. Unlike control or resilient mice,

vulnerable mice were characterized by plasma CORT non-suppression following the dexamethasone suppression test, a synthetic glucocorticoid which can be used to evaluate the function of the negative feedback control of the HPA axis [161] (Table 4). Conversely, a meta-analysis of 17 studies reported lower cortisol levels in PTSD patients when compared to controls without previous exposure to trauma [162]. However, no difference was found between PTSD patients and resilient trauma-exposed controls [162]. The variable findings of glucocorticoid levels in MDD and PTSD subjects have posed challenges for understanding the role of the glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) in risk and resilience to the development of stress-related disorders.

GRs are expressed widely in the brain, principally in the PVN and hippocampus, and play a key role in stress-related homeostasis through an inhibitory feedback response. In absence of its ligand, GR is inactive and resides in the cytoplasm where it associates with the heat shock protein 90 (Hsp90) and its co-chaperone FK506-binding protein 51 (Fkbp5). This heterocomplex lowers the affinity for glucocorticoid affinity and reduces the efficiency of the nuclear translocation, but upon binding its ligand, the GR dissociates from its chaperone, becomes activated and translocates into the nucleus for specific transcription. It appeared that some polymorphisms in the GR (Nr3c1) and Fkbp5 genes in combination with ELS strongly increases the risk of suffering from mood disorders in adulthood [163].

Clinical evidence suggests a reduced GR sensitivity to glucocorticoids in patients with depression, suggesting the impairment of GR-mediated negative feedback on the HPA axis [164]. Unlike the nuclear expression of GR, an increase in cytosolic GR protein expression in the PFC and the ventral hippocampus was observed in anhedonic adult Sprague-Dawley rats, induced by CMS [165]. This may be explained by the enhanced Fkbp5 protein expression [165], resulting in reduced function and translocation of GR to the nuclear compartment. Similarly, in a single prolonged stress model of PTSD, male Sprague-Dawley rats had a greater PFC and dorsal hippocampal GR levels than non-stressed animals but a decreased sensitivity to glucocorticoids [166]. Thereby, mice that underexpress functional GR may mimic patients with affective disorders, while in contrast, mice overexpressing GR may demonstrate stress resilience. In fact, forebrain-specific GR KO (FBGRKO) mice exhibited increased despair in the FST and the TST and increased anhedonia in the SPT [167] (Table 4). The GR-heterozygous mice displayed normal behavior under basal conditions in a test battery for anxiety- and depression-like behavior, but exhibited coping deficits after a stressful challenge caused by the LH paradigm [168] (Table 4). Moreover, these mice are non-suppressors in the dexamethasone suppression test or after a 30-minute restraint stress, suggesting that mice underexpressing GR have a predisposition for depression [168] (Table 4). Conversely, mice carrying 2 additional copies of the GR gene are more resistant to developing helplessness [168] (Table 4).

Because genome-wide association studies (GWAS) correlate variants in Fkbp5 to susceptibility to mood disorders [163], the emotional behavior of mice lacking this co-chaperone was assessed. Under basal conditions, ablation of Fkbp5 in young adult mice have no effect on emotional behavior, but after a 21-day SD paradigm, mice spent more time swimming in the FST [169] (Table 4). Yet, aged Fkbp5-deficient mice (17-20 months old) exhibit an antidepressant-like phenotype [170] (Table 4). Taken together, these data confirm that Fkbp5 is involved in stress vulnerability, since individuals with Fkbp5 deletion are less responsive to the adverse effects of acute or chronic stress.

Besides Fkbp5, the GR heterocomplex also needs Hsp90 for maintaining competent conformation for ligand binding. In a recent study, GR binding to Hsp90 was significantly elevated in SD vulnerable mice, coinciding with reduced Hsp90 acetylation and increased nuclear GR, while resilient mice did not show any difference compared to the control group [161] (Table 4). Moreover, viral overexpression of a mutant protein mimicking the hyperacetylated state of Hsp90 was sufficient to promote resilient phenotype after SD [161] (Table 4). Specifically, Hsp90 is under the control of the cytoplasmic histone deacetylases (HDACs). It is known that inhibition or depletion of HDAC6 leads to hyperacetylation of Hsp90, resulting in GR assembly impairment [171]. Consequently, targeting HDAC6 may be considered a novel key regulator of stress resilience. Under acute inescapable stressors condition, deletion of HDAC in the global brain or specifically in 5-HT neurons in male mice promotes an anxiolytic- and antidepressant-like phenotype [172, 173] (Table 4). Interestingly, HDAC6Pet1Cre KO mice exposed to SD for 10 consecutive days failed to display the typical SD-induced increase in social avoidance [173] (Table 4). It should be noted that acetylation of Hsp90 through pharmacological inhibition of HDAC6 may offer a possible method to mediate stress resilience in an at-risk population. Indeed, treating non-stressed mice with systemic injection of the HDAC6-selective inhibitor NCT-14b caused an antidepressant-like effect in the TST [172] (Table 4). More interestingly, treating mice with a systemic injection of the selective HDAC6 inhibitor, ACY-738, 10 days prior to SD procedure, prevented the enhancement of SD-induced social avoidance [161] (Table 4). In conclusion, preventative strategies targeting the GR heterocomplex may be quite effective, but to our knowledge, none of these molecules were clinically tested for MDD or anxiety disorders.

GRs and MRs operate in balance to coordinate stress-induced HPA axis activity. Compared to the GRs, the MRs hold a 10-fold higher affinity for cortisol and are predominantly expressed in limbic areas. Interestingly, dexamethasone-resistant depressed patients showed normal cortisol suppression by prednisolone, a glucocorticoid that binds to both the GR and the MR, suggesting a clear dissociation between GR and MR function in patients with depression [164]. Like GRs, there are numerous studies linking MRs with susceptibility for psychiatric disorders. Generally, MDD patients showed a decrease in MR mRNA expression in several brain regions (as reviewed in [174]). This is consistent with the findings of 3 independent studies, including data from a GWAS that looked at reduction of MR activity and correlated this with susceptibility to MDD, as well as hopelessness and rumination [175]. Conversely, women, but not men, carrying the functional MR haplotype 2, which is associated with higher MR activity, exhibited enhanced resilience to depression and displayed higher dispositional optimism and fewer thoughts of hopelessness [175]. Hence, men’s susceptibility or resilience to depression does not seem to be modulated by the MR gene variability. This observation has led to the hypothesis that high MR levels confer resilience to stress-related psychopathologies. Indeed, genetic forebrain-specific MR-overexpression in mice present an anxiolytic-like phenotype [176] (Table 4). More specifically, overexpression of MRs in the basolateral amygdala in adult male Wistar rats reduced restraint stress-induced corticosterone secretion 48h post-virus injection and reduced anxiety in non-stressed and restraint stress-exposed rats [177] (Table 4). Such results have yet to be validated in the clinic, but recent data has shown that MR stimulation with fludrocortisone may improve cognitive deficits in depressed patients [178].

In conclusion, the data presented are strong evidence that the HPA axis through CRH, GRs, and MRs are important in mediating stress that may lead to mood disorders. Modulating these receptors using novel therapeutics may promote resilience to stressful events.

10. OTHER NEUROTRANSMITTER SYSTEMS

10.1. The Dopaminergic System

Insight into the biological variations in susceptibility and resilience can be gained by studying the mesolimbic dopaminergic system, particularly VTA dopamine (DA) neurons in the brain’s reward circuit, which play a crucial role in mediating stress responses. Dysregulation of DA release or alterations in DA receptor expression has also been associated with depression, though some postmortem and imaging studies have provided conflicting results [179]. The Nestler laboratory supports the idea that resilience to chronic stress is mediated by diminished dopaminergic neurotransmission, as they showed that the VTA DA neuron firing rate was upregulated in SD susceptible mice, while there was no effect in SD resilient mice [180, 181]. But, reducing the firing rate of VTA DA neurons by overexpressing voltage-gated potassium (K+) channels was sufficient to reverse susceptible mice into a resilient phenotype [180]. Likewise, phasic stimulation of the VTA DA neurons made SD-resilient mice susceptible [182]. Conversely, Tye and colleagues found that selective optogenetic inhibition of VTA DA neurons in non-stressed mice induced depressive-like behavior in the TST and the SPT [183]. However, phasic optogenetic stimulation of VTA DA neurons rescued unpredictable CMS-induced increase in immobility in the TST as well as the decrease in the SPT [183].

10.2. The Norepinephrine System

A large body of evidence points to deficiencies in brain norepinephrine (NE) neurotransmission as a key player in the pathogenesis of anxiety and depressive disorders. Specifically, an increase in NE neurotransmission through blockade of α2-adrenergic autoreceptor or NE transporter alleviates anxious and depression symptoms. Acute and chronic stress elicits dysregulation of the NE pathway, 
specifically through close interactions with the HPA axis 
and cortisol secretion. Thus, chronic traumatic stress leads 
to an hypoactivity of the NE system in brains of depressed subjects, resulting in decreased in NE turnover in limbic 
areas and increased α2-adrenergic binding sites in the LC [184].

NE neurotransmission also plays a key role in stress resilience. Ten days after SD, resilient mice exhibited increased NE levels in the VTA, but not in other limbic areas [185]. Interestingly, the susceptible phenotype of SD mice was reversed into a resilient phenotype after a 1-week daily treatment with a serotonin norepinephrine reuptake inhibitor (SNRI) or an α2-adrenergic receptor antagonist [185], suggesting that α2-adrenergic receptors might be a target of interest to increase stress resilience. Specifically, α2a-adrenergic receptors may play a potential stress-protective role whereas stress susceptibility might be mediated by α2c-adrenergic receptor. Mice lacking the α2a-adrenergic receptor exhibited more immobility in the FST and lower exploration of the light compartment in the LDT [186]. Target inactivation of the gene encoding the α2c-adrenergic receptor resulted in an antidepressant-like phenotype in the FST; conversely, overexpression of α2c-adrenergic receptor increased immobility in the FST [187]. Finally, the projection from the LC NE neurons to VTA DA neurons might be a critical factor in determining resilience to emotional stress. Chronic optogenetic stimulation of LC NE neurons projecting onto VTA stimulation reversed susceptible phenotype into a resilient one [185].

10.3. The Cholinergic System

The cholinergic system has also been implicated in mood disorder pathology. In medication-free subjects with recurrent MDD, a decrease in availability of the ß2 subunit-containing nicotinic acetylcholine (ACh) receptor (nAChR) throughout much of the brain structures has been observed, probably due to higher endogenous ACh levels rather than a decrease in the total number of receptors [188]. Secondly, blocking the muscarinic receptors with antagonists reduced depression and anxiety rating scale scores in patients suffering from recurrent MDD and BP [189]. Lastly, inhibiting acetylcholinesterase (AChE), an enzyme responsible for the breakdown of ACh, as well as hippocampal AChE knockdown (KD) in rodents had a depressive- and anxiety-like effect, which are reversed by administration of an AChR antagonist [190]. Taken together, these observations suggest that hyperactivity of the cholinergic pathway contributes to the pathophysiology of anxiety and depression. This hypothesis was supported in preclinical studies as KD or KO mice for the ß2 nAChR subunit exhibited anxiolytic- and antidepressant-like phenotypes [191, 192]. Similarly, mice lacking the ß4-containing nAChR displayed decreased anxiety-like behavior in the LDT and reduced fear memory retention in the cue-induced fear-conditioning task [193]. However, the results were variable when in depressive-like tests, as mice lacking the ß4 nAChR subunit showed increased immobility in the FST, but decreased immobility in the TST [193].

In addition to being involved in mood disorder states, the cholinergic system might also confer resilience to chronic stress. Unlike control animals, a 1-day SD stress induced an increase in social avoidance in hippocampal KD of AChE mice [190]. Moreover, downregulation of the ß2 subunit in the amygdala decreased social avoidance in mice subjected to a 3-day SD stress, suggesting that blockade of ß2 signaling in the amygdala increases resilience to social stress [192]. These data support the importance for maintaining ACh at homeostatic levels for promoting behavioral resilience to stress and preventing mood disorders.

As data on stress resiliency thus far demonstrate that there are a number of potential risk factors that increase susceptibility for developing mood disorders, particularly anxiety disorders, an emerging field of research is also focused on resilience in the context of aging.

11. RESILIENCE THROUGHOUT AGING

As an individual grows older, the risk for developing mood disorders, particularly anxiety disorders, increases with each passing year [5-7]. There is often high comorbidity of neuropsychiatric and neurodegenerative disorders, as well as of depression and anxiety disorders in elderly individuals [194-196]. Neuropsychiatric disorders in particular have been associated with accelerated cellular aging and neuroprogression, as well as secondary medical illnesses such as cardiovascular disease, stroke, osteoporosis, dementia, diabetes, and metabolic syndrome [197-203]. The effects of stress throughout aging may contribute to heightened incidence of disease onset [204, 205]. The aging process may change biological processes (i.e. HPA axis function) that are essential to maintain resilience [206-209]; the breakdown of these systems could make a once-resilient individual susceptible, though the causality between symptoms and biological breakdown is still under speculation. Additionally, there may be risk factors during old age that contribute to this increased susceptibility to stress, such as neurodegenerative disorders and genetic factors such as telomere shortening associated with stress-related oxidative damage [196, 202, 205, 210, 211].

In order to combat this susceptible phenotype, it has been shown that increasing resiliency throughout the lifespan can slow the aging process, improve overall health, and protect against stressful situations in multiple species [212-214]. Importantly, several studies have discovered that adults aged 85 or older have a similar capacity for resilience as younger individuals [215-218]. However, it is equally likely that resilience is maintained in individuals throughout the lifespan, and that those with more susceptibility to disease are not included in this study due to earlier mortality. Despite this caveat, these studies have spurred an increased focus on developing behavioral interventions to attempt to improve resilience in older adults. Most research, however, is centered on developing measurement scales and identifying characteristics of resilience, and not many resilience interventions have been developed or thoroughly studied, especially for older adults [215]. Historically, resilience research has been largely centered on early childhood and adolescence, and has not been as widely studied in the adult or elderly population [219]. Only recently have researchers begun to study resilience in aging, and how the changes that occur in the body with age relate to the potential neurobiological underpinnings of stress resilience (reviewed in [220]). For instance, several groups have attempted pharmacologic approaches to increase the lifespan and improve resilience in pre-clinical models [211].

12. EFFECTS OF STRESS THROUGHOUT AGING

Previous work has shown that in a large sample of Canadians aged 18 and older, the perceived impact of stress exposure is strongest in older adults [221], suggesting that the level of observed stress correlates with the onset of stress-induced disorders [222]. It is generally accepted that the way that an individual ages is highly influenced by major life events and stressors (reviewed in [223]). Here, we will discuss the increased incidence of psychiatric disorders in older adults, and how mood disorders subsequently have a negative cyclical effect by accelerating aging.

12.1. Increased Incidence of Psychiatric Disorders in Older Adults

A literature review focused on World Health Organization (WHO) World Mental Health (WMH) surveys found that there was a proportional increase in projected lifetime risk versus prevalence with mood disorders. Additionally, estimates of projected lifetime risk of any DSM-IV mental disorder is increased throughout the lifespan [6]. It has been estimated that 25% of the population over 65, or 8.6 million people, contracts a mental disorder [224]. In particular, the overall rates of GAD are high (5-10% lifetime prevalence), but due to increased vulnerability in elderly people, late-life onset of GAD is as high as 25% [204, 225, 226]. Anxiety symptoms are common in older subjects, especially when they are medically ill, and depression and alcoholism are also often comorbid diagnoses [195]. Specifically, the prevalence of anxiety in community samples of older adults ranges from 15-52.3%, if sub-threshold anxiety symptoms are included as well [227]. Depressive symptoms are also prevalent in older adults, though some of the earlier literature reported values that varied from 1-23% depending on the population sampled [228]; however, a more recent review concluded that the prevalence of major depression is up to 16-42%, depending on whether the elderly were living in private households or in institutions [229]. Comorbid anxiety disorders and depression are common; one group found that as many as 35% of depressed patients aged 60 and older had at least one lifetime anxiety disorder diagnosis, and 23% had current diagnosis at the time of the study [230]. In younger individuals, the estimates range from 10 to 15% [231]. These studies, though varied, indicate that there is a general increased prevalence of psychiatric disorders with old age, and with it, an increased incidence of other mental disorders as well as secondary illnesses.

One specific group that experiences increased risk for a psychiatric disease throughout life is the veteran population. Several studies have recently shown that elderly individuals experience greater levels of stress after war, and respond differently to social support aimed to improve mental health [232, 233]. For instance, one study, which assessed individuals along the Lebanese border after the Second Lebanon War, found that the elderly reported significantly higher levels of stress symptoms, and lower levels of posttraumatic recovery as compared with younger age groups [232]. Additionally, older veterans experience PTSD at higher rates than younger veterans; in other words, the longer the time from the experience of war, the more likely an individual is to begin experiencing symptoms of PTSD, possibly due to the phenomenon of delayed-onset PTSD. One study found that approximately 1 in 10 US veterans experiences a clinically significant exacerbation of PTSD symptoms in late life, at an average of nearly 3 decades after their worst trauma [234]. Though the mechanisms of this late-life exacerbation were not discussed, it is possible that the aging process contributed to this intensification of trauma-related stress reactivity leading to PTSD.

In terms of factors that may contribute to the exacerbation of PTSD in older US veterans, a recent study was published that assessed loneliness [235]. The authors found that loneliness is prevalent among older veterans in the US, as 44% reported feeling lonely at least some of the time, with 10.4% reporting that they often felt lonely. The authors correlated increased age, trauma, perceived stress, and several other factors with the severity of loneliness and concluded that interventions might aid in mitigating loneliness, through social support and the eventual reduction of depressive symptoms. However, no specific interventions were suggested or outlined in the study. Another study echoed these effects and found that a significant portion of older male veterans in the US contemplates suicide, with a higher rate of suicidal ideation among combat veterans as compared with non-combat veterans [236]. Both studies conclude by suggesting interventions that improve social connectedness to decrease loneliness, though no specific interventions were proposed.

These data are in stark contrast to a study that assessed older veterans and found that 69.5% of US veterans aged 60 and older were considered resilient, which they characterized as a high number of lifetime traumas and low current psychological distress [237]. These individuals were highly likely to have been college or higher level educated, and had high levels of emotional stability, social connectedness, and positive perceptions of the military’s effect on one’s life. Whether these characteristics existed before the experience of these traumas, or were built as a result of them, is unknown, but it is interesting to consider the possibility that these veterans had with high levels of resilience to begin with, and despite their experiences with multiple lifetime traumas, came away without disease. It is also possible that the generation of the cohort may have an impact on the outcome; specifically, the older generation in this cohort may be more resilient in general than the younger generation, though to our knowledge, no studies have informed on generational differences in regards to stress resilience. According to the APA’s Stress in America survey from 2011, 23% of Baby Boomers, or those born between 1946 and 1964, reported decreased stress levels, and this may be because they were more likely to have grown up in two-parent households, with safe schools, job-security, and post-war prosperity; they also report decreased stress levels [238]. Thus, it is possible that there may exist a generational difference for stress resilience, and that this is manifested in the US veteran study, though no studies have directly tested this hypothesis.

12.2. The Role of Mood Disorders and Stress on Accelerated Aging

Experiencing chronic stress is a large risk factor for developing mood disorders, and both stress and mental illness may contribute to accelerated aging. It was shown that chronic psychological stress in rats affect genes that are critical for longevity [239]. Stress has been found to impact the developmental process, especially during critical developmental stages [240]. In general, however, there are complex interactions between stress and aging, as one can promote the other [241]. It has been shown that the peroxisome proliferator-activated receptor γ (PPARγ) may be a critical mechanistic link between aging and stress [241]. For example, PTSD has been associated with elevated risk for age-related diseases; it has been hypothesized that chronic PTSD is a form of persistent life stress that elevates oxidative stress and leads to accelerated cellular aging [202]. Elevated rates of aging-related biological and functional decline, specifically through accelerated cerebral white matter aging, was also found in patients with schizophrenia and MDD [203]. Stress experiences, therefore, appear to be interlinked with aging processes and psychiatric disorders, and the combination of the two may have large consequences on the rate of aging.

13. BENEFITS OF STRESS RESILIENCE IN AGED ADULTS

Though the risk of contracting mood disorders and other diseases increases with age, there are individuals who are able to avoid these risks, and succeed in living healthily throughout the lifespan. Thus, strategies that enhance resilience may slow aging [242]. Here, we will discuss how resiliency not only improves general well-being, but also how it may function to slow the process of aging.

13.1. Stress Resilience and Improved Health in Aged Adults

Having components of stress resiliency has been correlated with high functioning and successful aging in older adults. One group demonstrated that several markers of resiliency in persons older than 85 years include intact cognitive function, high mobility, and good nutritional status, and that these traits improved the likelihood of recovery from disease, and in performing activities of daily living (ADLs) [243]. Another group attempted to estimate the likelihood of and the factors associated with recovery from exhaustion in older adults, which is a common ailment for this population. They found that among individuals aged 69 or older, resiliency was associated with physical and psychological well-being in the face of persistent exhaustion [244].

13.2. Stress Resilience and Slowed Aging

In fact, as an individual grows older, stress resilience becomes increasingly important to continue to age successfully. Several studies have demonstrated that the older individuals are, the more resilience becomes a critical factor in maintaining health and longevity, and the more that adults self-rate as successfully aging [214]. For instance, one group studied centenarians, nonagenarians, and octogenarians, and found that nonagenarians aged 94-98 with increased resilience had a 43.1% higher likelihood of living to be 100, or becoming a centenarian, than those with lower resilience [245]. This suggests that the impact resilience becomes more pronounced at older ages, and greatly contributes to healthy lifespan.

In one review, the resilience framework was applied in two case studies of resilience among patients with Alzheimer disease (AD), and found that individuals with dementia may still lead meaningful lives, and that the resilience process may contribute to a more positive dementia experience [246]. The key point of this review is that the field of gerontology should be striving not for successful aging, but for resilience, which has a fundamental role in aging. Importantly, improving resilience and improving longevity is possible for all adults regardless of social and cultural background, or physical and cognitive impairments [246].

14. CHANGES THROUGHOUT AGING THAT SHIFT A RESILIENT PHENOTYPE INTO A SUSCEPTIBLE ONE

Though it is evident that increased resilience is beneficial throughout life, it is possible for an individual to gradually lose a resilient phenotype as a result of the aging process. There are several mechanisms that are critical for maintaining resilience that are vulnerable to wearing down and making a once-resilient individual susceptible to stressors and the development of mood disorders. These include HPA axis dysfunction, brain atrophy in regions that are most actively protective against stress, genetic causes, loss of coping abilities, and the onset of a neurodegenerative disorders. Additionally, early-life or critical-period stressors are factors that may accelerate the resilient mechanism breakdown.

14.1. HPA AXIS Dysfunction in Aging

As discussed previously in this review, the HPA axis modulates stress responses by facilitating responses to threat, and susceptible individuals exhibit persistently altered stress responses characterized by enhanced activity of the HPA axis and secretion of glucocorticoids [152, 153]. Importantly, the HPA axis may be perturbed by exposure to prolonged stress that leads to deleterious effects, and HPA axis dysfunction has been correlated with age-related diseases such as depression and cognitive disorders [208] and this predisposes individuals to deleterious effects such as unstable emotional regulation [209]. For example, there could be either decreased aldosterone, or hyperaldosteronism resulting from increased ACTH stimulation that occurs with age [206, 208]. Cortisol may also be increased with aging, though the evidence supporting this claim is not conclusive [209]. To date, there are few studies that assess specific changes in the HPA axis that occur throughout aging that may contribute to mood disorder onset, and much work needs to be done to elucidate the what contributes to HPA axis breakdown, and how this may be prevented.

14.2. Telomere Shortening and Genetic Factors Associated with Mood Disorder Onset

In addition to changes brain morphology and stress axis functioning, there are also genetic markers that may either predispose individuals to developing mood disorders, or epigenetic changes that induce a susceptible phenotype. A large body of research has been dedicated to investigating the relationship between telomere length and health [247, 248]. Reduced telomere length has been associated with a number of chronic somatic diseases normally associated with aging but also implicated in psychiatric diseases such as depression [249-251]. Despite the established role of telomere length in health and aging, there are only a few therapies have been developed to combat the telomere shortening process. For instance, there have been several small stress reduction and wellness studies that have examined the potential to elongate telomeres in vivo over short periods of time [252]. The recent interest in gene therapy has led to the development of telomere-enhancing drugs not only as a cancer therapy [253], but also as a method to potentially slow the aging process, as a recent biotechnical company BioViva, based in Texas, has proposed.

In regards to genetic markers and polymorphisms that may indicate resilience, if 5-HTTLPR short (s)-allele carriers have reduced resilience to stress in young adults, in older adults, it appeared that though the s allele had less of a direct effect on resilience, it did correlate with poorer self-rated successful aging, as well as cognitive performance, suggesting that carrying this gene throughout life has an impact on quality of life in old age [254]. A separate study found that SNPs in the FK506 binding protein 5 (FKBP51) gene is an indicator of increased risk to develop PTSD and MDD when exposed to traumatic events [255]. The trauma can induce changes in FKBP51 protein expression and impair glucocorticoid signaling [255].

Epigenetic changes throughout the lifespan may also greatly contribute to disease susceptibility, and are more commonly studied than single genes, as both aging and stress are associated with changes to the epigenome [256-258]. Indeed, DNA methylation changes are associated with an aging clock, which has been implicated as a biomarker of aging [259]. In general, aging has been known to affect specific regulatory gene elements such as enhancers, promoters, and CpG islands [260]. It was found that in mice, age-associated hypomethylation was enriched at super-enhancers at highly expressed genes that are critical for liver function, and hypermethylation was enriched at CpG islands. These changes were suppressed in calorie-restricted mice and in mice treated with rapamycin (two possible therapies discussed later in this review) [260]. Epigenetic changes as a result of stress may also occur at the level of microRNAs (miRNAs). For instance, the Nestler laboratory found that β-catenin mediates pro-resilient and anxiolytic effects in the NAc of mice, which affects miRNA regulation through Dicer1, which is important in maintaining stress resilience [261]. These studies suggest that lifestyle changes may confer epigenetic modifications that contribute to longevity. Overall, it seems that several genetic changes, either inherited or changed over time by telomere shortening, genetic polymorphisms, or epigenetic modifications, may contribute to age-related disease onset.

14.3. Correlation Between Age-Related Cognitive Decline and Mood Disorders

With the onset of a neurodegenerative disorders or age-related cognitive decline, the risk for developing a mood disorder increases, and vice versa [262-265]. Shimada and colleagues found that older adults with depressive symptoms have increased risk of cognitive impairment and dementia, particularly AD and vascular dementia [263, 266]. Additionally, anxiety is a common symptom among patients with a cognitive impairment [5]. In cases such as these, the mood disorders correlate with poorer outcomes in patients, and the two disorders often worsen in tandem [5]. It is possible that these cognitive impairments linked to these mood disorders are induced by cellular loss that occurs with age. In the aging brain, it is common to observe atrophy of various regions due to cell loss and tract-specific white matter degeneration [267, 268]. Although brain atrophy in aging has mostly been studied in AD and other cognitive disorders, there are several studies that indicate that depressive symptoms and other mood disorders are associated with changes in both gray and white matter, including atrophy [269]. White matter lesions are frequently found in patients with late-life depression, which is a characteristic that overlaps with dementia [269]. The hippocampus undergoes several structural changes both grossly and at the cellular level with aging, and these have been correlated with cognitive decline in both animals and humans [270]. Although similar hippocampal alterations occur both with normal and pathological aging, in the case of neuropathologies these changes are exacerbated (increase in inflammation and decrease in trophic factors, decreased histone acetylation) [270]. Smaller hippocampal volume has been also associated with depressive symptoms, as well as stress and anxiety [271-273]. The PFC and the amygdala also undergo a plethora of changes during aging [274]. The modulation of stress responses during aging could be affected by changes in PFC-basolateral nucleus of the amygdala. Thus, insensitivity of the PFC-basolateral amygdala pathway to stress could constitute a mark of health, while the alteration of this specific pathway could be involved in mood disorders [274].

Overall, changes in brain region integrity may have 
effects on brain network function, and specifically the default mood network, cognitive control network, affective/
frontolimbic network, and corticostriatal circuits, which may be directly relevant for mood disorder onset (reviewed in [275]).

14.4. Critical Period Stressors Contribute to Resilience Mechanism Breakdown

The breakdown of brain functions necessary to maintain resilience could likely be induced not just by age, but by an ELS that has long-term deleterious effects. Exposure to stress during critical periods, such as during childhood, adolescence, and early adulthood, may cause an individual to become even more susceptible to stress in the future, and can lead to disease and a number of neuropsychiatric diseases [242, 276, 277]. Specifically, adverse childhood experiences (ACEs), including physical, sexual, and emotional abuse, have been correlated with depression in older adults [277]. The number and types of childhood maltreatment also affect the risk of depression, as cumulative stress has a negative effect on likelihood for mood disorders [242]. It has been shown that several significant important predictors of lifetime history of MDD included non-verbal emotional abuse in males, and peer emotional abuse in females at 14 years of age [242]. For PTSD, the critical periods of vulnerability to ACEs have been found to be the pre-school (age 4-5) and pre-adolescent (age 8-9) periods, and that the type and timing of ACEs affect severity of symptoms [278].

Though little is known about the underlying biology of this effect of early childhood trauma on late-life disorders, it has been noted that early childhood adversity such as abuse, neglect, or exposure to violence increases pro-inflammatory responses [279], affects patterns of gene expression regulated by inflammatory signaling [280], and quickens the process of telomere shortening [249] (as described earlier in regards to stress vulnerability and accelerated aging). One study found that children exposed to maltreatment had smaller volumes of the PFC and the hippocampus, greater activation of the HPA axis, and changes in inflammation levels as adults [279]. Another study found that stressors induce persistent changes in the HPA axis and affect its ability to respond effectively to stress in adulthood [281]. It has also been shown that telomere length may be associated with severity of ELS [249, 282]. 5-HT has been suggested to play a critical role in the maturation of brain systems that modulate emotional functioning in adults [283]. Specifically, inactivation of 5-HT receptors during development produce behavioral abnormalities related to neuropsychiatric diseases such as anxiety and depression [283, 284]. 5-HT has a suggested role in resilience, as previously discussed in this review, and may represent an essential target system that is most sensitive during developmental periods and may induce later-life psychopathology. Overall, the effects of stress during early, vulnerable periods in life may confer long-lasting biological changes to processes necessary to maintain resilience such as nervous, endocrine, immune, and genetic systems; these changes affect an individual’s response to future stressors, and lead to faster aging and disease.

15. SEX DIFFERENCES IN AGING AND STRESS RESILIENCE

When considering factors that may contribute to stress susceptibility in old age, it is important to note the role that gender plays in regards to aging and in responding to stressful experiences.

15.1. Sex Differences in Aging

Several studies have attempted to assess sex differences in the aging process, and have discovered a number of characteristics that distinguish between male and female longevity. Women appear to live longer than men, as the average life expectancy for women is 83.5 years, and for men, 79.5 years [285]. This increased lifespan of women may be due to the rate at which telomeres shorten in males and females, though there has yet to be a study that directly links sex, telomeres, and lifespan observed across multiple species [286]. Another hypothesis links responsivity of the immune system to longevity and susceptibility, though the studies assessing this hypothesis are small and not conclusive [285]. A recent study used resting-state networks to study sex differences in normal age trajectory, and found that males and females both showed decreases in connectivity with age, but at different rates, which may explain the rates of aging [287]. Yet, paradoxically, despite living longer lives, women generally experience worse health throughout life than men [285, 288]. This decreased quality of health throughout life is likely due to an increased vulnerability to stress in women, as women seem to be more susceptible to stress than men throughout the lifespan [289].

15.2. Differential Sex Effects of Stress Throughout the Lifespan

Sex differences in stress responses are found at all ages, and are related to gonadal hormone changes that occur throughout development and maturation, and sex chromosome genes (reviewed in [290]). In general, females are twice as likely to develop anxiety disorders such as PTSD than males [233], even though some studies suggest that the probability of trauma exposure in females is lower [291]. Older depressed females were observed to have higher GR immunoreactivity in postmortem hippocampal tissue than elderly depressed males [292]. Women also seem to lose hippocampal volume more rapidly than men in older age, according to one study, which used cerebral magnetic resonance imaging (MRI) [271]. There also seems to be indication that epigenetic mechanisms may promote sexual dimorphism in the brain in regions such as the hypothalamus due to sex hormone exposure during development [293]. This differentiation may create susceptibilities between males and females for psychiatric disorders. Yet, despite some hints about how females seem more likely to develop mood disorders, the exact neurobiological underpinnings of this increased susceptibility are not well known, though several studies have provided insight into potential mechanisms. It has been hypothesized that prepubertal stress induces long-lasting changes in brain regions such as the hippocampus and that these effects are sex-specific [276]. Specifically, one study found there prepubertal stress impaired hippocampus-dependent behavior in males (CFC), yet enhanced hippocampus-dependent behavior in females (spatial navigation and memory retention), suggesting differential sex effects of brief, variable stress during the prepubertal phase.

15.3. Peri- and Post-menopausal Periods as Windows of Vulnerability for Women

Recent research has attempted to describe female’s heightened response to stressors by exploring the effects of stress during peri- and post-menopausal periods, as these represent windows of vulnerability for women to develop mood disorders [294-298]. Specifically, women are 2 to 3 times more likely to experience a first episode of depression during the peri-menopausal period [299, 300].

A reason for this increased susceptibility to stress could be due to the loss of gonadal hormones in females and their metabolites that are critical for stress regulation [290]. Ovarian estradiol secretion ceases at menopause and is followed by low levels of circulating estradiol [301]. Interestingly, one study found that age after menopause impacted the effect of estradiol hormone replacement therapy on mood after a psychosocial stress situation. This is important in light of previous studies, which demonstrated that estradiol improves mood in women in the menopause transition, but not older post-menopausal women. Thus, estradiol administration may be modulating emotional reactivity to stress by sensitizing older women to its adverse effects [302]. Though a specific mechanism is not addressed in this study, it suggests that estrogen receptor function shifts during the post-menopausal phase in women, and that this shift may be critical in mediating women’s responses to stress. Several other groups have demonstrated the effects of estrogen receptor function shifts by examining mood in pre- and post-menopausal women, in which estradiol treatment has differential effects in both populations [302-304]. Indeed, in line with these studies, it has been suggested that the rapid decline of gonadal hormones, as well as ovarian senescence, in women and the cellular aging process promotes stress dysregulation in women [290, 305]. Thus, females are at greater risk for mood disorders induced by stress throughout life, and this should be considered when developing preclinical models or stress resilience enhancers that would benefit both males and females. It is possible that the male and female populations may require separate therapeutic strategies based on the differences in their stress reactivity, as well as the significant influence of hormonal states.

16. BEHAVIORAL STRATEGIES OF PREVENTING MOOD DISORDERS EXACERBATED WITH AGE

As there currently exists no pharmaceutical therapies for prevention of mood disorders in old age, one option is to implement behavioral strategies to prevent mood disorders in aged adults. Though very few intervention programs exist for elderly adults, there are several intervention programs that have been developed to increase resilience for the general population.

16.1. Intervention Programs

One example of an intervention program aimed to increase resilience is the American Psychological Association (APA)’s Road to Resilience program, which sought to promote resilience in in middle and high school students affected by the 9/11 terrorist attacks through focus groups in several major cities [306]. There are plans to provide psychologist-led workshops, forums, and lectures, but this program is not validated in older adults [306]. Meditation and Physical (MAP) training was also implemented in a small cohort of young mothers who were recently homeless and suffered from physical and sexual abuse, addiction, and depression. After 8 weeks, measures of aerobic fitness increased, and symptoms of depression and anxiety decreased [307]. However, though these positive preliminary results are promising for developing more refined and effective interventions, it is important to note that these examples have been published as pilot studies, and that none of them have extensive or long-term evaluation of their impact on resilience, or very large sample sizes in their original studies (for review, see [215]).

For current active duty military members, there exists the US Army Ready and Resilient Campaign (R2C) is one that attempts to build resilience in soldiers, army civilians, and their families, and the Army spends about $50 million a year on this program alone. In 2013, there existed over 94 different prevention or resilience programs within the Department of Defense (DoD). As opposed to the aforementioned studies, the impact on resilience from these programs has been evaluated, but no consistent, significant impact has been uncovered yet [308]. Thus, overall, resilience behavioral training is in its infancy and there exists a dearth of evidence validating that these behavioral programs are actually effective [309].

The only intervention programs in existence for older adults are implemented primarily after the onset of symptoms. For instance, though the exact causes of increased depression in aging is unknown, some research suggests that deficits in biological rhythmicity can contribute to onset of depressive symptoms. One option for these patients is using bright light therapy (BLT), in which bright blue light is shone daily in elderly patients’ homes. In essence, the patients are to sit in front of a light box for 60 minutes each day for the duration of the study, during the middle of the day [310]. One group found that 3 weeks of BLT treatment caused a reduction in MDD symptoms, including mood, sleep and cortisol hyperexcretion [311]. There also exist elderly suicide prevention programs, which consist of community workshops, interpersonal psychotherapy, and individual counseling. One review summarized 19 studies that assessed suicide prevention programs for adults aged 60 or older, and found that most studies observed a reduction in the level of suicide ideation or suicide rate of their respective communities [312]. The authors point out, however, that none of the studies aimed to improve protective factors.

Despite these small studies, building resilience through behavioral interventions in older adults is a field that requires much more investigation. Most research concludes with suggestions rather than definitive evaluations; for instance, savoring positive experiences was examined in older adults, and the authors reached the conclusion that positive psychological interventions to enhance resilience would be beneficial [313]. In general, positive emotions correlates strongly with resilience and well-being, and these could be the foundation for building effective resilience programs, or happiness interventions, for both the general population and the elderly [222, 313-315]. However, to our knowledge, no specific interventions have been established. Thus, the field of resilience enhancement in the elderly can potentially gain insight from resilience studies on other populations such as children and adolescents, but would likely find more value in studies that have examined resilience among specific groups of older adults, such as veterans or groups that come from specific backgrounds or experienced similar adversities. Because resilience is such a dynamic process, one suggestion has been to develop individualized resilience enhancement programs for older adults [215, 316, 317].

16.2. Exercise Therapies and Caloric Restriction (CR)

A popular intervention is regular exercise or high mobility, as a number of studies have correlated regular exercise and physical strength with higher emotional resilience in adults [18, 318, 319], and contributes to successful aging [320-323]. In addition to its effectiveness in preventing or alleviating heart disease [324], exercise has also been shown to improve mental health in older populations [325]. Increasing interest has been focused on using exercise to promote healthy aging and as a treatment for older people with and without psychiatric illnesses [326]. In regards to resilience, in a study of participants 72 years or older, it was demonstrated that high mobility was a positive predictor and a marker of resiliency in this population [243]. In general, the molecular processes of exercise are ones that attenuate typical signs of aging, such as inflammatory processes and telomere length [327, 328]. Additionally, since maximal oxygen capacity decreases with age, due to decreases in aging skeletal muscles, exercise can have a restoring effect on health; it also has effects in neurodegenerative diseases and oxidative stress regulation, even in frail elderly [327]. These studies collectively suggest that exercise has a multitude of benefits, but importantly, they may increase health and resilience in elderly populations.

In line with exercise therapies, weight management over time has also been correlated with resilience, particularly in the older population [329]. Thus, caloric restriction (CR) has been used as a therapy to postpone the detrimental aspects of aging, and is defined as a decrease of 30% to 60% ad libitum feeding without malnutrition (reviewed in [211, 329]). It has been shown to be effective in improving resilience, but that the effects of exercise have proven more consistently beneficial [248]. Numerous studies since 1935 have shown that lifelong CR increases mean and maximum lifespan and delays age-associated disease in several species, including mice, rats, and rhesus monkeys [330-332]. For instance, some studies have found that some dietary regimes and over a dozen single-gene mutations can extend the mean and maximal lifespan in mice [333, 334]. It has been suggested that CR may suppress inflammatory cytokines, a common risk factor for a number of chronic diseases [335]. Though CR is not used primarily as a method to alleviate mood disorders, it has been shown to improve mood states such as depression, but to our knowledge, the relationship between CR and mood has only been examined in aged men [336, 337]. Altogether, these studies provide evidence that therapeutic interventions in older adults would benefit from incorporating exercise and CR in their programs, though some groups are working to achieve the effects of these therapies, as not all individuals can withstand the rigor of these approaches.

17. POTENTIAL METHODS TO TARGET NEUROBIOLOGICAL SYSTEMS OF RESILIENCE IN AGING AND ELDERLY ADULT POPULATIONS

Although the aforementioned behavioral interventions are currently the standard of care for elderly patients susceptible to mood disorders, or who already suffer from one or more, it may be possible in the near future to target biological mechanisms of resilience to protect against stress.

17.1. Stress and Aging in Invertebrates and Mammals

As early as the 1990s, it was known that mutations in the genes of the nematode Caenorhabditis elegans (C. elegans) led to extensions in their lifespan; it was discovered that these nematodes were resistant to oxidative and thermal stress, as well as other environmental stressors [220]. This ability to face multiple stressors and protect against multiple forms of injury was deemed “multiplex stress resistance” [213]. More thorough molecular observations revealed that mutations that lead to stress resistance involved the slowing of molecular chaperones and cellular protein accumulation, which represents failed homeostasis, and that enhancing chaperone activity via enhancement of the heat shock factor 1 (HSF-1) increased stress resilience and life span in C. elegans [338]. Though studies in mammals are less numerous, some cell culture work has revealed that fibroblasts from Snell dwarf mice and cells from long-lived species, are resistant to multiple forms of stress in culture [339, 340].

Pharmacologic strategies have also been proposed to improve indices of health and aging, though the effects on resilience have not been consistent [211]. As discussed, the benefits of CR and exercise are large, but committing to a rigorous dietary program is unfeasible for some individuals. Thus, novel research has focused on developing CR mimetics such as pharmacological approaches in order to confer the benefits of CR and exercise without the rigorous requirements. For instance, the mTOR inhibitor rapamycin has been shown to extend lifespan and have effects on immune processes when administered to mice [341-344], as well as other compounds such as resveratrol and metformin [329]. However, a recent study demonstrated that neither rapamycin nor CR has effects in older mice, and that there may even be deleterious effects to the aging immune system [250]. Resveratrol, a polyphenolic flavonoid found in grapes and red wine, and has been shown to enhance health and increase lifespan, and it may currently be taken as a therapeutic supplement in humans (reviewed in [345]). Other options such as drugs that inhibit the growth hormone/IGF-I axis, or ones that activate specific sirtuins are being discussed as potentially promising candidates to increase lifespan [346].

17.2. Translating Potential Biological Mechanisms to Humans

Given the aforementioned studies in invertebrates and mammals, as well as all that has been discussed in this review concerning the increased understanding of the neurobiology of stress resilience, there seems to be potential in targeting several different pathways to increase resilience in vulnerable populations. In addition to the potential exercise and CR mimetic drugs discussed in the previous section, there is potential to induce changes in biological pathways that have been implicated in resilience. For instance, this review provides evidence for a multitude of systems that are viable pharmacologic targets for increasing stress resistance: targeting the serotonergic system in more acute ways than common SSRIs; the glutamatergic system, and in particular the NMDA receptors as through ketamine administration; AMPA receptors; mGluR receptors; the GABA system; other neurotransmitter systems; and the HPA axis. These biological pathways are complex and integrated, but research has been aimed at uncovering the most effective pathway implicated in stress resilience, which may soon aid in improving health, longevity, and mental capacity, especially in susceptible populations.

CONCLUSION

Frequent exposure to adverse life events is an important risk factor for developing psychopathology. But, why do some individuals succumb to debilitating psychiatric disease whereas others age normally? The notion that gene-by-environment interactions affect susceptibility to stress-related disorders, especially genes enhancing vulnerability is well known. Identifying these genes that confer resilience to stressful events in adulthood has become even more important. In this review, we have provided a brief overview of the biological mechanisms underlying stress resilience, and have explored how resilience changes throughout age. We have also offered suggestions for manipulating these mechanisms to improve resilience throughout life and prevent against stress-induced mood disorders. The topic of resilience as it pertains to the aged population is particularly relevant today, as the US Census Bureau population data projects that by 2030, adults aged 65 and older will constitute nearly 20% of the population [215]. Specifically, Baby Boomers, or those born between 1946 and 1964, are the fastest growing age group in the US [215]. Thus, current research is focusing on how to aid these Baby Boomers as they reach old age to remain healthy and resilient.

While we have focused this review on neurobiological factors of resilience, it is important to emphasize once more that other individualistic factors such as strength, the ability to cope with adversity, optimism, savoring positive experiences, and flexibility also largely contribute to resilient aging [215, 313, 347, 348], as these psychosocial processes are highly integrated with resilience. For example, coping strategies and other psychosocial influences that were briefly discussed in this review are critical components to maintaining resilience. Though a more in-depth discussion of these factors was beyond the scope of this review, they are not trivial and should be considered when evaluating potential psychological interventions in conjunction with biologically-based therapeutics to increase resilience in susceptible populations. In a similar vein, as effective interventions are developed to increase resilience, there must also be a parallel improvement in resilience scales in order to properly capture what elements of health and wellness are being impacted by these interventions. Currently, there are no “gold-standard” resilience scales, as Windle and colleagues found that each of the 15 measures of resilience they reviewed were missing information regarding psychometric properties [349]. Much work needs to be done to most effectively capture resiliency in individuals.

Resiliency research is still in its infancy. It is unlikely that neurobiological mechanisms involved in stress resiliency can be summarized to one single brain structure or target; it is more likely that circuits and systems dynamically drive stress resiliency. Identifying a common circuit involved in drug-induced stress resiliency, such as that induced by a prophylactic ketamine treatment, would be beneficial for the success of future drug development.

We have also discussed here the emerging series of studies that have been conducted to probe the relationship between resilience and aging. Importantly, we prefaced this discussion with a summary of what is currently known about biological mechanisms of stress resilience, and provide suggestions for how to move forward by combining what is currently known about resilience, and how it changes throughout the aging process, to inform more effective preventative resilience-enhancement therapies that is effective throughout life. Ultimately, prophylactic approaches may reduce the worldwide burden of disability, saving billions of dollars a year for countries, and years of emotional and physiological distress for both patients and their loved ones.

CONSENT FOR PUBLICATION

Not applicable.

Fig. (1).

Fig. (1)

Factors associated with stress-induced emotional disorders and those believed to characterize stress resilience. Combination of psychosocial environment, genetics and early life stress largely determine vulnerability to MDD and anxiety disorders. In contrast, the strategy to improve positive emotions, cognitive flexibility, optimism and active coping leads to promote stress resilience.

Table 1.

Role of the different 5-HT pathway-related key players in the stress-vulnerability and resilience in animals.

Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
Tph2 Rat Long Evans
10 weeks
Vulnerable (MS180 - maternal separation) SD 12 days Tph2 mRNA in DRN: ↑ Gardner et al., 2009
Resilient (MS15 - early handling) Tph2 mRNA in DRN: ↓
Monkey Cynomolgus
7-9 years
Resilient (normal
ovulatory cyclicity)
(vs anovulatory
vulnerable monkeys)
Mild psychosocial stress + mild diet ± moderate exercise 5 days Tph2-positive cells in DR: ↑ Bethea et al., 2013
Mouse ND
Adult
Tph2 KO Ø acute SPT: ↑
EPM: ↓ time in CA, ↑ distance/entries in OA
OF: ↑ time in center
FST: no difference
Gutknecht et al., 2015
CMS 3 weeks EPM: ↑ time in CA
OF: ↑ entries in center
FST: no difference
SERT Mouse B6.129(Cg)-Slc6a4tm1Kpl/J
ND
Heterozygous 5-Htt KO Ø EZM: ↑ time/entries in OA
FST: no difference
van den Hove et al., 2011
Prenatal RS 5 days EZM: ↑ time in OA
FST: no difference
Rat Wistar Han
ND
Vulnerable (↓ sucrose consumption >20%) CMS 2 weeks SERT expression: no difference Zurawek et al., 2016
Resilient (no difference in sucrose consumption) SERT expression: ↓ in VTA
Mouse C57BL/6J
3.5 months
Vulnerable CMS 10 days SERT expression: ↑ in PFC, hippocampus, striatum Couch et al., 2013
Resilient (> 65% preference for sucrose) SERT expression: no difference
5-HT1A Mouse mixed 129S6/Sv; C57B6; CBA
11-13 weeks
1A-Low mice
(vs 1A-High mice)
Ø acute OF/LDT/TST: no difference
FST: ↓ immobility
5-HT levels: no difference in PFC, hippocampus
Richardson-Jones et al., 2010
Oral gavage 4 weeks OF/FST: no difference
TST: ↓ immobility
Monkey Cynomolgus
7-9 years
Resilient (normal ovulatory cyclicity)
(vs anovulatory vulnerable monkeys)
Mild psychosocial stress + mild diet ± moderate exercise 5 days 5-HT1A-positive cells in DR: ↑ Bethea et al., 2013
Drugs Rat Lewis
8-9 weeks
Fluoxetine (7.5 mg/kg/d, 21 days) SD EPM: reverse defeat-induced ↓ entries/time in OA Berton et al., 1999
Mouse 129S6/SvEvTac
8-10 weeks
Fluoxetine (18 mg/kg/d, 21 days) SD 2 weeks EPM/FST: did not reverse defeat-induced ↑ time in CA and ↑ immobility Brachman et al., 2016
Mouse C57BL/6NTac CORT 3 weeks EPM/NSF/FST/ST: did not reverse CORT-induced ↑ time in CA, ↑ latency to feed, ↑ grooming latency and ↑ immobility
Mouse 129S1/SvImJ 10 weeks Escitalopram (4 mg/kg, 14 days) CMS 14 days HBT: ↑ time in center, ↓ head dipping
FST: ↑ immobility
Binder et al., 2011

Abbreviations: 5-HT: serotonin; CA: closed arms; CMS: chronic mild stress; CORT: corticosterone; DR: dorsal raphe; EPM/EZM: elevated plus/zero maze; FST: forced swim test; HBT: hole board test; KO: knock-out; LDT: light/dark test; ND: non-determined; NSF: novelty suppressed feeding; OA: open arms; OF: open field; PFC: prefrontal cortex; SD: social defeat; RS: restraint stress; SERT: serotonin transporter; ST: splash test; SPT: sucrose preference test; Tph: tryptophan hydroxylase; TST: tail suspension test; VTA: ventral tegmental area.

Table 2.

Role of the different glutamatergic pathway-related key players in the stress-vulnerability and resilience in animals.

Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
Mouse C57BL/6J
Adult
Vulnerable (↓ social avoidance vs non-stressed) SD 10 days GluR1 expression: ↑ in NAc
GluR2 expression: ↓ in NAc
Vialou et al., 2010
Resilient (no difference in social avoidance vs non-stressed) GluR1 expression: no difference in NAc
GluR2 expression: ↑ in NAc
Mouse mixed 129S1/Sv- p+Tyr+KitlSl−J/+; 129X1/SvJ ♂♀
2-10 months
GluR1 KO
(vs WT)
Ø acute EPM: ↑ entries in OA, ↓ times in CA
LDT: ↑ entries in light
FST: ↓ immobility
Fitzgerald et al., 2010
Mouse B6N.129-Gria1tm2Rsp/J ♂♀
15-30 weeks
Ø acute EPM/OF: ↑ time in OA/center
FST/TST: ↓ immobility
Social interaction: ↑ interaction
Maksimovic et al., 2014
Mouse C57BL/6
3-6 months
Ø acute LH testing: ↑ latency to escape, ↑ escape failure Chourbaji et al., 2008
Mouse C57BL/6
12 weeks
GluR1 KO (glutamatergic neurons)
(vs WT)
Ø acute EZM: no difference
LH testing: no difference
Vogt et al., 2014
Mouse C57BL/6N
Adult
GluR1 KO 5-HT neurons)
(vs WT)
Ø acute EZM/LDT: ↓ time/entries in OA/light
FST/TST: no difference
LH testing: no difference
Weber et al., 2015
Mouse mixed C57BL/6J; 129Sv
ND
GluR1 KO Ø acute EPM: ↓ time in OA Mead et al., 2006
Mouse C57BL/6J
Adult
Vulnerable (↓ social avoidance vs non-stressed) SD 10 days GluR2 expression: ↓ in NAc Vialou et al., 2010
Resilient (no difference in social avoidance vs non-stressed) GluR2 expression: ↑ in NAc
Mouse CD1
ND
GluR2 KO Ø acute EPM: ↑ time in OA, ↓ entries in CA Mead et al., 2006
Mouse C57BL/6J
Adult
mGluR2 KO Ø acute EPM/B&W Alley/OF: no difference De Filippis et al., 2015
Mouse C57BL/6J
6 weeks
Vulnerable (↓ sucrose preference/↑ immobility in FST vs non-stressed) CMS 28 days mGluR2 expression: ↓ in PFC Nasca et al., 2015
Resilient (no difference in sucrose preference/immobility vs non-stressed) mGluR2 expression: ↓ in PFC/hippocampus
Mouse ND ND mGluR2 KO FST: ↑ immobility
Coat-state: ↑ deterioration
Mouse mixed C57BL/6; 129Sv/Ev
Adult
GluK2 KO
(vs WT)
Ø acute EPM/OF:↑ time/entries in OA/center
FST: ↓ immobility
Shaltiel et al., 2008
Mouse C57BL/6J
Adult
mGluR3 KO
(vs WT)
Ø acute EPM/B&W Alley/OF: no difference De Filippis et al., 2015
Mouse mixed C57BL/6J; 129Sv
Adult
GluK4 KO
(vs WT)
Ø acute EZM: ↑ time/distance in OA
Marble burying: ↓ marble
NSF: ↓ latency to feed
FST: ↓ immobility
Catches et al., 2012
Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
Mouse C57BL/6J
12-20 weeks
Vulnerable (↓ social avoidance vs non-stressed) SD 12 days mGluR5 expression: ↓ in NAc Shin et al., 2015
Resilient (no difference in social avoidance vs non-stressed) mGluR5 expression: no difference in NAc
mGluR5 KO
(vs WT)
LH 3 days LH testing: ↑ latency to escape, ↑ escape failure
FST/TST: ↑ immobility
SD 3 days Social interaction: ↑ social avoidance
RS 3 days SPT: ↓
Mouse C57BL/6J
ND
mGluR5 KO
(vs WT)
Ø acute FST: ↓ immobility Li et al., 2006
Mouse C57BL/6J ND
10-14 weeks
mGluR7 KO
(vs WT)
Ø acute FST/TST: ↓ immobility
LDT: ↑ transitions
EPM: ↑ time/entries in OA
Cryan et al., 2003
Mouse mixed 129Sv; C57BL/6J; ICR
12 weeks
mGluR8 KO
(vs WT)
Ø acute EPM: ↓ time/entries in OA Linden et al., 2002
RS 30 min EPM: no difference
Mouse C57BL/6J ♂♀
> 10 weeks
NR2A KO
(vs WT)
Ø acute EPM/LDT: ↑ time/entries in OA/light
OF: ↑ time in center
FST/TST: ↓ immobility
Boyce-Rustay & Holmes, 2006
Mouse ND
50-70 days
NR2B KO (cortical neurons)
(vs WT)
Ø acute EPM: ↑ time in OA
FST/TST: ↓ immobility
SPT: no difference
Miller et al., 2014
Mouse C57BL/6J ND
3 months
NR2D KO
(vs WT)
Ø acute EPM: ↑ time/entries in OA
LDT: ↑ time in light
FST: ↓ immobility
Miyamoto et al., 2002
Mouse ND
Adult
Ø acute NSF/Marble burying: no difference
TST: ↑ immobility
SPT: ↓
Yamamoto et al., 2017
Mouse 129S6/SvEvTac
8-10 weeks
Ketamine (30 mg/kg)
1 week before stress
SD 2 weeks EPM: did not reverse defeat-induced ↑ time in CA
Social interaction: ↓ social avoidance
FST: reverse defeat-induced ↑ immobility
CFC/NSF: no difference
Brachman et al., 2016
Ketamine (30 mg/kg)
1 week before stress
LH testing: ↓ latency to escape
Mouse C57BL/6NTac Ketamine (90 mg/kg)
1 week before stress
CORT 3 weeks EPM: did not reverse CORT-induced ↑ time in CA
NSF/ST/FST: reverse CORT-induced ↑ latency to feed, ↑ grooming latency, ↑ immobility,
Mouse 129S6/SvEvTac
8 weeks
Ketamine (30 mg/kg)
24 hours, 1 week, 1 month before stress
3-shock CFC Fear extinction: ↓ freezing at 1 week
Fear reinstatement: no difference
McGowan et al., 2017
Rat Sprague Dawley
ND
Ketamine (10 mg/kg)
2 hours, 1 week, 2 weeks before stress
IS Social interaction: reverse IS-induced social avoidance at any of time Amat et al., 2016
Mouse/Rat NIH-Swiss mice
Sprague–Dawley rats

ND
LY392098 (0.5, 1 mg/kg)
1 hour before testing
FST: ↓ immobility Li et al., 2001

Abbreviations: 5-HT: serotonin; B&W: black & white; CA: closed arms; CFC: contextual fear conditioning; CMS: chronic mild stress; CORT: corticosterone; EPM/EZM: elevated plus/zero maze; FST: forced swim test; IS: inescapable shock; KO: knock-out; LDT: light/dark test; LH: learned helplessness; ND: non-determined; NAc: nucleus accumbens; ND: non-determined; NSF: novelty suppressed feeding; OA: open arms; OF: open field; PFC: prefrontal cortex; RS: restraint stress; SD: social defeat; SPT: sucrose preference test; ST: splash test; TST: tail suspension test; WT: wild-type.

Table 3.

Role of the different GABAergic pathway-related key players in the stress-vulnerability and resilience in animals.

Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
GABAergic neurons Rat Wistar rats
5–6 weeks
Resilient (< 10% within subject ↓ sucrose intake)
(vs vulnerable)
CMS 7 weeks CCK-positive GABAergic neurons: ↑ in ventral orbitofrontal cortex
NPY-positive GABAergic neurons: ↑ in lateral orbitofrontal cortex
Varga et al., 2017
GABA(A) receptors Mouse mixed C57BL/6; FVB ND α1 KO (CRH neurons)
(vs WT)
Ø acute EPM/OF: ↓ time in OA/center
TST: no difference
Fear extinction: ↑ freezing
Gafford et al., 2012
Mouse 129×1/SvJ
ND
α2 KO (vs WT) Ø acute FST: ↑ immobility in first 2 minutes, no difference in last 4 minutes
TST: ↑ immobility
NSF: trend to increase latency
Vollenweider et al., 2011

ND
FST: ↑ immobility in first 2 minutes
TST/NSF: no difference
Mouse mixed C57BL/6J; 129X1/SvJ
12 weeks
α3 KO (vs WT) Ø acute FST: ↓ immobility
Sucrose preference: no difference
Fiorelli et al., 2008
Mouse mixed C57BL/6; 129/SvJ ND γ2 KO (vs WT) Ø acute EPM/LDT: ↓ time/entries in OA/light
CFC: no difference
Crestani et al., 1999
Mouse mixed C57BL/6J; 129S1/X1 ND
Adult
γ2 KD (vs WT) Ø acute EPM: ↓ time/entries in OA Chandra et al., 2005
GABA(B) receptors Mouse BALB/c ♀♂
Adult
1A KO (vs WT) Ø acute Sucrose preference: no difference
FST: ↓ immobility
TST: ↑ immobility
Social interaction: ↓
O'Leary et al., 2014
MS 14 days Sucrose preference: ↓
FST: no difference
TST: ↑ immobility
EPM: no difference
SD 10 days Social interaction/sucrose preference: ↓
1B KO (vs WT) Ø acute Sucrose preference: no difference
FST/TST: ↓ immobility
Social interaction: no difference
MS 14 days Sucrose preference: no difference
FST/TST: ↓ immobility
EPM: no difference
SD 10 days Social interaction/sucrose preference: no difference
GAT Mouse ND
6-8 weeks
GAT-1 KO (vs WT) Ø acute OF/EZM: ↑ time/entries in center Gong et al., 2015
Mouse C57BL/6J
12–18 weeks
Ø acute FST/TST: ↓ immobility
EPM/LDT: ↑ time/entries in OA/ light
Liu et al., 2007
GAD Mouse mixed C57BL/6; CBA2
Adult
GAD65 KO (vs WT) Ø acute LDT: ↓ entries/distance in light
FST: ↓ immobility
Stork et al., 2000
Mouse C57BL/6
> 3 months
Ø acute OF/EZM: ↓ time in center/OA Kash et al., 1999
Mouse C57BL/6 ND
8-12 weeks
Ø acute Fear extinction: ↑ freezing Sangha et al., 2009
Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
GAD Mouse C57BL/6NTac
8-12 weeks
GAD65 haplodeficiency (vs WT) Ø acute Generalized contextual fear and anxiety: no difference
Pre-conditioning anxiety: no difference
Depression-like behavior: no difference
Muller et al., 2014
juvenile stress 3 days Generalized contextual fear and anxiety: no difference
Pre-conditioning anxiety: ↑ emotionality score
Depression-like behavior: no difference
isolation stress for months Generalized contextual fear and anxiety: ↑ emotionality score
Pre-conditioning anxiety: no difference
Depression-like behavior: no difference
JS + IS ND Generalized contextual fear and anxiety: ↑ emotionality score
Pre-conditioning anxiety: no difference
BZD Rat Wistar
Adult
midazolam (1.5 mg/kg)
5 minutes before stress
RS 30 minutes CFC: reverse stress-induced ↑ freezing Maldonado et al., 2011
midazolam (1.0 µg in amygdala)
5 minutes before stress
CFC: reverse stress-induced ↑ freezing
Rat Wistar
ND
midazolam (0.25 mg/kg)
30 minutes before re-exposure to stress
Fear reinstatement: no difference in high-anxiety
Fear reinstatement: ↓ re-conditioning freezing in low-anxiety
Skorzewska et al., 2015

Abbreviations: BZD: benzodiazepine; CCK: cholecystokinin; CMS: chronic mild stress; CRH: corticotrophin-releasing hormone; EPM/EZM: elevated plus/zero maze; FST: forced swim test; GAD: glutamate decarboxylase; GAT: GABA transporter; IS: isolation stress; JS: juvenile variable stress; KD: knockdown; KO: knock-out; LDT: light/dark test; MS: maternal separation; ND: non-determined; NPY: neuropeptide Y; OA: open arms; OF: open field; RS: restraint stress; SD: social defeat; TST: tail suspension test; WT: wild-type.

Table 4.

Role of the different HPA axis-related key players in the stress-vulnerability and resilience in animals.

Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
CRH Mouse C57BL/6
8 weeks
Vulnerable (↑ social avoidance vs non-stressed) SD 10 days Crh mRNA levels: ↑ in PVN Elliott et al., 2010
Resilient (no difference in social avoidance vs non-stressed) Crh mRNA levels: no difference in PVN
CRH1 Rat Sprague–Dawley
Adult
Vulnerable (mean escape latency times > 2 SD vs non-stressed) IS 2 hours CRH1 expression: no difference in LC, amygdala, striatum
CRH1 expression: ↓ in cortex
Taneja et al., 2011
Resilient (escape latency times within 2 SD vs non-stressed) CRH1 expression: ↓ in LC, amygdala, cortex
CRH1 expression: no difference in striatum
Mouse C57BL/6 ♂♀
Adult
CRH1 KO (in pituitary) Ø acute LDT: ↑ time in light
EPM: ↑ time/entries in OA
Smith et al., 1998
Mouse mixed 129/Sv; C57BL/6
3-5 months
CRH1 KO (in anterior forebrain and limbic structures) Ø acute LDT: ↑ time/entries in light
EPM: ↑ entries in OA
Muller et al., 2003
CORT Mouse C57BL/6
8-12 weeks
Vulnerable (↑ social avoidance vs non-stressed) SD 10 days CORT level after DEX challenge: ↑ Jochems et al., 2015
Resilient (no difference in social avoidance vs non-stressed) CORT level after DEX challenge: no difference
GR Mouse mixed C57BL/6; 129SV; CBA ND
6 months
FBGRKO (forebrain-specific GR KO)
(vs WT)
Ø acute FST/TST: ↓ immobility
SPT: ↓
Boyle et al., 2005
Mouse mixed C57BL/6; FVB/N
3-6 months
GR-heterozygous KO (50% reduced GR gene) Ø acute EZM/LDT/CFC: no difference
FST: no difference
CORT level after DEX challenge: ↑
Ridder et al., 2005
LH 2 days LH testing: ↑ latency to escape, ↑ escape failure
RS 30 min CORT level: ↑
YGR (two-fold increased GR gene) Ø acute EZM/LDT/CFC: no difference
FST: no difference
CORT level after DEX challenge: ↓
LH 2 days LH testing: ↓ latency to escape,
↓ escape failure
RS 30 min CORT level: ↓
Mouse mixed 129/SvJ; C57BL/6 ND
17-22 months
FKBP5 KO
(vs WT)
Ø acute EPM/LDT: no difference
FST/TST: ↓ immobility
O'Leary et al., 2011
RS 10 min CORT level: ↓
Mouse mixed 129/SvJ; C57BL/6
10-16 weeks
FKBP5 KO
(vs WT)
Ø acute EPM/LDT/FST: no difference
CORT level: no difference
Hartmann et al., 2012
SD 21 days EPM: no difference
FST: ↑ swimming
CORT level: ↓
Species Strain Sex/Age Model/Treatment Stress Duration Results (vs Control) Refs.
GR Mouse C57BL/6
8-12 weeks
Vulnerable (interaction ratio < 100) SD 10 days CORT level after DEX challenge: ↑
Nuclear GR protein expression/GR-Hsp90 interaction: ↑
Hsp90 acetylation: ↓
Jochems et al., 2015
Resilient (interaction ratio ≥ 100) CORT level after DEX challenge: no difference
Nuclear GR protein expression/GR-Hsp90 interaction/Hsp90 Acetylation: no difference
viral overexpression of acetyl-mimic Hsp90 mutant
(in DRN) (vs WT)
SD 10 days Social interaction: no difference
Mouse 129/Sv
3-12 months
HDAC6 KO Ø acute OF: no difference
EPM: ↑ time/entries in OA
TST: ↓ immobility
Fukada et al., 2012
Mouse C57BL/6
8-12 weeks
HDAC6Pet1Cre KO (in 5-HT neurons) Ø acute FST/TST: ↓ immobility Espallergues et al., 2012
SD 10 days Social interaction: no difference
MR Mouse C57BL/6J
3 months
MR overexpressing
(vs WT)
Ø acute OF: ↑ time/entries in center
LDT: ↓ latency to enter in light, ↑ entries in light
CORT level (before and after stress): no difference
Lai et al., 2007
Rat Wistar
10 weeks
MR overexpressing (in BLA)
(vs WT)
Ø acute EPM: ↑ entries in OA, ↑ time in OA (trend) Mitra et al., 2009
RS 2 hours CORT level: ↓
EPM: ↑ entries in OA, ↑ time in OA (trend)
OF: ↑ time in center
Drugs Mouse 129/Sv
3-12 months
NCT-14b (HDAC6 inhibitor) Ø acute TST: ↓ immobility Fukada et al., 2012
Mouse C57BL/6
8-12 weeks
ACY-738 (selective HDAC6 inhibitor, 5 mg/kg)
10 days before stress and 10 days throughout the stress
SD 10 days Social interaction: reverse stress-induced ↑ social avoidance Jochems et al., 2015

Abbreviations: 5-HT: serotonin; CORT: corticosterone; BLA: basolateral amygdala; CFC: contextual fear conditioning; CORT: corticosterone; CRH: corticotrophin-releasing hormone; DEX: dexamethasone; DRN: dorsal raphe nucleus; EPM/EZM: elevated plus/zero maze; FKBP5: FK506-binding protein 51; FST: forced swim test; GR: glucocorticoid receptor; HDAC: histone deacetylase; HPA: hypothalamic-pituitary-adrenal; Hsp90: heat shock protein; IS: inescapable stress; KO: knock-out; LC: locus cœruleus; LDT: light/dark test; LH: learned helplessness; MR: mineralocorticoid receptor; ND: non-determined; OA: open arms; OF: open field; PVN: paraventricular nucleus; RS: restraint stress; SD: social defeat; SPT: sucrose preference test; TST: tail suspension test; WT: wild-type.

ACKNOWLEDGEMENTS

Declared none.

CONFLICT OF INTEREST

Dr. Christine A. Denny and Josephine C. McGowan are named on non-provisional patent applications for the prophylactic use of ketamine against stress-related psychiatric disorders.

REFERENCES

  • 1.Reddy M.S. Depression: the disorder and the burden. Indian J. Psychol. Med. 2010;32(1):1–2. doi: 10.4103/0253-7176.70510. [http://dx.doi.org/10.4103/0253-7176.70510]. [PMID: 21799550]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Andlin-Sobocki P., Jönsson B., Wittchen H.U., Olesen J. Cost of disorders of the brain in Europe. Eur. J. Neurol. 2005;12(Suppl. 1):1–27. doi: 10.1111/j.1468-1331.2005.01202.x. [http://dx.doi.org/10.1111/j.1468-1331.2005.01202.x]. [PMID: 15877774]. [DOI] [PubMed] [Google Scholar]
  • 3.Greenberg P.E., Kessler R.C., Birnbaum H.G., Leong S.A., Lowe S.W., Berglund P.A., Corey-Lisle P.K. The economic burden of depression in the United States: how did it change between 1990 and 2000? J. Clin. Psychiatry. 2003;64(12):1465–1475. doi: 10.4088/jcp.v64n1211. [http://dx.doi.org/10.4088/JCP.v64n1211]. [PMID: 14728109]. [DOI] [PubMed] [Google Scholar]
  • 4.Greenberg P.E., Sisitsky T., Kessler R.C., Finkelstein S.N., Berndt E.R., Davidson J.R., Ballenger J.C., Fyer A.J. The economic burden of anxiety disorders in the 1990s. J. Clin. Psychiatry. 1999;60(7):427–435. doi: 10.4088/jcp.v60n0702. [http://dx.doi.org/10.4088/JCP.v60n0702]. [PMID: 10453795]. [DOI] [PubMed] [Google Scholar]
  • 5.Gomoll B.P., Kumar A. Managing anxiety associated with neurodegenerative disor-ders. 2015. [DOI] [PMC free article] [PubMed]
  • 6.Kessler R.C., Amminger G.P., Aguilar-Gaxiola S., Alonso J., Lee S., Ustün T.B. Age of onset of mental disorders: a review of recent literature. Curr. Opin. Psychiatry. 2007;20(4):359–364. doi: 10.1097/YCO.0b013e32816ebc8c. [http:// dx.doi.org/10.1097/YCO.0b013e32816ebc8c]. [PMID: 17551351]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Forlani C., Morri M., Ferrari B., Dalmonte E., Menchetti M., De Ronchi D., Atti A.R. Prevalence and gender differences in late-life depression: a population-based study. Am. J. Geriatr. Psychiatry. 2014;22(4):370–380. doi: 10.1016/j.jagp.2012.08.015. [http://dx.doi.org/10.1016/j.jagp. 2012.08.015]. [PMID: 23567427]. [DOI] [PubMed] [Google Scholar]
  • 8.Johnson S.A., Fournier N.M., Kalynchuk L.E. Effect of different doses of corticosterone on depression-like behavior and HPA axis responses to a novel stressor. Behav. Brain Res. 2006;168(2):280–288. doi: 10.1016/j.bbr.2005.11.019. [http://dx.doi.org/10.1016/j.bbr.2005.11.019]. [PMID: 16386319]. [DOI] [PubMed] [Google Scholar]
  • 9.Kudielka B.M., Buske-Kirschbaum A., Hellhammer D.H., Kirschbaum C. HPA axis responses to laboratory psychosocial stress in healthy elderly adults, younger adults, and children: impact of age and gender. Psychoneuroendocrinology. 2004;29(1):83–98. doi: 10.1016/s0306-4530(02)00146-4. [http://dx.doi.org/10.1016/S0306-4530(02)00146-4]. [PMID: 14575731]. [DOI] [PubMed] [Google Scholar]
  • 10.Southwick S.M., Vythilingam M., Charney D.S. The psychobiology of depression and resilience to stress: implications for prevention and treatment. Annu. Rev. Clin. Psychol. 2005;1:255–291. doi: 10.1146/annurev.clinpsy.1.102803.143948. [http://dx.doi.org/10.1146/annurev.clinpsy.1.102803.143948]. [PMID: 17716089]. [DOI] [PubMed] [Google Scholar]
  • 11.Gloria C.T., Steinhardt M.A. Relationships among positive emotions, coping, resilience and mental health. Stress Health. 2016;32(2):145–156. doi: 10.1002/smi.2589. [http://dx.doi.org/10.1002/smi.2589]. [PMID: 24962138]. [DOI] [PubMed] [Google Scholar]
  • 12.Galatzer-Levy I.R., Brown A.D., Henn-Haase C., Metzler T.J., Neylan T.C., Marmar C.R. Positive and negative emotion prospectively predict trajectories of resilience and distress among high-exposure police officers. Emotion. 2013;13(3):545–553. doi: 10.1037/a0031314. [http://dx.doi.org/10.1037/a0031314]. [PMID: 23339621]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Breton J.J., Labelle R., Berthiaume C., Royer C., St-Georges M., Ricard D., Abadie P., Gérardin P., Cohen D., Guilé J.M. Protective factors against depression and suicidal behaviour in adolescence. Can. J. Psychiatry. 2015;60(2) Suppl. 1:S5–S15. [PMID: 25886672]. [PMC free article] [PubMed] [Google Scholar]
  • 14.Davidovich S., Collishaw S., Thapar A.K., Harold G., Thapar A., Rice F. Do better executive functions buffer the effect of current parental depression on adolescent depressive symptoms? J. Affect. Disord. 2016;199:54–64. doi: 10.1016/j.jad.2016.03.049. [http://dx.doi.org/10.1016/j.jad. 2016.03.049]. [PMID: 27085164]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Burton N.W., Pakenham K.I., Brown W.J. Feasibility and effectiveness of psychosocial resilience training: a pilot study of the READY program. Psychol. Health Med. 2010;15(3):266–277. doi: 10.1080/13548501003758710. [http:// dx.doi.org/10.1080/13548501003758710]. [PMID: 20480432]. [DOI] [PubMed] [Google Scholar]
  • 16.Waite P.J., Richardson G.E. Determining the efficacy of resiliency training in the work site. J. Allied Health. 2004;33(3):178–183. [PMID: 15503750]. [PubMed] [Google Scholar]
  • 17.Steensma H., Den Heijer M., Stallen V. [DOI] [PubMed]
  • 18.Childs E., de Wit H. Regular exercise is associated with emotional resilience to acute stress in healthy adults. Front. Physiol. 2014;5:161. doi: 10.3389/fphys.2014.00161. [http://dx.doi.org/10.3389/fphys.2014.00161]. [PMID: 24822048]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dell’Osso L., Carmassi C., Mucci F., Marazziti D. Depression, serotonin and tryptophan. Curr. Pharm. Des. 2016;22(8):949–954. doi: 10.2174/1381612822666151214104826. [http://dx.doi.org/10.2174/1381612822666151214104826]. [PMID: 26654774]. [DOI] [PubMed] [Google Scholar]
  • 20.Richell R.A., Deakin J.F., Anderson I.M. Effect of acute tryptophan depletion on the response to controllable and uncontrollable noise stress. Biol. Psychiatry. 2005;57(3):295–300. doi: 10.1016/j.biopsych.2004.10.010. [http://dx.doi. org/10.1016/j.biopsych.2004.10.010]. [PMID: 15691531]. [DOI] [PubMed] [Google Scholar]
  • 21.Gutknecht L., Popp S., Waider J., Sommerlandt F.M., Göppner C., Post A., Reif A., van den Hove D., Strekalova T., Schmitt A., Colaςo M.B., Sommer C., Palme R., Lesch K.P. Interaction of brain 5-HT synthesis deficiency, chronic stress and sex differentially impact emotional behavior in Tph2 knockout mice. Psychopharmacology (Berl.) 2015;232(14):2429–2441. doi: 10.1007/s00213-015-3879-0. [http://dx.doi. org/10.1007/s00213-015-3879-0]. [PMID: 25716307]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gardner K.L., Hale M.W., Oldfield S., Lightman S.L., Plotsky P.M., Lowry C.A. Adverse experience during early life and adulthood interact to elevate tph2 mRNA expression in serotonergic neurons within the dorsal raphe nucleus. Neuroscience. 2009;163(4):991–1001. doi: 10.1016/j.neuroscience.2009.07.055. [http://dx.doi.org/10.1016/j.neuroscience.2009. 07.055]. [PMID: 19647049]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Stein M.B., Campbell-Sills L., Gelernter J. Genetic variation in 5HTTLPR is associated with emotional resilience. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2009;150B(7):900–906. doi: 10.1002/ajmg.b.30916. [http:// dx.doi.org/10.1002/ajmg.b.30916]. [PMID: 19152387]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gunthert K.C., Conner T.S., Armeli S., Tennen H., Covault J., Kranzler H.R. Serotonin transporter gene polymorphism (5-HTTLPR) and anxiety reactivity in daily life: a daily process approach to gene-environment interaction. Psychosom. Med. 2007;69(8):762–768. doi: 10.1097/PSY.0b013e318157ad42. [http://dx.doi.org/10.1097/PSY.0b013e318157ad42]. [PMID: 17942837]. [DOI] [PubMed] [Google Scholar]
  • 25.Markus C.R., De Raedt R. Differential effects of 5-HTTLPR genotypes on inhibition of negative emotional information following acute stress exposure and tryptophan challenge. Neuropsychopharmacology. 2011;36(4):819–826. doi: 10.1038/npp.2010.221. [http://dx.doi.org/10.1038/ npp.2010.221]. [PMID: 21150915]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Caspi A., Sugden K., Moffitt T.E., Taylor A., Craig I.W., Harrington H., McClay J., Mill J., Martin J., Braithwaite A., Poulton R. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301(5631):386–389. doi: 10.1126/science.1083968. [http://dx.doi.org/10.1126/science.1083968]. [PMID: 12869766]. [DOI] [PubMed] [Google Scholar]
  • 27.Kaufman J., Yang B.Z., Douglas-Palumberi H., Grasso D., Lipschitz D., Houshyar S., Krystal J.H., Gelernter J. Brain-derived neurotrophic factor-5-HTTLPR gene interactions and environmental modifiers of depression in children. Biol. Psychiatry. 2006;59(8):673–680. doi: 10.1016/j.biopsych.2005.10.026. [http://dx.doi.org/10.1016/j.biopsych.2005.10.026]. [PMID: 16458264]. [DOI] [PubMed] [Google Scholar]
  • 28.Lesch K.P., Bengel D., Heils A., Sabol S.Z., Greenberg B.D., Petri S., Benjamin J., Müller C.R., Hamer D.H., Murphy D.L. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science. 1996;274(5292):1527–1531. doi: 10.1126/science.274.5292.1527. [http://dx.doi.org/10.1126/science.274.5292. 1527]. [PMID: 8929413]. [DOI] [PubMed] [Google Scholar]
  • 29.Telch M.J., Beevers C.G., Rosenfield D., Lee H.J., Reijntjes A., Ferrell R.E., Hariri A.R. 5-HTTLPR genotype potentiates the effects of war zone stressors on the emergence of PTSD, depressive and anxiety symptoms in soldiers deployed to Iraq. World Psychiatry. 2015;14(2):198–206. doi: 10.1002/wps.20215. [http://dx.doi.org/10.1002/wps.20215]. [PMID: 26043338]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.van den Hove D.L., Jakob S.B., Schraut K.G., Kenis G., Schmitt A.G., Kneitz S., Scholz C.J., Wiescholleck V., Ortega G., Prickaerts J., Steinbusch H., Lesch K.P. Differential effects of prenatal stress in 5-Htt deficient mice: towards molecular mechanisms of gene × environment interactions. PLoS One. 2011;6(8):e22715. doi: 10.1371/journal.pone.0022715. [http://dx.doi.org/10.1371/journal.pone.0022715]. [PMID: 21857948]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Couch Y., Anthony D.C., Dolgov O., Revischin A., Festoff B., Santos A.I., Steinbusch H.W., Strekalova T. Microglial activation, increased TNF and SERT expression in the prefrontal cortex define stress-altered behaviour in mice susceptible to anhedonia. Brain Behav. Immun. 2013;29:136–146. doi: 10.1016/j.bbi.2012.12.017. [http://dx.doi.org/ 10.1016/j.bbi.2012.12.017]. [PMID: 23305936]. [DOI] [PubMed] [Google Scholar]
  • 32.Zurawek D. Reciprocal microRNA expression in mesocortical circuit and its interplay with serotonin transporter define resilient rats in the chronic mild stress. Mol. Neurobiol. 2017;54:5741. doi: 10.1007/s12035-016-0107-9. [PMID: 27660265]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bethea C.L., Phu K., Reddy A.P., Cameron J.L. The effect of short-term stress on serotonin gene expression in high and low resilient macaques. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2013;44:143–153. doi: 10.1016/j.pnpbp.2013.01.013. [http://dx.doi.org/10.1016/j.pnpbp.2013.01.013]. [PMID: 23357537]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bethea C.L., Streicher J.M., Mirkes S.J., Sanchez R.L., Reddy A.P., Cameron J.L. Serotonin-related gene expression in female monkeys with individual sensitivity to stress. Neuroscience. 2005;132(1):151–166. doi: 10.1016/j.neuroscience.2004.11.022. [http://dx.doi.org/10.1016/j.neuroscience.2004.11. 022]. [PMID: 15780474]. [DOI] [PubMed] [Google Scholar]
  • 35.David D.J., Gardier A.M. The pharmacological basis of the serotonin system: Application to antidepressant response. Encephale. 2016;42(3):255–263. doi: 10.1016/j.encep.2016.03.012. [http://dx.doi.org/10.1016/j.encep.2016.03.012]. [PMID: 27112704]. [DOI] [PubMed] [Google Scholar]
  • 36.Samuels B.A., Mendez-David I., Faye C., David S.A. ; Pierz K.A., Gardier A.M., Hen R., David D.J. Serotonin 1A and serotonin 4 receptors: Essential mediators of the neurogenic and behavioral actions of antidepressants. Neuroscientist. 2016;22(1):26–45. doi: 10.1177/1073858414561303. [http://dx.doi.org/10.1177/1073858414561303]. [PMID: 25488850]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lemonde S., Turecki G., Bakish D., Du L., Hrdina P.D., Bown C.D., Sequeira A., Kushwaha N., Morris S.J., Basak A., Ou X.M., Albert P.R. Impaired repression at a 5-hydroxytryptamine 1A receptor gene polymorphism associated with major depression and suicide. J. Neurosci. 2003;23(25):8788–8799. doi: 10.1523/JNEUROSCI.23-25-08788.2003. [PMID: 14507979]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Richardson-Jones J.W., Craige C.P., Guiard B.P., Stephen A., Metzger K.L., Kung H.F., Gardier A.M., Dranovsky A., David D.J., Beck S.G., Hen R., Leonardo E.D. 5-HT1A autoreceptor levels determine vulnerability to stress and response to antidepressants. Neuron. 2010;65(1):40–52. doi: 10.1016/j.neuron.2009.12.003. [http://dx.doi.org/10.1016/ j.neuron.2009.12.003]. [PMID: 20152112]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Attar-Lévy D., Martinot J.L., Blin J., Dao-Castellana M.H., Crouzel C., Mazoyer B., Poirier M.F., Bourdel M.C., Aymard N., Syrota A., Féline A. The cortical serotonin2 receptors studied with positron-emission tomography and [18F]-setoperone during depressive illness and antidepressant treatment with clomipramine. Biol. Psychiatry. 1999;45(2):180–186. doi: 10.1016/s0006-3223(98)00007-9. [http://dx.doi.org/10.1016/ S0006-3223(98)00007-9]. [PMID: 9951565]. [DOI] [PubMed] [Google Scholar]
  • 40.Biver F., Wikler D., Lotstra F., Damhaut P., Goldman S., Mendlewicz J. Serotonin 5-HT2 receptor imaging in major depression: focal changes in orbito-insular cortex. Br. J. Psychiatry. 1997;171:444–448. doi: 10.1192/bjp.171.5.444. [http://dx.doi.org/10.1192/bjp.171.5.444]. [PMID: 9463603]. [DOI] [PubMed] [Google Scholar]
  • 41.Larisch R., Klimke A., Mayoral F., Hamacher K., Herzog H.R., Vosberg H., Tosch M., Gaebel W., Rivas F., Coenen H.H., Müller-Gärtner H.W. Disturbance of serotonin 5HT2 receptors in remitted patients suffering from hereditary depressive disorder. Nucl. Med. (Stuttg.) 2001;40(4):129–134. [PMID: 11556203]. [PubMed] [Google Scholar]
  • 42.Mintun M.A., Sheline Y.I., Moerlein S.M., Vlassenko A.G., Huang Y., Snyder A.Z. Decreased hippocampal 5-HT2A receptor binding in major depressive disorder: In vivo measurement with [18F]altanserin positron emission tomography. Biol. Psychiatry. 2004;55(3):217–224. doi: 10.1016/j.biopsych.2003.08.015. [http://dx.doi.org/10.1016/j.biopsych.2003. 08.015]. [PMID: 14744461]. [DOI] [PubMed] [Google Scholar]
  • 43.Sheline Y.I., Mintun M.A., Barch D.M., Wilkins C., Snyder A.Z., Moerlein S.M. Decreased hippocampal 5-HT(2A) receptor binding in older depressed patients using [18F]altanserin positron emission tomography. Neuropsychopharmacology. 2004;29(12):2235–2241. doi: 10.1038/sj.npp.1300555. [http://dx.doi.org/10.1038/sj.npp.1300555]. [PMID: 15367923]. [DOI] [PubMed] [Google Scholar]
  • 44.Adams K.H., Hansen E.S., Pinborg L.H., Hasselbalch S.G., Svarer C., Holm S., Bolwig T.G., Knudsen G.M. Patients with obsessive-compulsive disorder have increased 5-HT2A receptor binding in the caudate nuclei. Int. J. Neuropsychopharmacol. 2005;8(3):391–401. doi: 10.1017/S1461145705005055. [http://dx.doi.org/10.1017/S1461145705005055]. [PMID: 15801987]. [DOI] [PubMed] [Google Scholar]
  • 45.Berton O., Aguerre S., Sarrieau A., Mormede P., Chaouloff F. Differential effects of social stress on central serotonergic activity and emotional reactivity in Lewis and spontaneously hypertensive rats. Neuroscience. 1998;82(1):147–159. doi: 10.1016/s0306-4522(97)00282-0. [http://dx.doi.org/10. 1016/S0306-4522(97)00282-0]. [PMID: 9483511]. [DOI] [PubMed] [Google Scholar]
  • 46.Farhang S., Barar J., Fakhari A., Mesgariabbasi M., Khani S., Omidi Y., Farnam A. Asymmetrical expression of BDNF and NTRK3 genes in frontoparietal cortex of stress-resilient rats in an animal model of depression. Synapse. 2014;68(9):387–393. doi: 10.1002/syn.21746. [http://dx.doi.org/10.1002/syn.21746]. [PMID: 24753016]. [DOI] [PubMed] [Google Scholar]
  • 47.Blier P., Keller M.B., Pollack M.H., Thase M.E., Zajecka J.M., Dunner D.L. Preventing recurrent depression: long-term treatment for major depressive disorder. J. Clin. Psychiatry. 2007;68(3):e06. [http://dx.doi.org/10.4088/JCP.0307e06]. [PMID: 17388700]. [PubMed] [Google Scholar]
  • 48.Hirschfeld R.M. Guidelines for the long-term treatment of depression. J. Clin. Psychiatry. 1994;55(Suppl.):61–69. [PMID: 7814359]. [PubMed] [Google Scholar]
  • 49.Gilaberte I., Montejo A.L., de la Gandara J., Perez-Sola V., Bernardo M., Massana J., Martin-Santos R., Santiso A., Noguera R., Casais L., Perez-Camo V., Arias M., Judge R. Fluoxetine in the prevention of depressive recurrences: a double-blind study. J. Clin. Psychopharmacol. 2001;21(4):417–424. doi: 10.1097/00004714-200108000-00009. [http://dx.doi.org/10.1097/00004714-200108000-00009]. [PMID: 11476126]. [DOI] [PubMed] [Google Scholar]
  • 50.Montgomery S.A., Dufour H., Brion S., Gailledreau J., Laqueille X., Ferrey G., Moron P., Parant-Lucena N., Singer L., Danion J.M. The prophylactic efficacy of fluoxetine in unipolar depression. Br. J. Psychiatry Suppl. 1988;(3):69–76. [PMID: 3150694]. [PubMed] [Google Scholar]
  • 51.Montgomery S.A., Dunbar G. Paroxetine is better than placebo in relapse prevention and the prophylaxis of recurrent depression. Int. Clin. Psychopharmacol. 1993;8(3):189–195. doi: 10.1097/00004850-199300830-00009. [http://dx.doi.org/ 10.1097/00004850-199300830-00009]. [PMID: 8263317]. [DOI] [PubMed] [Google Scholar]
  • 52.Montgomery S.A., Rasmussen J.G., Tanghøj P. A 24-week study of 20 mg citalopram, 40 mg citalopram, and placebo in the prevention of relapse of major depression. Int. Clin. Psychopharmacol. 1993;8(3):181–188. doi: 10.1097/00004850-199300830-00008. [http://dx.doi.org/10.1097/00004850-199300830-00008]. [PMID: 8263316]. [DOI] [PubMed] [Google Scholar]
  • 53.Doogan D.P., Caillard V. Sertraline in the prevention of depression. Br. J. Psychiatry. 1992;160:217–222. doi: 10.1192/bjp.160.2.217. [http://dx.doi.org/10. 1192/bjp.160.2.217]. [PMID: 1540762]. [DOI] [PubMed] [Google Scholar]
  • 54.Peselow E.D., Tobia G., Karamians R., Pizano D. IsHak, W.W. Prophylactic efficacy of fluoxetine, escitalopram, sertraline, paroxetine, and concomitant psychotherapy in major depressive disorder: outcome after long-term follow-up. Psychiatry Res. 2015;225(3):680–686. doi: 10.1016/j.psychres.2014.11.022. [http://dx.doi.org/10.1016/j.psychres.2014.11. 022]. [PMID: 25496869]. [DOI] [PubMed] [Google Scholar]
  • 55.Berton O., Durand M., Aguerre S., Mormède P., Chaouloff F. Behavioral, neuroendocrine and serotonergic consequences of single social defeat and repeated fluoxetine pretreatment in the Lewis rat strain. Neuroscience. 1999;92(1):327–341. doi: 10.1016/s0306-4522(98)00742-8. [http://dx.doi.org/ 10.1016/S0306-4522(98)00742-8]. [PMID: 10392854]. [DOI] [PubMed] [Google Scholar]
  • 56.Baek I.S., Park J.Y., Han P.L. Chronic antidepressant treatment in normal mice induces anxiety and impairs stress-coping ability. Exp. Neurobiol. 2015;24(2):156–168. doi: 10.5607/en.2015.24.2.156. [http://dx.doi.org/10.5607/ en.2015.24.2.156]. [PMID: 26113795]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Brachman R.A., McGowan J.C., Perusini J.N., Lim S.C. ; Pham T.H., Faye C., Gardier A.M., Mendez-David I., David D.J., Hen R., Denny C.A. Ketamine as a prophylactic against stress-induced depressive-like behavior. Biol. Psychiatry. 2016;79(9):776–786. doi: 10.1016/j.biopsych.2015.04.022. [http://dx.doi.org/10.1016/j.biopsych.2015.04.022]. [PMID: 26037911]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Binder E., Malki K., Paya-Cano J.L., Fernandes C., Aitchison K.J., Mathé A.A., Sluyter F., Schalkwyk L.C. Antidepressants and the resilience to early-life stress in inbred mouse strains. Pharmacogenet. Genomics. 2011;21(12):779–789. doi: 10.1097/FPC.0b013e32834b3f35. [http://dx. doi.org/10.1097/FPC.0b013e32834b3f35]. [PMID: 22016050]. [DOI] [PubMed] [Google Scholar]
  • 59.Trullas R., Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur. J. Pharmacol. 1990;185(1):1–10. doi: 10.1016/0014-2999(90)90204-j. [http://dx.doi.org/10.1016/0014-2999(90) 90204-J]. [PMID: 2171955]. [DOI] [PubMed] [Google Scholar]
  • 60.Hashimoto K., Sawa A., Iyo M. Increased levels of glutamate in brains from patients with mood disorders. Biol. Psychiatry. 2007;62(11):1310–1316. doi: 10.1016/j.biopsych.2007.03.017. [http://dx.doi.org/10.1016/j.biopsych.2007.03. 017]. [PMID: 17574216]. [DOI] [PubMed] [Google Scholar]
  • 61.Sanacora G., Gueorguieva R., Epperson C.N., Wu Y.T., Appel M., Rothman D.L., Krystal J.H., Mason G.F. Subtype-specific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Arch. Gen. Psychiatry. 2004;61(7):705–713. doi: 10.1001/archpsyc.61.7.705. [http://dx.doi.org/10.1001/archpsyc.61.7.705]. [PMID: 15237082]. [DOI] [PubMed] [Google Scholar]
  • 62.Hasler G., van der Veen J.W., Tumonis T., Meyers N., Shen J., Drevets W.C. Reduced prefrontal glutamate/glutamine and gamma-aminobutyric acid levels in major depression determined using proton magnetic resonance spectroscopy. Arch. Gen. Psychiatry. 2007;64(2):193–200. doi: 10.1001/archpsyc.64.2.193. [http://dx.doi.org/10.1001/archpsyc. 64.2.193]. [PMID: 17283286]. [DOI] [PubMed] [Google Scholar]
  • 63.Yildiz-Yesiloglu A., Ankerst D.P. Review of 1H magnetic resonance spectroscopy findings in major depressive disorder: a meta-analysis. Psychiatry Res. 2006;147(1):1–25. doi: 10.1016/j.pscychresns.2005.12.004. [http://dx.doi.org/ 10.1016/j.pscychresns.2005.12.004]. [PMID: 16806850]. [DOI] [PubMed] [Google Scholar]
  • 64.Luykx J.J., Laban K.G., van den Heuvel M.P., Boks M.P., Mandl R.C., Kahn R.S., Bakker S.C. Region and state specific glutamate downregulation in major depressive disorder: a meta-analysis of (1)H-MRS findings. Neurosci. Biobehav. Rev. 2012;36(1):198–205. doi: 10.1016/j.neubiorev.2011.05.014. [http://dx.doi.org/10.1016/j.neubiorev.2011.05. 014]. [PMID: 21672551]. [DOI] [PubMed] [Google Scholar]
  • 65.Rosenberg D.R., Macmaster F.P., Mirza Y., Smith J.M., Easter P.C., Banerjee S.P., Bhandari R., Boyd C., Lynch M., Rose M., Ivey J., Villafuerte R.A., Moore G.J., Renshaw P. Reduced anterior cingulate glutamate in pediatric major depression: a magnetic resonance spectroscopy study. Biol. Psychiatry. 2005;58(9):700–704. doi: 10.1016/j.biopsych.2005.05.007. [http://dx.doi.org/10.1016/j.biopsych.2005.05.007]. [PMID: 16084860]. [DOI] [PubMed] [Google Scholar]
  • 66.Hermens D.F., Chitty K.M., Lee R.S., Tickell A., Haber P.S., Naismith S.L., Hickie I.B., Lagopoulos J. Hippocampal glutamate is increased and associated with risky drinking in young adults with major depression. J. Affect. Disord. 2015;186:95–98. doi: 10.1016/j.jad.2015.07.009. [http://dx.doi.org/10.1016/j.jad.2015.07.009]. [PMID: 26233319]. [DOI] [PubMed] [Google Scholar]
  • 67.de Diego-Adeliño J., Portella M.J., Gómez-Ansón B., López-Moruelo O., Serra-Blasco M., Vives Y., Puigdemont D., Pérez-Egea R., Álvarez E., Pérez V. Hippocampal abnormalities of glutamate/glutamine, N-acetylaspartate and choline in patients with depression are related to past illness burden. J. Psychiatry Neurosci. 2013;38(2):107–116. doi: 10.1503/jpn.110185. [http://dx.doi.org/10.1503/jpn.110185]. [PMID: 23425950]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Block W., Träber F., von Widdern O., Metten M., Schild H., Maier W., Zobel A., Jessen F. Proton MR spectroscopy of the hippocampus at 3 T in patients with unipolar major depressive disorder: correlates and predictors of treatment response. Int. J. Neuropsychopharmacol. 2009;12(3):415–422. doi: 10.1017/S1461145708009516. [http://dx.doi.org/10. 1017/S1461145708009516]. [PMID: 18845018]. [DOI] [PubMed] [Google Scholar]
  • 69.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(5):1650–1657. doi: 10.1111/j.1471-4159.1993.tb13387.x. [http://dx.doi.org/10.1111/j.1471-4159.1993.tb13387.x]. [PMID: 8097232]. [DOI] [PubMed] [Google Scholar]
  • 70.Ghasemi M., Phillips C., Trillo L., De Miguel Z., Das D., Salehi A. The role of NMDA receptors in the pathophysiology and treatment of mood disorders. Neurosci. Biobehav. Rev. 2014;47:336–358. doi: 10.1016/j.neubiorev.2014.08.017. [http://dx.doi.org/10.1016/j.neubiorev.2014.08.017]. [PMID: 25218759]. [DOI] [PubMed] [Google Scholar]
  • 71.Boyce-Rustay J.M., Holmes A. Genetic inactivation of the NMDA receptor NR2A subunit has anxiolytic- and antidepressant-like effects in mice. Neuropsychopharmacology. 2006;31(11):2405–2414. doi: 10.1038/sj.npp.1301039. [http://dx.doi.org/10.1038/sj.npp.1301039]. [PMID: 16482087]. [DOI] [PubMed] [Google Scholar]
  • 72.Miller O.H., Yang L., Wang C.C., Hargroder E.A., Zhang Y., Delpire E., Hall B.J. GluN2B-containing NMDA receptors regulate depression-like behavior and are critical for the rapid antidepressant actions of ketamine. eLife. 2014;3:e03581. doi: 10.7554/eLife.03581. [http://dx.doi.org/10.7554/eLife.03581]. [PMID: 25340958]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Miyamoto Y., Yamada K., Noda Y., Mori H., Mishina M., Nabeshima T. Lower sensitivity to stress and altered monoaminergic neuronal function in mice lacking the NMDA receptor epsilon 4 subunit. J. Neurosci. 2002;22(6):2335–2342. doi: 10.1523/JNEUROSCI.22-06-02335.2002. [PMID: 11896172]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yamamoto H. 2017.
  • 75.Zarate C.A., Jr, Singh J.B., Carlson P.J., Brutsche N.E., Ameli R., Luckenbaugh D.A., Charney D.S., Manji H.K. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch. Gen. Psychiatry. 2006;63(8):856–864. doi: 10.1001/archpsyc.63.8.856. [http://dx.doi.org/10.1001/archpsyc.63.8.856]. [PMID: 16894061]. [DOI] [PubMed] [Google Scholar]
  • 76.Covvey J.R., Crawford A.N., Lowe D.K. Intravenous ketamine for treatment-resistant major depressive disorder. Ann. Pharmacother. 2012;46(1):117–123. doi: 10.1345/aph.1Q371. [http://dx.doi.org/10.1345/aph.1Q371]. [PMID: 22190250]. [DOI] [PubMed] [Google Scholar]
  • 77.Murrough J.W., Iosifescu D.V., Chang L.C., Al Jurdi R.K., Green C.E., Perez A.M., Iqbal S., Pillemer S., Foulkes A., Shah A., Charney D.S., Mathew S.J. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am. J. Psychiatry. 2013;170(10):1134–1142. doi: 10.1176/appi.ajp.2013.13030392. [http://dx.doi.org/10.1176/appi.ajp.2013.13030392]. [PMID: 23982301]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lapidus K.A., Levitch C.F., Perez A.M., Brallier J.W., Parides M.K., Soleimani L., Feder A., Iosifescu D.V., Charney D.S., Murrough J.W. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol. Psychiatry. 2014;76(12):970–976. doi: 10.1016/j.biopsych.2014.03.026. [http://dx.doi.org/10.1016/j.biopsych.2014.03.026]. [PMID: 24821196]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Berman R.M., Cappiello A., Anand A., Oren D.A., Heninger G.R., Charney D.S., Krystal J.H. Antidepressant effects of ketamine in depressed patients. Biol. Psychiatry. 2000;47(4):351–354. doi: 10.1016/s0006-3223(99)00230-9. [http://dx.doi.org/10.1016/S0006-3223(99)00230-9]. [PMID: 10686270]. [DOI] [PubMed] [Google Scholar]
  • 80.Irwin S.A., Iglewicz A. Oral ketamine for the rapid treatment of depression and anxiety in patients receiving hospice care. J. Palliat. Med. 2010;13(7):903–908. doi: 10.1089/jpm.2010.9808. [http://dx.doi.org/10.1089/jpm.2010. 9808]. [PMID: 20636166]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ray S.M., Kious B.M. Sustained resolution of panic disorder, agoraphobia, and generalized anxiety disorder with a single ketamine infusion: A case report. Prim. Care Companion CNS Disord. 2016;18(4) doi: 10.4088/PCC.15l01899. [PMID: 27828703]. [DOI] [PubMed] [Google Scholar]
  • 82.Feder A., Parides M.K., Murrough J.W., Perez A.M., Morgan J.E., Saxena S., Kirkwood K., Aan Het Rot M., Lapidus K.A., Wan L.B., Iosifescu D., Charney D.S. Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA Psychiatry. 2014;71(6):681–688. doi: 10.1001/jamapsychiatry.2014.62. [http://dx.doi.org/10.1001/jamapsychiatry.2014.62]. [PMID: 24740528]. [DOI] [PubMed] [Google Scholar]
  • 83.Rodriguez C.I., Kegeles L.S., Levinson A., Feng T., Marcus S.M., Vermes D., Flood P., Simpson H.B. Randomized controlled crossover trial of ketamine in obsessive-compulsive disorder: proof-of-concept. Neuropsychopharmacology. 2013;38(12):2475–2483. doi: 10.1038/npp.2013.150. [http://dx.doi.org/10.1038/npp.2013.150]. [PMID: 23783065]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Juven-Wetzler A., Cohen H., Kaplan Z., Kohen A., Porat O., Zohar J. Immediate ketamine treatment does not prevent posttraumatic stress responses in an animal model for PTSD. Eur. Neuropsychopharmacol. 2014;24(3):469–479. doi: 10.1016/j.euroneuro.2013.08.007. [http://dx.doi.org/10. 1016/j.euroneuro.2013.08.007]. [PMID: 24239430]. [DOI] [PubMed] [Google Scholar]
  • 85.Amat J., Dolzani S.D., Tilden S., Christianson J.P., Kubala K.H., Bartholomay K., Sperr K., Ciancio N., Watkins L.R., Maier S.F. Previous ketamine produces an enduring blockade of neurochemical and behavioral effects of uncontrollable stress. J. Neurosci. 2016;36(1):153–161. doi: 10.1523/JNEUROSCI.3114-15.2016. [http://dx.doi.org/10.1523/ JNEUROSCI.3114-15.2016]. [PMID: 26740657]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.McGowan J.C., LaGamma C.T., Lim S.C., Tsitsiklis M., Neria Y., Brachman R.A., Denny C.A. Prophylactic ketamine attenuates learned feaR. Neuropsychopharmacology. 2017;42(8):1577–1589. doi: 10.1038/npp.2017.19. [http://dx.doi.org/10.1038/npp.2017.19]. [PMID: 28128336]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Maeng S., Zarate C.A., Jr, Du J., Schloesser R.J., McCammon J., Chen G., Manji H.K. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol. Psychiatry. 2008;63(4):349–352. doi: 10.1016/j.biopsych.2007.05.028. [http://dx.doi.org/10.1016/j.biopsych.2007.05.028]. [PMID: 17643398]. [DOI] [PubMed] [Google Scholar]
  • 88.Gibbons A.S., Brooks L., Scarr E., Dean B. AMPA receptor expression is increased post-mortem samples of the anterior cingulate from subjects with major depressive disorder. J. Affect. Disord. 2012;136(3):1232–1237. doi: 10.1016/j.jad.2011.10.001. [http://dx.doi.org/10.1016/j.jad. 2011.10.001]. [PMID: 22036795]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Beneyto M., Kristiansen L.V., Oni-Orisan A., McCullumsmith R.E., Meador-Woodruff J.H. Abnormal glutamate receptor expression in the medial temporal lobe in schizophrenia and mood disorders. Neuropsychopharmacology. 2007;32(9):1888–1902. doi: 10.1038/sj.npp.1301312. [http://dx.doi.org/10.1038/sj.npp.1301312]. [PMID: 17299517]. [DOI] [PubMed] [Google Scholar]
  • 90.Freudenberg F., Celikel T., Reif A. The role of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in depression: central mediators of pathophysiology and antidepressant activity? Neurosci. Biobehav. Rev. 2015;52:193–206. doi: 10.1016/j.neubiorev.2015.03.005. [http:// dx.doi.org/10.1016/j.neubiorev.2015.03.005]. [PMID: 25783220]. [DOI] [PubMed] [Google Scholar]
  • 91.Fitzgerald P.J., Barkus C., Feyder M., Wiedholz L.M., Chen Y.C., Karlsson R.M., Machado-Vieira R., Graybeal C., Sharp T., Zarate C., Harvey-White J., Du J., Sprengel R., Gass P., Bannerman D., Holmes A. Does gene deletion of AMPA GluA1 phenocopy features of schizoaffective disorder? Neurobiol. Dis. 2010;40(3):608–621. doi: 10.1016/j.nbd.2010.08.005. [http://dx.doi.org/10.1016/j.nbd.2010.08.005]. [PMID: 20699120]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Maksimovic M., Vekovischeva O.Y., Aitta-aho T., Korpi E.R. Chronic treatment with mood-stabilizers attenuates abnormal hyperlocomotion of GluA1-subunit deficient mice. PLoS One. 2014;9(6):e100188. doi: 10.1371/journal.pone.0100188. [http://dx.doi.org/10.1371/journal.pone.0100188]. [PMID: 24932798]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chourbaji S., Vogt M.A., Fumagalli F., Sohr R., Frasca A., Brandwein C., Hörtnagl H., Riva M.A., Sprengel R., Gass P. AMPA receptor subunit 1 (GluR-A) knockout mice model the glutamate hypothesis of depression. FASEB J. 2008;22(9):3129–3134. doi: 10.1096/fj.08-106450. [http://dx.doi.org/10.1096/fj.08-106450]. [PMID: 18492725]. [DOI] [PubMed] [Google Scholar]
  • 94.Vogt M.A., Elkin H., Pfeiffer N., Sprengel R., Gass P., Inta D. Impact of adolescent GluA1 AMPA receptor ablation in forebrain excitatory neurons on behavioural correlates of mood disorders. Eur. Arch. Psychiatry Clin. Neurosci. 2014;264(7):625–629. doi: 10.1007/s00406-014-0509-5. [http://dx.doi.org/10.1007/s00406-014-0509-5]. [PMID: 24895223]. [DOI] [PubMed] [Google Scholar]
  • 95.Weber T., Vogt M.A., Gartside S.E., Berger S.M., Lujan R., Lau T., Herrmann E., Sprengel R., Bartsch D., Gass P. Adult AMPA GLUA1 receptor subunit loss in 5-HT neurons results in a specific anxiety-phenotype with evidence for dysregulation of 5-HT neuronal activity. Neuropsychopharmacology. 2015;40(6):1471–1484. doi: 10.1038/npp.2014.332. [http://dx.doi.org/10.1038/npp.2014.332]. [PMID: 25547714]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Vialou V., Robison A.J., Laplant Q.C., Covington H.E., III, Dietz D.M., Ohnishi Y.N., Mouzon E., Rush A.J., III, Watts E.L., Wallace D.L., Iñiguez S.D., Ohnishi Y.H., Steiner M.A., Warren B.L., Krishnan V., Bolaños C.A., Neve R.L., Ghose S., Berton O., Tamminga C.A., Nestler E.J. DeltaFosB in brain reward circuits mediates resilience to stress and antidepressant responses. Nat. Neurosci. 2010;13(6):745–752. doi: 10.1038/nn.2551. [http://dx.doi.org/ 10.1038/nn.2551]. [PMID: 20473292]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mead A.N., Morris H.V., Dixon C.I., Rulten S.L., Mayne L.V., Zamanillo D., Stephens D.N. AMPA receptor GluR2, but not GluR1, subunit deletion impairs emotional response conditioning in mice. Behav. Neurosci. 2006;120(2):241–248. doi: 10.1037/0735-7044.120.2.241. [http://dx.doi.org/ 10.1037/0735-7044.120.2.241]. [PMID: 16719688]. [DOI] [PubMed] [Google Scholar]
  • 98.Li X., Tizzano J.P., Griffey K., Clay M., Lindstrom T. [DOI] [PubMed]; Skolnick P. Antidepressant-like actions of an AMPA receptor potentiator (LY392098). Neuropharmacology. 2001;40(8):1028–1033. doi: 10.1016/s0028-3908(00)00194-5. [http://dx.doi.org/10.1016/S0028-3908(00)00194-5]. [PMID: 11406194]. [DOI] [PubMed] [Google Scholar]
  • 99.Schmidt M.V., Trümbach D., Weber P., Wagner K., Scharf S.H., Liebl C., Datson N., Namendorf C., Gerlach T., Kühne C., Uhr M., Deussing J.M., Wurst W., Binder E.B., Holsboer F., Müller M.B. Individual stress vulnerability is predicted by short-term memory and AMPA receptor subunit ratio in the hippocampus. J. Neurosci. 2010;30(50):16949–16958. doi: 10.1523/JNEUROSCI.4668-10.2010. [http://dx.doi.org/ 10.1523/JNEUROSCI.4668-10.2010]. [PMID: 21159965]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Knable M.B., Torrey E.F., Webster M.J., Bartko J.J. Multivariate analysis of prefrontal cortical data from the Stanley Foundation Neuropathology Consortium. Brain Res. Bull. 2001;55(5):651–659. doi: 10.1016/s0361-9230(01)00521-4. [http://dx.doi.org/10.1016/S0361-9230(01)00521-4]. [PMID: 11576762]. [DOI] [PubMed] [Google Scholar]
  • 101.Laje G., Paddock S., Manji H., Rush A.J., Wilson A.F., Charney D., McMahon F.J. Genetic markers of suicidal ideation emerging during citalopram treatment of major depression. Am. J. Psychiatry. 2007;164(10):1530–1538. doi: 10.1176/appi.ajp.2007.06122018. [http://dx.doi.org/ 10.1176/appi.ajp.2007.06122018]. [PMID: 17898344]. [DOI] [PubMed] [Google Scholar]
  • 102.Myung W., Song J., Lim S.W., Won H.H., Kim S., Lee Y., Kang H.S., Lee H., Kim J.W., Carroll B.J., Kim D.K. Genetic association study of individual symptoms in depression. Psychiatry Res. 2012;198(3):400–406. doi: 10.1016/j.psychres.2011.12.037. [http://dx.doi.org/10.1016/j.psychres. 2011.12.037]. [PMID: 22429480]. [DOI] [PubMed] [Google Scholar]
  • 103.Schiffer H.H., Heinemann S.F. Association of the human kainate receptor GluR7 gene (GRIK3) with recurrent major depressive disorder. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2007;144B(1):20–26. doi: 10.1002/ajmg.b.30374. [http://dx.doi.org/10.1002/ajmg.b.30374]. [PMID: 16958029]. [DOI] [PubMed] [Google Scholar]
  • 104.Pickard B.S., Malloy M.P., Christoforou A., Thomson P.A., Evans K.L., Morris S.W., Hampson M., Porteous D.J., Blackwood D.H., Muir W.J. Cytogenetic and genetic evidence supports a role for the kainate-type glutamate receptor gene, GRIK4, in schizophrenia and bipolar disorder. Mol. Psychiatry. 2006;11(9):847–857. doi: 10.1038/sj.mp.4001867. [http://dx.doi.org/10.1038/sj.mp.4001867]. [PMID: 16819533]. [DOI] [PubMed] [Google Scholar]
  • 105.Shaltiel G., Maeng S., Malkesman O., Pearson B., Schloesser R.J., Tragon T., Rogawski M., Gasior M., Luckenbaugh D., Chen G., Manji H.K. Evidence for the involvement of the kainate receptor subunit GluR6 (GRIK2) in mediating behavioral displays related to behavioral symptoms of mania. Mol. Psychiatry. 2008;13(9):858–872. doi: 10.1038/mp.2008.20. [http://dx.doi.org/10.1038/mp.2008.20]. [PMID: 18332879]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Catches J.S., Xu J., Contractor A. Genetic ablation of the GluK4 kainate receptor subunit causes anxiolytic and antidepressant-like behavior in mice. Behav. Brain Res. 2012;228(2):406–414. doi: 10.1016/j.bbr.2011.12.026. [http://dx.doi.org/10.1016/j.bbr.2011.12.026]. [PMID: 22203159]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Terbeck S., Akkus F., Chesterman L.P., Hasler G. The role of metabotropic glutamate receptor 5 in the pathogenesis of mood disorders and addiction: combining preclinical evidence with human Positron Emission Tomography (PET) studies. Front. Neurosci. 2015;9:86. doi: 10.3389/fnins.2015.00086. [http://dx.doi.org/10.3389/fnins.2015.00086]. [PMID: 25852460]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kovačević T., Skelin I., Minuzzi L., Rosa-Neto P., Diksic M. Reduced metabotropic glutamate receptor 5 in the flinders sensitive line of rats, an animal model of depression: an autoradiographic study. Brain Res. Bull. 2012;87(4-5):406–412. doi: 10.1016/j.brainresbull.2012.01.010. [http://dx. doi.org/10.1016/j.brainresbull.2012.01.010]. [PMID: 22310150]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Iyo A.H., Feyissa A.M., Chandran A., Austin M.C., Regunathan S., Karolewicz B. Chronic corticosterone administration down-regulates metabotropic glutamate receptor 5 protein expression in the rat hippocampus. Neuroscience. 2010;169(4):1567–1574. doi: 10.1016/j.neuroscience.2010.06.023. [http://dx.doi.org/10.1016/j.neuroscience.2010.06.023]. [PMID: 20600666]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Van den Hove D.L., Kenis G., Brass A., Opstelten R., Rutten B.P., Bruschettini M., Blanco C.E., Lesch K.P., Steinbusch H.W., Prickaerts J. Vulnerability versus resilience to prenatal stress in male and female rats; implications from gene expression profiles in the hippocampus and frontal cortex. Eur. Neuropsychopharmacol. 2013;23(10):1226–1246. doi: 10.1016/j.euroneuro.2012.09.011. [http://dx.doi.org/10.1016/ j.euroneuro.2012.09.011]. [PMID: 23199416]. [DOI] [PubMed] [Google Scholar]
  • 111.Shin S., Kwon O., Kang J.I., Kwon S., Oh S., Choi J., Kim C.H., Kim D.G. mGluR5 in the nucleus accumbens is critical for promoting resilience to chronic stress. Nat. Neurosci. 2015;18(7):1017–1024. doi: 10.1038/nn.4028. [http://dx.doi.org/10.1038/nn.4028]. [PMID: 26005851]. [DOI] [PubMed] [Google Scholar]
  • 112.Li X., Need A.B., Baez M., Witkin J.M. Metabotropic glutamate 5 receptor antagonism is associated with antidepressant-like effects in mice. J. Pharmacol. Exp. Ther. 2006;319(1):254–259. doi: 10.1124/jpet.106.103143. [http:// dx.doi.org/10.1124/jpet.106.103143]. [PMID: 16803860]. [DOI] [PubMed] [Google Scholar]
  • 113.Feyissa A.M., Woolverton W.L., Miguel-Hidalgo J.J., Wang Z., Kyle P.B., Hasler G., Stockmeier C.A., Iyo A.H., Karolewicz B. Elevated level of metabotropic glutamate receptor 2/3 in the prefrontal cortex in major depression. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2010;34(2):279–283. doi: 10.1016/j.pnpbp.2009.11.018. [http://dx.doi.org/10.1016/ j.pnpbp.2009.11.018]. [PMID: 19945495]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Muguruza C., Miranda-Azpiazu P., Díez-Alarcia R., Morentin B., González-Maeso J., Callado L.F., Meana J.J. Evaluation of 5-HT2A and mGlu2/3 receptors in postmortem prefrontal cortex of subjects with major depressive disorder: effect of antidepressant treatment. Neuropharmacology. 2014;86:311–318. doi: 10.1016/j.neuropharm.2014.08.009. [http://dx. doi.org/10.1016/j.neuropharm.2014.08.009]. [PMID: 25150943]. [DOI] [PubMed] [Google Scholar]
  • 115.Du Jardin K.G., Müller H.K., Sanchez C., Wegener G., Elfving B. Gene expression related to serotonergic and glutamatergic neurotransmission is altered in the flinders sensitive line rat model of depression: Effect of ketamine. Synapse. 2017;71(1):37–45. doi: 10.1002/syn.21940. [http://dx.doi.org/10.1002/syn.21940]. [PMID: 27589698]. [DOI] [PubMed] [Google Scholar]
  • 116.Matrisciano F., Caruso A., Orlando R., Marchiafava M., Bruno V., Battaglia G., Gruber S.H., Melchiorri D., Tatarelli R., Girardi P., Mathè A.A., Nicoletti F. Defective group-II metaboropic glutamate receptors in the hippocampus of spontaneously depressed rats. Neuropharmacology. 2008;55(4):525–531. doi: 10.1016/j.neuropharm.2008.05.014. [http:// dx.doi.org/10.1016/j.neuropharm.2008.05.014]. [PMID: 18590921]. [DOI] [PubMed] [Google Scholar]
  • 117.Nasca C., Bigio B., Zelli D., Nicoletti F., McEwen B.S. Mind the gap: glucocorticoids modulate hippocampal glutamate tone underlying individual differences in stress susceptibility. Mol. Psychiatry. 2015;20(6):755–763. doi: 10.1038/mp.2014.96. [http://dx.doi.org/10.1038/mp.2014. 96]. [PMID: 25178162]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.De Filippis B., Lyon L., Taylor A., Lane T., Burnet P.W., Harrison P.J., Bannerman D.M. The role of group II metabotropic glutamate receptors in cognition and anxiety: comparative studies in GRM2(-/-), GRM3(-/-) and GRM2/3(-/-) knockout mice. Neuropharmacology. 2015;89:19–32. doi: 10.1016/j.neuropharm.2014.08.010. [http://dx.doi.org/10.1016/ j.neuropharm.2014.08.010]. [PMID: 25158312]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Cryan J.F., Kelly P.H., Neijt H.C., Sansig G., Flor P.J., van Der Putten H. Antidepressant and anxiolytic-like effects in mice lacking the group III metabotropic glutamate receptor mGluR7. Eur. J. Neurosci. 2003;17(11):2409–2417. doi: 10.1046/j.1460-9568.2003.02667.x. [http://dx.doi.org/10.1046/ j.1460-9568.2003.02667.x]. [PMID: 12814372]. [DOI] [PubMed] [Google Scholar]
  • 120.Linden A.M., Johnson B.G., Peters S.C., Shannon H.E., Tian M., Wang Y., Yu J.L., Köster A., Baez M., Schoepp D.D. Increased anxiety-related behavior in mice deficient for metabotropic glutamate 8 (mGlu8) receptor. Neuropharmacology. 2002;43(2):251–259. doi: 10.1016/s0028-3908(02)00079-5. [http://dx.doi.org/10.1016/S0028-3908(02)00079-5]. [PMID: 12213279]. [DOI] [PubMed] [Google Scholar]
  • 121.Nutt D.J., Malizia A.L. New insights into the role of the GABA(A)-benzodiazepine receptor in psychiatric disorder. Br. J. Psychiatry. 2001;179:390–396. doi: 10.1192/bjp.179.5.390. [http://dx.doi.org/10.1192/bjp.179. 5.390]. [PMID: 11689393]. [DOI] [PubMed] [Google Scholar]
  • 122.Schür R.R., Draisma L.W., Wijnen J.P., Boks M.P., Koevoets M.G., Joëls M., Klomp D.W., Kahn R.S., Vinkers C.H. Brain GABA levels across psychiatric disorders: A systematic literature review and meta-analysis of (1) H-MRS studies. Hum. Brain Mapp. 2016;37(9):3337–3352. doi: 10.1002/hbm.23244. [http://dx.doi.org/10.1002/hbm. 23244]. [PMID: 27145016]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Vaiva G., Boss V., Ducrocq F., Fontaine M., Devos P., Brunet A., Laffargue P., Goudemand M., Thomas P. Relationship between posttrauma GABA plasma levels and PTSD at 1-year follow-up. Am. J. Psychiatry. 2006;163(8):1446–1448. doi: 10.1176/ajp.2006.163.8.1446. [http://dx.doi. org/10.1176/ajp.2006.163.8.1446]. [PMID: 16877663]. [DOI] [PubMed] [Google Scholar]
  • 124.Varga Z., Csabai D., Miseta A., Wiborg O., Czéh B. Chronic stress affects the number of GABAergic neurons in the orbitofrontal cortex of rats. Behav. Brain Res. 2017;316:104–114. doi: 10.1016/j.bbr.2016.08.030. [http://dx.doi.org/10.1016/j.bbr.2016.08.030]. [PMID: 27555539]. [DOI] [PubMed] [Google Scholar]
  • 125.Birkenhäger T.K., Moleman P., Nolen W.A. Benzodiazepines for depression? A review of the literature. Int. Clin. Psychopharmacol. 1995;10(3):181–195. doi: 10.1097/00004850-199510030-00008. [http://dx.doi.org/10.1097/00004850-199510030-00008]. [PMID: 8675972]. [DOI] [PubMed] [Google Scholar]
  • 126.Pham X., Sun C., Chen X., van den Oord E.J., Neale M.C., Kendler K.S., Hettema J.M. Association study between GABA receptor genes and anxiety spectrum disorders. Depress. Anxiety. 2009;26(11):998–1003. doi: 10.1002/da.20628. [http://dx.doi.org/10.1002/da.20628]. [PMID: 19842164]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Feusner J., Ritchie T., Lawford B., Young R.M., Kann B., Noble E.P. GABA(A) receptor beta 3 subunit gene and psychiatric morbidity in a post-traumatic stress disorder population. Psychiatry Res. 2001;104(2):109–117. doi: 10.1016/s0165-1781(01)00296-7. [http://dx.doi.org/10.1016/S0165-1781(01)00296-7]. [PMID: 11711165]. [DOI] [PubMed] [Google Scholar]
  • 128.Gafford G.M., Guo J.D., Flandreau E.I., Hazra R., Rainnie D.G., Ressler K.J. Cell-type specific deletion of GABA(A)α1 in corticotropin-releasing factor-containing neurons enhances anxiety and disrupts fear extinction. Proc. Natl. Acad. Sci. USA. 2012;109(40):16330–16335. doi: 10.1073/pnas.1119261109. [http://dx.doi.org/10.1073/pnas.1119261109]. [PMID: 22992651]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Vollenweider I., Smith K.S., Keist R., Rudolph U. Antidepressant-like properties of α2-containing GABA(A) receptors. Behav. Brain Res. 2011;217(1):77–80. doi: 10.1016/j.bbr.2010.10.009. [http://dx.doi.org/10.1016/j.bbr. 2010.10.009]. [PMID: 20965216]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Fiorelli R., Rudolph U., Straub C.J., Feldon J., Yee B.K. Affective and cognitive effects of global deletion of alpha3-containing gamma-aminobutyric acid-A receptors. Behav. Pharmacol. 2008;19(5-6):582–596. doi: 10.1097/FBP.0b013e32830dc0c7. [http://dx.doi.org/10.1097/FBP.0b013e32830dc0c7]. [PMID: 18690113]. [DOI] [PubMed] [Google Scholar]
  • 131.Engin E., Smith K.S., Gao Y., Nagy D., Foster R.A., Tsvetkov E., Keist R., Crestani F., Fritschy J.M., Bolshakov V.Y., Hajos M., Heldt S.A., Rudolph U. Modulation of anxiety and fear via distinct intrahippocampal circuits. eLife. 2016;5:e14120. doi: 10.7554/eLife.14120. [http://dx.doi.org/10.7554/eLife.14120]. [PMID: 26971710]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Yee B.K., Keist R., von Boehmer L., Studer R., Benke D., Hagenbuch N., Dong Y., Malenka R.C., Fritschy J.M., Bluethmann H., Feldon J., Möhler H., Rudolph U. A schizophrenia-related sensorimotor deficit links alpha 3-containing GABAA receptors to a dopamine hyperfunction. Proc. Natl. Acad. Sci. USA. 2005;102(47):17154–17159. doi: 10.1073/pnas.0508752102. [http://dx.doi.org/10.1073/pnas. 0508752102]. [PMID: 16284244]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chandra D., Korpi E.R., Miralles C.P., De Blas A.L., Homanics G.E. GABAA receptor gamma 2 subunit knockdown mice have enhanced anxiety-like behavior but unaltered hypnotic response to benzodiazepines. BMC Neurosci. 2005;6:30. doi: 10.1186/1471-2202-6-30. [http://dx.doi.org/ 10.1186/1471-2202-6-30]. [PMID: 15850489]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Crestani F., Lorez M., Baer K., Essrich C., Benke D., Laurent J.P., Belzung C., Fritschy J.M., Lüscher B., Mohler H. Decreased GABAA-receptor clustering results in enhanced anxiety and a bias for threat cues. Nat. Neurosci. 1999;2(9):833–839. doi: 10.1038/12207. [http://dx.doi.org/10.1038/12207]. [PMID: 10461223]. [DOI] [PubMed] [Google Scholar]
  • 135.Levinson A.J., Fitzgerald P.B., Favalli G., Blumberger D.M., Daigle M., Daskalakis Z.J. Evidence of cortical inhibitory deficits in major depressive disorder. Biol. Psychiatry. 2010;67(5):458–464. doi: 10.1016/j.biopsych.2009.09.025. [http://dx.doi.org/10.1016/j.biopsych.2009.09.025]. [PMID: 19922906]. [DOI] [PubMed] [Google Scholar]
  • 136.O’Leary O.F., Felice D., Galimberti S., Savignac H.M., Bravo J.A., Crowley T., El Yacoubi M., Vaugeois J.M., Gassmann M., Bettler B., Dinan T.G., Cryan J.F. GABAB(1) receptor subunit isoforms differentially regulate stress resilience. Proc. Natl. Acad. Sci. USA. 2014;111(42):15232–15237. doi: 10.1073/pnas.1404090111. [http://dx.doi.org/10.1073/ pnas.1404090111]. [PMID: 25288769]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Thoeringer C.K., Ripke S., Unschuld P.G., Lucae S., Ising M., Bettecken T., Uhr M., Keck M.E., Mueller-Myhsok B., Holsboer F., Binder E.B., Erhardt A. The GABA transporter 1 (SLC6A1): a novel candidate gene for anxiety disorders. J. Neural Transm. (Vienna) 2009;116(6):649–657. doi: 10.1007/s00702-008-0075-y. [http://dx.doi.org/10. 1007/s00702-008-0075-y]. [PMID: 18607529]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Schwartz T.L., Nihalani N. Tiagabine in anxiety disorders. Expert Opin. Pharmacother. 2006;7(14):1977–1987. doi: 10.1517/14656566.7.14.1977. [http://dx.doi.org/ 10.1517/14656566.7.14.1977]. [PMID: 17020423]. [DOI] [PubMed] [Google Scholar]
  • 139.Liu G.X., Cai G.Q., Cai Y.Q., Sheng Z.J., Jiang J., Mei Z., Wang Z.G., Guo L., Fei J. Reduced anxiety and depression-like behaviors in mice lacking GABA transporter subtype 1. Neuropsychopharmacology. 2007;32(7):1531–1539. doi: 10.1038/sj.npp.1301281. [http://dx.doi.org/10. 1038/sj.npp.1301281]. [PMID: 17164814]. [DOI] [PubMed] [Google Scholar]
  • 140.Gong X., Shao Y., Li B., Chen L., Wang C., Chen Y. γ-aminobutyric acid transporter-1 is involved in anxiety-like behaviors and cognitive function in knockout mice. Exp. Ther. Med. 2015;10(2):653–658. doi: 10.3892/etm.2015.2577. [http://dx.doi.org/10.3892/etm.2015.2577]. [PMID: 26622370]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Karolewicz B., Maciag D., O’Dwyer G., Stockmeier C.A., Feyissa A.M., Rajkowska G. Reduced level of glutamic acid decarboxylase-67 kDa in the prefrontal cortex in major depression. Int. J. Neuropsychopharmacol. 2010;13(4):411–420. doi: 10.1017/S1461145709990587. [http://dx.doi. org/10.1017/S1461145709990587]. [PMID: 20236554]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Gao S.F., Klomp A., Wu J.L., Swaab D.F., Bao A.M. Reduced GAD(65/67) immunoreactivity in the hypothalamic paraventricular nucleus in depression: a postmortem study. J. Affect. Disord. 2013;149(1-3):422–425. doi: 10.1016/j.jad.2012.12.003. [http://dx.doi.org/10.1016/j.jad.2012.12.003]. [PMID: 23312397]. [DOI] [PubMed] [Google Scholar]
  • 143.Gilabert-Juan J., Castillo-Gomez E., Guirado R., Moltó M.D., Nacher J. Chronic stress alters inhibitory networks in the medial prefrontal cortex of adult mice. Brain Struct. Funct. 2013;218(6):1591–1605. doi: 10.1007/s00429-012-0479-1. [http://dx.doi.org/10.1007/s00429-012-0479-1]. [PMID: 23179864]. [DOI] [PubMed] [Google Scholar]
  • 144.Pochwat B., Nowak G., Szewczyk B. Brain glutamic acid decarboxylase-67kDa alterations induced by magnesium treatment in olfactory bulbectomy and chronic mild stress models in rats. Pharmacol. Rep. 2016;68(5):881–885. doi: 10.1016/j.pharep.2016.04.011. [http://dx.doi.org/10.1016/j. pharep.2016.04.011]. [PMID: 27351943]. [DOI] [PubMed] [Google Scholar]
  • 145.Stork O., Ji F.Y., Kaneko K., Stork S., Yoshinobu Y., Moriya T., Shibata S., Obata K. Postnatal development of a GABA deficit and disturbance of neural functions in mice lacking GAD65. Brain Res. 2000;865(1):45–58. doi: 10.1016/s0006-8993(00)02206-x. [http://dx.doi.org/10.1016/S0006-8993(00)02206-X]. [PMID: 10814732]. [DOI] [PubMed] [Google Scholar]
  • 146.Kash S.F., Tecott L.H., Hodge C., Baekkeskov S. Increased anxiety and altered responses to anxiolytics in mice deficient in the 65-kDa isoform of glutamic acid decarboxylase. Proc. Natl. Acad. Sci. USA. 1999;96(4):1698–1703. doi: 10.1073/pnas.96.4.1698. [http://dx.doi.org/10.1073/pnas. 96.4.1698]. [PMID: 9990087]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Sangha S., Narayanan R.T., Bergado-Acosta J.R., Stork O., Seidenbecher T., Pape H.C. Deficiency of the 65 kDa isoform of glutamic acid decarboxylase impairs extinction of cued but not contextual fear memory. J. Neurosci. 2009;29(50):15713–15720. doi: 10.1523/JNEUROSCI.2620-09.2009. [http://dx.doi.org/10.1523/JNEUROSCI.2620-09.2009]. [PMID: 20016086]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Müller I., Obata K., Richter-Levin G., Stork O. GAD65 haplodeficiency conveys resilience in animal models of stress-induced psychopathology. Front. Behav. Neurosci. 2014;8:265. doi: 10.3389/fnbeh.2014.00265. [PMID: 25147515]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Shaheen A.A., Hamdy M.A., Kheir-Eldin A.A., Lindström P., el-Fattah A.A. Effect of pretreatment with vitamin E or diazepam on brain metabolism of stressed rats. Biochem. Pharmacol. 1993;46(1):194–197. doi: 10.1016/0006-2952(93)90367-6. [http://dx.doi.org/10.1016/0006-2952(93)90367-6]. [PMID: 8394075]. [DOI] [PubMed] [Google Scholar]
  • 150.Maldonado N.M., Martijena I.D., Molina V.A. Facilitating influence of stress on the consolidation of fear memory induced by a weak training: reversal by midazolam pretreatment. Behav. Brain Res. 2011;225(1):77–84. doi: 10.1016/j.bbr.2011.06.035. [http://dx.doi.org/10.1016/j.bbr.2011.06. 035]. [PMID: 21763355]. [DOI] [PubMed] [Google Scholar]
  • 151.Skórzewska A., Lehner M., Wisłowska-Stanek A., Turzyńska D., Sobolewska A., Krząścik P., Płaźnik A. Midazolam treatment before re-exposure to contextual fear reduces freezing behavior and amygdala activity differentially in high- and low-anxiety rats. Pharmacol. Biochem. Behav. 2015;129:34–44. doi: 10.1016/j.pbb.2014.11.020. [http://dx. doi.org/10.1016/j.pbb.2014.11.020]. [PMID: 25482326]. [DOI] [PubMed] [Google Scholar]
  • 152.Holsboer F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology. 2000;23(5):477–501. doi: 10.1016/S0893-133X(00)00159-7. [http://dx. doi.org/10.1016/S0893-133X(00)00159-7]. [PMID: 11027914]. [DOI] [PubMed] [Google Scholar]
  • 153.Pariante C.M., Lightman S.L. The HPA axis in major depression: classical theories and new developments. Trends Neurosci. 2008;31(9):464–468. doi: 10.1016/j.tins.2008.06.006. [http://dx.doi.org/10.1016/j.tins.2008.06.006]. [PMID: 18675469]. [DOI] [PubMed] [Google Scholar]
  • 154.David D.J., Samuels B.A., Rainer Q., Wang J.W., Marsteller D., Mendez I., Drew M., Craig D.A., Guiard B.P., Guilloux J.P., Artymyshyn R.P., Gardier A.M., Gerald C., Antonijevic I.A., Leonardo E.D., Hen R. Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron. 2009;62(4):479–493. doi: 10.1016/j.neuron.2009.04.017. [http://dx.doi.org/10.1016/j.neuron. 2009.04.017]. [PMID: 19477151]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Stetler C., Miller G.E. Depression and hypothalamic-pituitary-adrenal activation: a quantitative summary of four decades of research. Psychosom. Med. 2011;73(2):114–126. doi: 10.1097/PSY.0b013e31820ad12b. [http://dx. doi.org/10.1097/PSY.0b013e31820ad12b]. [PMID: 21257974]. [DOI] [PubMed] [Google Scholar]
  • 156.Elliott E., Ezra-Nevo G., Regev L., Neufeld-Cohen A., Chen A. Resilience to social stress coincides with functional DNA methylation of the Crf gene in adult mice. Nat. Neurosci. 2010;13(11):1351–1353. doi: 10.1038/nn.2642. [http://dx.doi.org/10.1038/nn.2642]. [PMID: 20890295]. [DOI] [PubMed] [Google Scholar]
  • 157.Smith G.W., Aubry J.M., Dellu F., Contarino A., Bilezikjian L.M., Gold L.H., Chen R., Marchuk Y., Hauser C., Bentley C.A., Sawchenko P.E., Koob G.F., Vale W., Lee K.F. Corticotropin releasing factor receptor 1-deficient mice display decreased anxiety, impaired stress response, and aberrant neuroendocrine development. Neuron. 1998;20(6):1093–1102. doi: 10.1016/s0896-6273(00)80491-2. [http://dx.doi.org/10. 1016/S0896-6273(00)80491-2]. [PMID: 9655498]. [DOI] [PubMed] [Google Scholar]
  • 158.Müller M.B., Zimmermann S., Sillaber I., Hagemeyer T.P., Deussing J.M., Timpl P., Kormann M.S., Droste S.K., Kühn R., Reul J.M., Holsboer F., Wurst W. Limbic corticotropin-releasing hormone receptor 1 mediates anxiety-related behavior and hormonal adaptation to stress. Nat. Neurosci. 2003;6(10):1100–1107. doi: 10.1038/nn1123. [http://dx.doi.org/10.1038/nn1123]. [PMID: 12973355]. [DOI] [PubMed] [Google Scholar]
  • 159.Taneja M., Salim S., Saha K., Happe H.K., Qutna N., Petty F., Bylund D.B., Eikenburg D.C. Differential effects of inescapable stress on locus coeruleus GRK3, alpha2-adrenoceptor and CRF1 receptor levels in learned helpless and non-helpless rats: a potential link to stress resilience. Behav. Brain Res. 2011;221(1):25–33. doi: 10.1016/j.bbr.2011.02.018. [http://dx.doi.org/10.1016/j.bbr.2011.02.018]. [PMID: 21333691]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Bradley R.G., Binder E.B., Epstein M.P., Tang Y., Nair H.P., Liu W., Gillespie C.F., Berg T., Evces M., Newport D.J., Stowe Z.N., Heim C.M., Nemeroff C.B., Schwartz A., Cubells J.F., Ressler K.J. Influence of child abuse on adult depression: moderation by the corticotropin-releasing hormone receptor gene. Arch. Gen. Psychiatry. 2008;65(2):190–200. doi: 10.1001/archgenpsychiatry.2007.26. [http://dx.doi.org/ 10.1001/archgenpsychiatry.2007.26]. [PMID: 18250257]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Jochems J., Teegarden S.L., Chen Y., Boulden J., Challis C., Ben-Dor G.A., Kim S.F., Berton O. Enhancement of stress resilience through histone deacetylase 6-mediated regulation of glucocorticoid receptor chaperone dynamics. Biol. Psychiatry. 2015;77(4):345–355. doi: 10.1016/j.biopsych.2014.07.036. [http://dx.doi.org/10.1016/j.biopsych.2014.07.036]. [PMID: 25442004]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Meewisse M.L., Reitsma J.B., de Vries G.J., Gersons B.P., Olff M. Cortisol and post-traumatic stress disorder in adults: systematic review and meta-analysis. Br. J. Psychiatry. 2007;191:387–392. doi: 10.1192/bjp.bp.106.024877. [http://dx.doi.org/10.1192/bjp.bp.106.024877]. [PMID: 17978317]. [DOI] [PubMed] [Google Scholar]
  • 163.Klengel T., Binder E.B. Gene-environment interactions in major depressive disorder. Can. J. Psychiatry. 2013;58(2):76–83. doi: 10.1177/070674371305800203. [http:// dx.doi.org/10.1177/070674371305800203]. [PMID: 23442893]. [DOI] [PubMed] [Google Scholar]
  • 164.Juruena M.F., Cleare A.J., Papadopoulos A.S., Poon L., Lightman S., Pariante C.M. Different responses to dexamethasone and prednisolone in the same depressed patients. Psychopharmacology (Berl.) 2006;189(2):225–235. doi: 10.1007/s00213-006-0555-4. [http://dx.doi.org/10.1007/s00213-006-0555-4]. [PMID: 17016711]. [DOI] [PubMed] [Google Scholar]
  • 165.Guidotti G., Calabrese F., Anacker C., Racagni G., Pariante C.M., Riva M.A. Glucocorticoid receptor and FKBP5 expression is altered following exposure to chronic stress: modulation by antidepressant treatment. Neuropsychopharmacology. 2013;38(4):616–627. doi: 10.1038/npp.2012.225. [http://dx.doi.org/10.1038/npp.2012.225]. [PMID: 23169346]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.George S.A., Rodriguez-Santiago M., Riley J., Rodriguez E., Liberzon I. The effect of chronic phenytoin administration on single prolonged stress induced extinction retention deficits and glucocorticoid upregulation in the rat medial prefrontal cortex. Psychopharmacology (Berl.) 2015;232(1):47–56. doi: 10.1007/s00213-014-3635-x. [http://dx.doi. org/10.1007/s00213-014-3635-x]. [PMID: 24879497]. [DOI] [PubMed] [Google Scholar]
  • 167.Boyle M.P., Brewer J.A., Funatsu M., Wozniak D.F., Tsien J.Z., Izumi Y., Muglia L.J. Acquired deficit of forebrain glucocorticoid receptor produces depression-like changes in adrenal axis regulation and behavior. Proc. Natl. Acad. Sci. USA. 2005;102(2):473–478. doi: 10.1073/pnas.0406458102. [http://dx.doi.org/10.1073/pnas.0406458102]. [PMID: 15623560]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ridder S., Chourbaji S., Hellweg R., Urani A., Zacher C., Schmid W., Zink M., Hörtnagl H., Flor H., Henn F.A., Schütz G., Gass P. Mice with genetically altered glucocorticoid receptor expression show altered sensitivity for stress-induced depressive reactions. J. Neurosci. 2005;25(26):6243–6250. doi: 10.1523/JNEUROSCI.0736-05.2005. [http://dx.doi.org/ 10.1523/JNEUROSCI.0736-05.2005]. [PMID: 15987954]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Hartmann J., Wagner K.V., Liebl C., Scharf S.H., Wang X.D., Wolf M., Hausch F., Rein T., Schmidt U., Touma C., Cheung-Flynn J., Cox M.B., Smith D.F., Holsboer F., Müller M.B., Schmidt M.V. The involvement of FK506-binding protein 51 (FKBP5) in the behavioral and neuroendocrine effects of chronic social defeat stress. Neuropharmacology. 2012;62(1):332–339. doi: 10.1016/j.neuropharm.2011.07.041. [http://dx.doi.org/10.1016/j.neuropharm.2011.07.041]. [PMID: 21839098]. [DOI] [PubMed] [Google Scholar]
  • 170.O’Leary J.C., III, Dharia S., Blair L.J., Brady S., Johnson A.G., Peters M., Cheung-Flynn J., Cox M.B., de Erausquin G., Weeber E.J., Jinwal U.K., Dickey C.A. A new anti-depressive strategy for the elderly: ablation of FKBP5/FKBP51. PLoS One. 2011;6(9):e24840. doi: 10.1371/journal.pone.0024840. [http://dx.doi.org/10.1371/journal.pone.0024840]. [PMID: 21935478]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Murphy P.J., Morishima Y., Kovacs J.J., Yao T.P., Pratt W.B. Regulation of the dynamics of hsp90 action on the glucocorticoid receptor by acetylation/deacetylation of the chaperone. J. Biol. Chem. 2005;280(40):33792–33799. doi: 10.1074/jbc.M506997200. [http://dx.doi.org/10.1074/ jbc.M506997200]. [PMID: 16087666]. [DOI] [PubMed] [Google Scholar]
  • 172.Fukada M., Hanai A., Nakayama A., Suzuki T., Miyata N., Rodriguiz R.M., Wetsel W.C., Yao T.P., Kawaguchi Y. Loss of deacetylation activity of Hdac6 affects emotional behavior in mice. PLoS One. 2012;7(2):e30924. doi: 10.1371/journal.pone.0030924. [http://dx.doi.org/10.1371/journal. pone.0030924]. [PMID: 22328923]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Espallergues J., Teegarden S.L., Veerakumar A., Boulden J., Challis C., Jochems J., Chan M., Petersen T., Deneris E., Matthias P., Hahn C.G., Lucki I., Beck S.G., Berton O. HDAC6 regulates glucocorticoid receptor signaling in serotonin pathways with critical impact on stress resilience. J. Neurosci. 2012;32(13):4400–4416. doi: 10.1523/JNEUROSCI.5634-11.2012. [http://dx.doi.org/10.1523/JNEUROSCI.5634-11.2012]. [PMID: 22457490]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.ter Heegde F., De Rijk R.H., Vinkers C.H. The brain mineralocorticoid receptor and stress resilience. Psychoneuroendocrinology. 2015;52:92–110. doi: 10.1016/j.psyneuen.2014.10.022. [http://dx.doi.org/10.1016/j.psyneuen.2014.10. 022]. [PMID: 25459896]. [DOI] [PubMed] [Google Scholar]
  • 175.Klok M.D., Giltay E.J., Van der Does A.J., Geleijnse J.M., Antypa N., Penninx B.W., de Geus E.J., Willemsen G., Boomsma D.I., van Leeuwen N., Zitman F.G., de Kloet E.R., DeRijk R.H. A common and functional mineralocorticoid receptor haplotype enhances optimism and protects against depression in females. Transl. Psychiatry. 2011;1:e62. doi: 10.1038/tp.2011.59. [http://dx.doi.org/10.1038/tp. 2011.59]. [PMID: 22832354]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Lai M., Horsburgh K., Bae S.E., Carter R.N., Stenvers D.J., Fowler J.H., Yau J.L., Gomez-Sanchez C.E., Holmes M.C., Kenyon C.J., Seckl J.R., Macleod M.R. Forebrain mineralocorticoid receptor overexpression enhances memory, reduces anxiety and attenuates neuronal loss in cerebral ischaemia. Eur. J. Neurosci. 2007;25(6):1832–1842. doi: 10.1111/j.1460-9568.2007.05427.x. [http://dx.doi.org/10.1111/j.1460-9568.2007.05427.x]. [PMID: 17432969]. [DOI] [PubMed] [Google Scholar]
  • 177.Mitra R., Ferguson D., Sapolsky R.M. Mineralocorticoid receptor overexpression in basolateral amygdala reduces corticosterone secretion and anxiety. Biol. Psychiatry. 2009;66(7):686–690. doi: 10.1016/j.biopsych.2009.04.016. [http:// dx.doi.org/10.1016/j.biopsych.2009.04.016]. [PMID: 19500777]. [DOI] [PubMed] [Google Scholar]
  • 178.de Kloet E.R., Otte C., Kumsta R., Kok L., Hillegers M.H., Hasselmann H., Kliegel D., Joëls M. Stress and depression: a crucial role of the mineralocorticoid receptor. J. Neuroendocrinol. 2016;28(8) doi: 10.1111/jne.12379. [http://dx.doi.org/10.1111/jne.12379]. [PMID: 26970338]. [DOI] [PubMed] [Google Scholar]
  • 179.Dunlop B.W., Nemeroff C.B. The role of dopamine in the pathophysiology of depression. Arch. Gen. Psychiatry. 2007;64(3):327–337. doi: 10.1001/archpsyc.64.3.327. [http://dx.doi.org/10.1001/archpsyc.64.3.327]. [PMID: 17339521]. [DOI] [PubMed] [Google Scholar]
  • 180.Krishnan V., Han M.H., Graham D.L., Berton O., Renthal W., Russo S.J., Laplant Q., Graham A., Lutter M., Lagace D.C., Ghose S., Reister R., Tannous P., Green T.A., Neve R.L., Chakravarty S., Kumar A., Eisch A.J., Self D.W., Lee F.S., Tamminga C.A., Cooper D.C., Gershenfeld H.K., Nestler E.J. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131(2):391–404. doi: 10.1016/j.cell.2007.09.018. [http://dx.doi.org/10.1016/j.cell.2007.09.018]. [PMID: 17956738]. [DOI] [PubMed] [Google Scholar]
  • 181.Cao J.L., Covington H.E., III, Friedman A.K., Wilkinson M.B., Walsh J.J., Cooper D.C., Nestler E.J., Han M.H. Mesolimbic dopamine neurons in the brain reward circuit mediate susceptibility to social defeat and antidepressant action. J. Neurosci. 2010;30(49):16453–16458. doi: 10.1523/JNEUROSCI.3177-10.2010. [http://dx.doi.org/10.1523/JNEUROSCI.3177-10.2010]. [PMID: 21147984]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Chaudhury D., Walsh J.J., Friedman A.K., Juarez B., Ku S.M., Koo J.W., Ferguson D., Tsai H.C., Pomeranz L., Christoffel D.J., Nectow A.R., Ekstrand M., Domingos A., Mazei-Robison M.S., Mouzon E., Lobo M.K., Neve R.L., Friedman J.M., Russo S.J., Deisseroth K., Nestler E.J., Han M.H. Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature. 2013;493(7433):532–536. doi: 10.1038/nature11713. [http://dx. doi.org/10.1038/nature11713]. [PMID: 23235832]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Tye K.M., Mirzabekov J.J., Warden M.R., Ferenczi E.A., Tsai H.C., Finkelstein J., Kim S.Y., Adhikari A., Thompson K.R., Andalman A.S., Gunaydin L.A., Witten I.B., Deisseroth K. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature. 2013;493(7433):537–541. doi: 10.1038/nature11740. [http://dx.doi.org/10.1038/nature11740]. [PMID: 23235822]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Goddard A.W., Ball S.G., Martinez J., Robinson M.J., Yang C.R., Russell J.M., Shekhar A. Current perspectives of the roles of the central norepinephrine system in anxiety and depression. Depress. Anxiety. 2010;27(4):339–350. doi: 10.1002/da.20642. [http://dx.doi.org/10.1002/ da.20642]. [PMID: 19960531]. [DOI] [PubMed] [Google Scholar]
  • 185.Isingrini E., Perret L., Rainer Q., Amilhon B., Guma E., Tanti A., Martin G., Robinson J., Moquin L., Marti F., Mechawar N., Williams S., Gratton A., Giros B. Resilience to chronic stress is mediated by noradrenergic regulation of dopamine neurons. Nat. Neurosci. 2016;19(4):560–563. doi: 10.1038/nn.4245. [http://dx.doi.org/10.1038/nn.4245]. [PMID: 26878672]. [DOI] [PubMed] [Google Scholar]
  • 186.Schramm N.L., McDonald M.P., Limbird L.E. The alpha(2a)-adrenergic receptor plays a protective role in mouse behavioral models of depression and anxiety. J. Neurosci. 2001;21(13):4875–4882. doi: 10.1523/JNEUROSCI.21-13-04875.2001. [PMID: 11425914]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Sallinen J., Haapalinna A., MacDonald E., Viitamaa T., Lähdesmäki J., Rybnikova E., Pelto-Huikko M., Kobilka B.K., Scheinin M. Genetic alteration of the alpha2-adrenoceptor subtype c in mice affects the development of behavioral despair and stress-induced increases in plasma corticosterone levels. Mol. Psychiatry. 1999;4(5):443–452. doi: 10.1038/sj.mp.4000543. [http://dx.doi.org/10.1038/sj.mp.4000543]. [PMID: 10523817]. [DOI] [PubMed] [Google Scholar]
  • 188.Saricicek A., Esterlis I., Maloney K.H., Mineur Y.S., Ruf B.M., Muralidharan A., Chen J.I., Cosgrove K.P., Kerestes R., Ghose S., Tamminga C.A., Pittman B., Bois F., Tamagnan G., Seibyl J., Picciotto M.R., Staley J.K., Bhagwagar Z. Persistent β2*-nicotinic acetylcholinergic receptor dysfunction in major depressive disorder. Am. J. Psychiatry. 2012;169(8):851–859. doi: 10.1176/appi.ajp.2012.11101546. [http://dx. doi.org/10.1176/appi.ajp.2012.11101546]. [PMID: 22772158]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Furey M.L., Drevets W.C. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial. Arch. Gen. Psychiatry. 2006;63(10):1121–1129. doi: 10.1001/archpsyc.63.10.1121. [http://dx.doi.org/10.1001/archpsyc.63.10.1121]. [PMID: 17015814]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Mineur Y.S., Obayemi A., Wigestrand M.B., Fote G.M., Calarco C.A., Li A.M., Picciotto M.R. Cholinergic signaling in the hippocampus regulates social stress resilience and anxiety- and depression-like behavior. Proc. Natl. Acad. Sci. USA. 2013;110(9):3573–3578. doi: 10.1073/pnas.1219731110. [http://dx.doi.org/10.1073/pnas.1219731110]. [PMID: 23401542]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Caldarone B.J., Harrist A., Cleary M.A., Beech R.D., King S.L., Picciotto M.R. High-affinity nicotinic acetylcholine receptors are required for antidepressant effects of amitriptyline on behavior and hippocampal cell proliferation. Biol. Psychiatry. 2004;56(9):657–664. doi: 10.1016/j.biopsych.2004.08.010. [http://dx.doi.org/10.1016/j.biopsych.2004.08.010]. [PMID: 15522249]. [DOI] [PubMed] [Google Scholar]
  • 192.Mineur Y.S., Fote G.M., Blakeman S., Cahuzac E.L., Newbold S.A., Picciotto M.R. Multiple nicotinic acetylcholine receptor subtypes in the mouse amygdala regulate affective behaviors and response to social stress. Neuropsychopharmacology. 2016;41(6):1579–1587. doi: 10.1038/npp.2015.316. [http://dx.doi.org/10.1038/npp.2015.316]. [PMID: 26471256]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Semenova S., Contet C., Roberts A.J., Markou A. Mice lacking the β4 subunit of the nicotinic acetylcholine receptor show memory deficits, altered anxiety- and depression-like behavior, and diminished nicotine-induced analgesia. Nicotine Tob. Res. 2012;14(11):1346–1355. doi: 10.1093/ntr/nts107. [http://dx.doi.org/10.1093/ntr/nts107]. [PMID: 22573727]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Lenze E.J., Mulsant B.H., Shear M.K., Alexopoulos G.S., Frank E., Reynolds C.F., III Comorbidity of depression and anxiety disorders in later life. Depress. Anxiety. 2001;14(2):86–93. doi: 10.1002/da.1050. [http:// dx.doi.org/10.1002/da.1050]. [PMID: 11668661]. [DOI] [PubMed] [Google Scholar]
  • 195.Forlani M., Morri M., Belvederi M.M., Bernabei V., Moretti F., Attili T., Biondini A., De Ronchi D., Atti A.R. Anxiety symptoms in 74+ community-dwelling elderly: associations with physical morbidity, depression and alcohol consumption. PLoS One. 2014;9(2):e89859. doi: 10.1371/journal.pone.0089859. [http://dx.doi.org/10.1371/journal.pone. 0089859]. [PMID: 24587079]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Sibille E. Molecular aging of the brain, neuroplasticity, and vulnerability to depression and other brain-related disorders. Dialogues Clin. Neurosci. 2013;15(1):53–65. doi: 10.31887/DCNS.2013.15.1/esibille. [PMID: 23576889]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Perna G., Iannone G., Alciati A., Caldirola D. Are anxiety disorders associated with accelerated aging? A focus on neuroprogression. Neural Plast. 2016;2016:8457612. doi: 10.1155/2016/8457612. [http://dx.doi.org/ 10.1155/2016/8457612]. [PMID: 26881136]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Wolkowitz O.M., Reus V.I., Mellon S.H. Of sound mind and body: depression, disease, and accelerated aging. Dialogues Clin. Neurosci. 2011;13(1):25–39. doi: 10.31887/DCNS.2011.13.1/owolkowitz. [PMID: 21485744]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Evans D.L., Charney D.S., Lewis L., Golden R.N., Gorman J.M., Krishnan K.R., Nemeroff C.B., Bremner J.D., Carney R.M., Coyne J.C., Delong M.R., Frasure-Smith N., Glassman A.H., Gold P.W., Grant I., Gwyther L., Ironson G., Johnson R.L., Kanner A.M., Katon W.J., Kaufmann P.G., Keefe F.J., Ketter T., Laughren T.P., Leserman J., Lyketsos C.G., McDonald W.M., McEwen B.S., Miller A.H., Musselman D., O’Connor C., Petitto J.M., Pollock B.G., Robinson R.G., Roose S.P., Rowland J., Sheline Y., Sheps D.S., Simon G., Spiegel D., Stunkard A., Sunderland T., Tibbits P., Jr, Valvo W.J. Mood disorders in the medically ill: scientific review and recommendations. Biol. Psychiatry. 2005;58(3):175–189. doi: 10.1016/j.biopsych.2005.05.001. [http://dx.doi.org/ 10.1016/j.biopsych.2005.05.001]. [PMID: 16084838]. [DOI] [PubMed] [Google Scholar]
  • 200.Wolkowitz O.M., Epel E.S., Reus V.I., Mellon S.H. Depression gets old fast: do stress and depression accelerate cell aging? Depress. Anxiety. 2010;27(4):327–338. doi: 10.1002/da.20686. [http://dx.doi.org/10.1002/ da.20686]. [PMID: 20376837]. [DOI] [PubMed] [Google Scholar]
  • 201.Verhoeven J.E., Révész D., Epel E.S., Lin J., Wolkowitz O.M., Penninx B.W. Major depressive disorder and accelerated cellular aging: results from a large psychiatric cohort study. Mol. Psychiatry. 2014;19(8):895–901. doi: 10.1038/mp.2013.151. [http://dx.doi.org/10.1038/mp.2013.151]. [PMID: 24217256]. [DOI] [PubMed] [Google Scholar]
  • 202.Miller M.W., Sadeh N. Traumatic stress, oxidative stress and post-traumatic stress disorder: neurodegeneration and the accelerated-aging hypothesis. Mol. Psychiatry. 2014;19(11):1156–1162. doi: 10.1038/mp.2014.111. [http://dx.doi.org/10.1038/mp.2014.111]. [PMID: 25245500]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Kochunov P., Glahn D.C., Rowland L.M., Olvera R.L., Winkler A., Yang Y.H., Sampath H., Carpenter W.T., Duggirala R., Curran J., Blangero J., Hong L.E. Testing the hypothesis of accelerated cerebral white matter aging in schizophrenia and major depression. Biol. Psychiatry. 2013;73(5):482–491. doi: 10.1016/j.biopsych.2012.10.002. [http://dx.doi.org/ 10.1016/j.biopsych.2012.10.002]. [PMID: 23200529]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Zhang X., Norton J., Carrière I., Ritchie K., Chaudieu I., Ancelin M.L. Risk factors for late-onset generalized anxiety disorder: results from a 12-year prospective cohort (the ESPRIT study). Transl. Psychiatry. 2015;5:e536. doi: 10.1038/tp.2015.31. [http://dx.doi.org/10.1038/tp. 2015.31]. [PMID: 25826111]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Maurya P.K., Noto C., Rizzo L.B., Rios A.C., Nunes S.O., Barbosa D.S., Sethi S., Zeni M., Mansur R.B., Maes M., Brietzke E. The role of oxidative and nitrosative stress in accelerated aging and major depressive disorder. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2016;65:134–144. doi: 10.1016/j.pnpbp.2015.08.016. [http://dx.doi. org/10.1016/j.pnpbp.2015.08.016]. [PMID: 26348786]. [DOI] [PubMed] [Google Scholar]
  • 206.Belvederi Murri M., Pariante C., Mondelli V., Masotti M., Atti A.R., Mellacqua Z., Antonioli M., Ghio L., Menchetti M., Zanetidou S., Innamorati M., Amore M. HPA axis and aging in depression: systematic review and meta-analysis. Psychoneuroendocrinology. 2014;41:46–62. doi: 10.1016/j.psyneuen.2013.12.004. [http://dx.doi.org/10.1016/j.psyneuen. 2013.12.004]. [PMID: 24495607]. [DOI] [PubMed] [Google Scholar]
  • 207.Yokoyama K., Yamada T., Mitani H., Yamada S., Pu S., Yamanashi T., Matsumura H., Nakagome K., Kaneko K. Relationship between hypothalamic-pituitary-adrenal axis dysregulation and insulin resistance in elderly patients with depression. Psychiatry Res. 2015;226(2-3):494–498. doi: 10.1016/j.psychres.2015.01.026. [http://dx.doi.org/10.1016/ j.psychres.2015.01.026]. [PMID: 25757913]. [DOI] [PubMed] [Google Scholar]
  • 208.Gupta D., Morley J.E. Hypothalamic-pituitary-adrenal (HPA) axis and aging. Compr. Physiol. 2014;4(4):1495–1510. doi: 10.1002/cphy.c130049. [http://dx.doi. org/10.1002/cphy.c130049]. [PMID: 25428852]. [DOI] [PubMed] [Google Scholar]
  • 209.Gaffey A.E., Bergeman C.S., Clark L.A., Wirth M.M. Aging and the HPA axis: Stress and resilience in older adults. Neurosci. Biobehav. Rev. 2016;68:928–945. doi: 10.1016/j.neubiorev.2016.05.036. [http://dx.doi.org/10.1016/ j.neubiorev.2016.05.036]. [PMID: 27377692]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Simon N.M., Smoller J.W., McNamara K.L., Maser R.S., Zalta A.K., Pollack M.H., Nierenberg A.A., Fava M., Wong K.K. Telomere shortening and mood disorders: preliminary support for a chronic stress model of accelerated aging. Biol. Psychiatry. 2006;60(5):432–435. doi: 10.1016/j.biopsych.2006.02.004. [http://dx.doi.org/10.1016/j.biopsych.2006.02.004]. [PMID: 16581033]. [DOI] [PubMed] [Google Scholar]
  • 211.Huffman D.M., Schafer M.J., LeBrasseur N.K. Energetic interventions for healthspan and resiliency with aging. Exp. Gerontol. 2016;86:73–83. doi: 10.1016/j.exger.2016.05.012. [http://dx.doi.org/10.1016/j.exger.2016.05.012]. [PMID: 27260561]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Hamilton K.L., Miller B.F. What is the evidence for stress resistance and slowed aging? Exp. Gerontol. 2016;82:67–72. doi: 10.1016/j.exger.2016.06.001. [http://dx.doi.org/10.1016/j.exger.2016.06.001]. [PMID: 27268049]. [DOI] [PubMed] [Google Scholar]
  • 213.Miller R.A. Cell stress and aging: new emphasis on multiplex resistance mechanisms. J. Gerontol. A Biol. Sci. Med. Sci. 2009;64(2):179–182. doi: 10.1093/gerona/gln072. [http://dx.doi.org/10.1093/gerona/gln072]. [PMID: 19225033]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Jeste D.V., Savla G.N., Thompson W.K., Vahia I.V., Glorioso D.K., Martin A.S., Palmer B.W., Rock D., Golshan S., Kraemer H.C., Depp C.A. Association between older age and more successful aging: critical role of resilience and depression. Am. J. Psychiatry. 2013;170(2):188–196. doi: 10.1176/appi.ajp.2012.12030386. [http://dx.doi.org/10.1176/appi.ajp. 2012.12030386]. [PMID: 23223917]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.MacLeod S., Musich S., Hawkins K., Alsgaard K., Wicker E.R. The impact of resilience among older adults. Geriatr. Nurs. 2016;37(4):266–272. doi: 10.1016/j.gerinurse.2016.02.014. [http://dx.doi.org/10.1016/j.gerinurse.2016.02.014]. [PMID: 27055911]. [DOI] [PubMed] [Google Scholar]
  • 216.Hamarat E., Thompson D., Aysan F., Steele D., Matheny K., Simons C. Age differences in coping resources and satisfaction with life among middle-aged, young-old, and oldest-old adults. J. Genet. Psychol. 2002;163(3):360–367. doi: 10.1080/00221320209598689. [http://dx.doi.org/10.1080/ 00221320209598689]. [PMID: 12230155]. [DOI] [PubMed] [Google Scholar]
  • 217.Nygren B., Aléx L., Jonsén E., Gustafson Y., Norberg A., Lundman B. Resilience, sense of coherence, purpose in life and self-transcendence in relation to perceived physical and mental health among the oldest old. Aging Ment. Health. 2005;9(4):354–362. doi: 10.1080/1360500114415. [http://dx.doi.org/10.1080/1360500114415]. [PMID: 16019292]. [DOI] [PubMed] [Google Scholar]
  • 218.Gooding P.A., Hurst A., Johnson J., Tarrier N. Psychological resilience in young and older adults. Int. J. Geriatr. Psychiatry. 2012;27(3):262–270. doi: 10.1002/gps.2712. [http://dx.doi.org/10.1002/gps.2712]. [PMID: 21472780]. [DOI] [PubMed] [Google Scholar]
  • 219.Ong A.D., Bergeman C.S., Boker S.M. Resilience comes of age: defining features in later adulthood. J. Pers. 2009;77(6):1777–1804. doi: 10.1111/j.1467-6494.2009.00600.x. [http://dx.doi.org/10.1111/j.1467-6494.2009.00600.x]. [PMID: 19807864]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Epel E.S., Lithgow G.J. Stress biology and aging mechanisms: toward understanding the deep connection between adaptation to stress and longevity. J. Gerontol. A Biol. Sci. Med. Sci. 2014;69(Suppl. 1):S10–S16. doi: 10.1093/gerona/glu055. [http://dx.doi.org/10.1093/gerona/glu055]. [PMID: 24833580]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Cairney J., Krause N. Negative life events and age-related decline in mastery: are older adults more vulnerable to the control-eroding effect of stress? J. Gerontol. B Psychol. Sci. Soc. Sci. 2008;63(3):S162–S170. doi: 10.1093/geronb/63.3.s162. [http://dx.doi.org/10.1093/geronb/63.3.S162]. [PMID: 18559691]. [DOI] [PubMed] [Google Scholar]
  • 222.Osmanovic-Thunström A., Mossello E., Åkerstedt T., Fratiglioni L., Wang H.X. Do levels of perceived stress increase with increasing age after age 65? A population-based study. Age Ageing. 2015;44(5):828–834. doi: 10.1093/ageing/afv078. [http://dx.doi.org/10.1093/ageing/afv078]. [PMID: 26187986]. [DOI] [PubMed] [Google Scholar]
  • 223.Novais A., Monteiro S., Roque S., Correia-Neves M., Sousa N. How age, sex and genotype shape the stress response. Neurobiol. Stress. 2016;6:44–56. doi: 10.1016/j.ynstr.2016.11.004. [http://dx.doi.org/10.1016/j.ynstr.2016.11. 004]. [PMID: 28229108]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Flood M., Buckwalter K.C. Recommendations for mental health care of older adults: Part 1--an overview of depression and anxiety. J. Gerontol. Nurs. 2009;35(2):26–34. doi: 10.3928/00989134-20090201-03. [http://dx.doi.org/10.3928/ 00989134-20090201-03]. [PMID: 19263919]. [DOI] [PubMed] [Google Scholar]
  • 225.Parmentier H., García-Campayo J., Prieto R. Comprehensive review of generalized anxiety disorder in primary care in Europe. Curr. Med. Res. Opin. 2013;29(4):355–367. doi: 10.1185/03007995.2013.770731. [http://dx.doi.org/10. 1185/03007995.2013.770731]. [PMID: 23356728]. [DOI] [PubMed] [Google Scholar]
  • 226.Zhang X., Norton J., Carrière I., Ritchie K., Chaudieu I., Ancelin M.L. Generalized anxiety in community-dwelling elderly: Prevalence and clinical characteristics. J. Affect. Disord. 2015;172:24–29. doi: 10.1016/j.jad.2014.09.036. [http://dx.doi.org/10.1016/j.jad.2014.09.036]. [PMID: 25451391]. [DOI] [PubMed] [Google Scholar]
  • 227.Bryant C., Jackson H., Ames D. The prevalence of anxiety in older adults: methodological issues and a review of the literature. J. Affect. Disord. 2008;109(3):233–250. doi: 10.1016/j.jad.2007.11.008. [http://dx.doi.org/10.1016/ j.jad.2007.11.008]. [PMID: 18155775]. [DOI] [PubMed] [Google Scholar]
  • 228.Beekman A.T., Copeland J.R., Prince M.J. Review of community prevalence of depression in later life. Br. J. Psychiatry. 1999;174:307–311. doi: 10.1192/bjp.174.4.307. [http://dx.doi.org/10.1192/bjp.174.4.307]. [PMID: 10533549]. [DOI] [PubMed] [Google Scholar]
  • 229.Djernes J.K. Prevalence and predictors of depression in populations of elderly: a review. Acta Psychiatr. Scand. 2006;113(5):372–387. doi: 10.1111/j.1600-0447.2006.00770.x. [http://dx.doi.org/10.1111/j.1600-0447.2006.00770.x]. [PMID: 16603029]. [DOI] [PubMed] [Google Scholar]
  • 230.Lenze E.J., Mulsant B.H., Shear M.K., Schulberg H.C., Dew M.A., Begley A.E., Pollock B.G., Reynolds C.F., III Comorbid anxiety disorders in depressed elderly patients. Am. J. Psychiatry. 2000;157(5):722–728. doi: 10.1176/appi.ajp.157.5.722. [http://dx.doi.org/10.1176/appi.ajp.157.5. 722]. [PMID: 10784464]. [DOI] [PubMed] [Google Scholar]
  • 231.Cummings C.M., Caporino N.E., Kendall P.C. Comorbidity of anxiety and depression in children and adolescents: 20 years after. Psychol. Bull. 2014;140(3):816–845. doi: 10.1037/a0034733. [http://dx.doi.org/10.1037/ a0034733]. [PMID: 24219155]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Kimhi S., Hantman S., Goroshit M., Eshel Y., Zysberg L. Elderly people coping with the aftermath of war: resilience versus vulnerability. Am. J. Geriatr. Psychiatry. 2012;20(5):391–401. doi: 10.1097/JGP.0b013e31821106b3. [http:// dx.doi.org/10.1097/JGP.0b013e31821106b3]. [PMID: 21358387]. [DOI] [PubMed] [Google Scholar]
  • 233.Kessler R.C., Berglund P., Demler O., Jin R., Merikangas K.R., Walters E.E. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch. Gen. Psychiatry. 2005;62(6):593–602. doi: 10.1001/archpsyc.62.6.593. [http://dx.doi. org/10.1001/archpsyc.62.6.593]. [PMID: 15939837]. [DOI] [PubMed] [Google Scholar]
  • 234.Mota N., Tsai J., Kirwin P.D., Harpaz-Rotem I., Krystal J.H., Southwick S.M., Pietrzak R.H. Late-life exacerbation of PTSD symptoms in US veterans: results from the National Health and Resilience in Veterans Study. J. Clin. Psychiatry. 2016;77(3):348–354. doi: 10.4088/JCP.15m10101. [http://dx.doi.org/10.4088/JCP.15m10101]. [PMID: 27046308]. [DOI] [PubMed] [Google Scholar]
  • 235.Kuwert P., Knaevelsrud C., Pietrzak R.H. Loneliness among older veterans in the United States: results from the National Health and Resilience in Veterans Study. Am. J. Geriatr. Psychiatry. 2014;22(6):564–569. doi: 10.1016/j.jagp.2013.02.013. [http://dx.doi.org/10.1016/j.jagp.2013.02.013]. [PMID: 23806682]. [DOI] [PubMed] [Google Scholar]
  • 236.Fanning J.R., Pietrzak R.H. Suicidality among older male veterans in the United States: results from the National Health and Resilience in Veterans Study. J. Psychiatr. Res. 2013;47(11):1766–1775. doi: 10.1016/j.jpsychires.2013.07.015. [http://dx.doi.org/10.1016/j.jpsychires.2013.07.015]. [PMID: 23992768]. [DOI] [PubMed] [Google Scholar]
  • 237.Pietrzak R.H., Cook J.M. Psychological resilience in older U.S. veterans: results from the national health and resilience in veterans study. Depress. Anxiety. 2013;30(5):432–443. doi: 10.1002/da.22083. [http://dx.doi.org/ 10.1002/da.22083]. [PMID: 23468170]. [DOI] [PubMed] [Google Scholar]
  • 238.Association A.P. Stress in America. 2011 http://www.apa.org/ news/press/releases/stress/2011/generations.aspx
  • 239.Sánchez-Hidalgo A.C., Muñoz M.F., Herrera A.J., Espinosa-Oliva A.M., Stowell R., Ayala A., Machado A., Venero J.L., de Pablos R.M. Chronic stress alters the expression levels of longevity-related genes in the rat hippocampus. Neurochem. Int. 2016;97:181–192. doi: 10.1016/j.neuint.2016.04.009. [http://dx.doi.org/10.1016/j.neuint.2016.04.009]. [PMID: 27120255]. [DOI] [PubMed] [Google Scholar]
  • 240.Koenig J.I., Walker C.D., Romeo R.D., Lupien S.J. Effects of stress across the lifespan. Stress. 2011;14(5):475–480. doi: 10.3109/10253890.2011.604879. [http:// dx.doi.org/10.3109/10253890.2011.604879]. [PMID: 21848435]. [DOI] [PubMed] [Google Scholar]
  • 241.Ulrich-Lai Y.M., Ryan K.K. PPARγ and stress: implications for aging. Exp. Gerontol. 2013;48(7):671–676. doi: 10.1016/j.exger.2012.08.011. [http://dx.doi.org/10. 1016/j.exger.2012.08.011]. [PMID: 22960592]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Khan A., McCormack H.C., Bolger E.A., McGreenery C.E., Vitaliano G., Polcari A., Teicher M.H. Childhood maltreatment, depression, and suicidal ideation: critical importance of parental and peer emotional abuse during developmental sensitive periods in males and females. Front. Psychiatry. 2015;6:42. doi: 10.3389/fpsyt.2015.00042. [http://dx. doi.org/10.3389/fpsyt.2015.00042]. [PMID: 25870565]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Gill T.M., Robison J.T., Tinetti M.E. Predictors of recovery in activities of daily living among disabled older persons living in the community. J. Gen. Intern. Med. 1997;12(12):757–762. doi: 10.1046/j.1525-1497.1997.07161.x. [http:// dx.doi.org/10.1046/j.1525-1497.1997.07161.x]. [PMID: 9436895]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Whitson H.E., Thielke S., Diehr P., O’Hare A.M., Chaves P.H., Zakai N.A., Arnold A., Chaudhry S., Ives D., Newman A.B. Patterns and predictors of recovery from exhaustion in older adults: the cardiovascular health study. J. Am. Geriatr. Soc. 2011;59(2):207–213. doi: 10.1111/j.1532-5415.2010.03238.x. [http://dx.doi.org/10.1111/j.1532-5415.2010.03238.x]. [PMID: 21288229]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Zeng Y., Shen K. Resilience significantly contributes to exceptional longevity. Curr. Gerontol. Geriatr. Res. 2010;2010:525693. doi: 10.1155/2010/525693. [http://dx.doi.org/10.1155/2010/525693]. [PMID: 21197075]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Harris P.B. Another wrinkle in the debate about successful aging: the undervalued concept of resilience and the lived experience of dementia. Int. J. Aging Hum. Dev. 2008;67(1):43–61. doi: 10.2190/AG.67.1.c. [http://dx. doi.org/10.2190/AG.67.1.c]. [PMID: 18630190]. [DOI] [PubMed] [Google Scholar]
  • 247.Shalev I., Entringer S., Wadhwa P.D., Wolkowitz O.M. ; Puterman E., Lin J., Epel E.S. Stress and telomere biology: a lifespan perspective. Psychoneuroendocrinology. 2013;38(9):1835–1842. doi: 10.1016/j.psyneuen.2013.03.010. [http://dx.doi.org/10.1016/j.psyneuen.2013.03.010]. [PMID: 23639252]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Starkweather A.R., Alhaeeri A.A., Montpetit A., Brumelle J., Filler K., Montpetit M., Mohanraj L., Lyon D.E., Jackson-Cook C.K. An integrative review of factors associated with telomere length and implications for biobehavioral research. Nurs. Res. 2014;63(1):36–50. doi: 10.1097/NNR.0000000000000009. [http://dx.doi.org/10.1097/NNR.0000000000000009]. [PMID: 24335912]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Price L.H., Kao H.T., Burgers D.E., Carpenter L.L., Tyrka A.R. Telomeres and early-life stress: an overview. Biol. Psychiatry. 2013;73(1):15–23. doi: 10.1016/j.biopsych.2012.06.025. [http://dx.doi.org/10.1016/j.biopsych.2012.06. 025]. [PMID: 22831981]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Goldberg E.L., Romero-Aleshire M.J., Renkema K.R., Ventevogel M.S., Chew W.M., Uhrlaub J.L., Smithey M.J., Limesand K.H., Sempowski G.D., Brooks H.L., Nikolich-Žugich J. Lifespan-extending caloric restriction or mTOR inhibition impair adaptive immunity of old mice by distinct mechanisms. Aging Cell. 2015;14(1):130–138. doi: 10.1111/acel.12280. [http://dx.doi.org/10.1111/acel.12280]. [PMID: 25424641]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Zhang L., Hu X.Z., Li X., Li H., Smerin S., Russell D., Ursano R.J. Telomere length - a cellular aging marker for depression and Post-traumatic Stress Disorder. Med. Hypotheses. 2014;83(2):182–185. doi: 10.1016/j.mehy.2014.04.033. [http://dx.doi.org/10.1016/j.mehy.2014.04.033]. [PMID: 24875221]. [DOI] [PubMed] [Google Scholar]
  • 252.Epel E. How “reversible” is telomeric aging? Cancer Prev. Res. (Phila.) 2012;5(10):1163–1168. doi: 10.1158/1940-6207.CAPR-12-0370. [http://dx.doi.org/10.1158/1940-6207.CAPR-12-0370]. [PMID: 23041472]. [DOI] [PubMed] [Google Scholar]
  • 253.Saretzki G., von Zglinicki T. Telomerase as a promising target for human cancer gene therapy. Drugs Today (Barc) 2003;39(4):265–276. doi: 10.1358/dot.2003.39.4.799403. [http://dx.doi.org/10.1358/dot.2003.39.4.799403]. [PMID: 12743642]. [DOI] [PubMed] [Google Scholar]
  • 254.OʼHara R.; Marcus, P.; Thompson, W.K.; Flournoy, J.; Vahia, I.; Lin, X.; Hallmayer, J.; Depp, C.; Jeste, D.V. 5-HTTLPR short allele, resilience, and successful aging in older adults. Am. J. Geriatr. Psychiatry. 2012;20(5):452–456. doi: 10.1097/JGP.0b013e31823e2d03. [http://dx.doi.org/10.1097/JGP. 0b013e31823e2d03]. [PMID: 22233775]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Sabbagh J.J., O’Leary J.C., III, Blair L.J., Klengel T., Nordhues B.A., Fontaine S.N., Binder E.B., Dickey C.A. Age-associated epigenetic upregulation of the FKBP5 gene selectively impairs stress resiliency. PLoS One. 2014;9(9):e107241. doi: 10.1371/journal.pone.0107241. [http://dx.doi.org/ 10.1371/journal.pone.0107241]. [PMID: 25191701]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Berdasco M., Esteller M. Hot topics in epigenetic mechanisms of aging: 2011. Aging Cell. 2012;11(2):181–186. doi: 10.1111/j.1474-9726.2012.00806.x. [http://dx.doi.org/ 10.1111/j.1474-9726.2012.00806.x]. [PMID: 22321768]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Adams P.D., Jasper H., Rudolph K.L. Aging-induced stem cell mutations as drivers for disease and cancer. Cell Stem Cell. 2015;16(6):601–612. doi: 10.1016/j.stem.2015.05.002. [http://dx.doi.org/10.1016/j.stem.2015.05.002]. [PMID: 26046760]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Babenko O., Kovalchuk I., Metz G.A. Stress-induced perinatal and transgenerational epigenetic programming of brain development and mental health. Neurosci. Biobehav. Rev. 2015;48:70–91. doi: 10.1016/j.neubiorev.2014.11.013. [http:// dx.doi.org/10.1016/j.neubiorev.2014.11.013]. [PMID: 25464029]. [DOI] [PubMed] [Google Scholar]
  • 259.López-Otín C., Galluzzi L., Freije J.M., Madeo F., Kroemer G. Metabolic control of longevity. Cell. 2016;166(4):802–821. doi: 10.1016/j.cell.2016.07.031. [http://dx.doi.org/10.1016/j.cell.2016.07.031]. [PMID: 27518560]. [DOI] [PubMed] [Google Scholar]
  • 260.Cole J.J., Robertson N.A., Rather M.I., Thomson J.P., McBryan T., Sproul D., Wang T., Brock C., Clark W., Ideker T., Meehan R.R., Miller R.A., Brown-Borg H.M., Adams P.D. Diverse interventions that extend mouse lifespan suppress shared age-associated epigenetic changes at critical gene regulatory regions. Genome Biol. 2017;18(1):58. doi: 10.1186/s13059-017-1185-3. [http://dx.doi.org/10.1186/s13059-017-1185-3]. [PMID: 28351383]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Dias C., Feng J., Sun H., Shao N.Y., Mazei-Robison M.S., Damez-Werno D., Scobie K., Bagot R., LaBonté B., Ribeiro E., Liu X., Kennedy P., Vialou V., Ferguson D., Peña C., Calipari E.S., Koo J.W., Mouzon E., Ghose S., Tamminga C., Neve R., Shen L., Nestler E.J. β-catenin mediates stress resilience through Dicer1/microRNA regulation. Nature. 2014;516(7529):51–55. doi: 10.1038/nature13976. [PMID: 25383518]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Rao J.A., Kassel M.T., Weldon A.L., Avery E.T., Briceno E.M., Mann M., Cornett B., Kales H.C., Zubieta J.K., Welsh R.C., Langenecker S.A., Weisenbach S.L. The double burden of age and major depressive disorder on the cognitive control network. Psychol. Aging. 2015;30(2):475–485. doi: 10.1037/pag0000027. [http://dx.doi.org/10.1037/ pag0000027]. [PMID: 26030776]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Shimada H., Park H., Makizako H., Doi T., Lee S., Suzuki T. Depressive symptoms and cognitive performance in older adults. J. Psychiatr. Res. 2014;57:149–156. doi: 10.1016/j.jpsychires.2014.06.004. [http://dx.doi.org/10.1016/ j.jpsychires.2014.06.004]. [PMID: 25023083]. [DOI] [PubMed] [Google Scholar]
  • 264.Diniz B.S., Sibille E., Ding Y., Tseng G., Aizenstein H.J., Lotrich F., Becker J.T., Lopez O.L., Lotze M.T., Klunk W.E., Reynolds C.F., Butters M.A. Plasma biosignature and brain pathology related to persistent cognitive impairment in late-life depression. Mol. Psychiatry. 2015;20(5):594–601. doi: 10.1038/mp.2014.76. [http://dx.doi.org/ 10.1038/mp.2014.76]. [PMID: 25092249]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Wilkins C.H., Mathews J., Sheline Y.I. Late life depression with cognitive impairment: evaluation and treatment. Clin. Interv. Aging. 2009;4:51–57. [PMID: 19503765]. [PMC free article] [PubMed] [Google Scholar]
  • 266.Nozaki S., Yoshimura K., Mimura M. Depression and dementia: perspectives from clinical studies. Brain Nerve. 2012;64(12):1387–1397. [PMID: 23209065]. [PubMed] [Google Scholar]
  • 267.Oh H., Madison C., Villeneuve S., Markley C., Jagust W.J. Association of gray matter atrophy with age, β-amyloid, and cognition in aging. Cereb. Cortex. 2014;24(6):1609–1618. doi: 10.1093/cercor/bht017. [http://dx. doi.org/10.1093/cercor/bht017]. [PMID: 23389995]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.de Groot M., Ikram M.A., Akoudad S., Krestin G.P., Hofman A., van der Lugt A., Niessen W.J., Vernooij M.W. Tract-specific white matter degeneration in aging: the Rotterdam Study. Alzheimers Dement. 2015;11(3):321–330. doi: 10.1016/j.jalz.2014.06.011. [http://dx.doi.org/10.1016/ j.jalz.2014.06.011]. [PMID: 25217294]. [DOI] [PubMed] [Google Scholar]
  • 269.Vu N.Q., Aizenstein H.J. Depression in the elderly: brain correlates, neuropsychological findings, and role of vascular lesion load. Curr. Opin. Neurol. 2013;26(6):656–661. doi: 10.1097/WCO.0000000000000028. [http://dx.doi.org/10. 1097/WCO.0000000000000028]. [PMID: 24184971]. [DOI] [PubMed] [Google Scholar]
  • 270.Bettio L.E., Rajendran L., Gil-Mohapel J. The effects of aging in the hippocampus and cognitive decline. Neurosci. Biobehav. Rev. 2017;79:66–86. doi: 10.1016/j.neubiorev.2017.04.030. [http://dx.doi.org/10.1016/j.neubiorev.2017.04. 030]. [PMID: 28476525]. [DOI] [PubMed] [Google Scholar]
  • 271.Elbejjani M., Fuhrer R., Abrahamowicz M., Mazoyer B., Crivello F., Tzourio C., Dufouil C. Depression, depressive symptoms, and rate of hippocampal atrophy in a longitudinal cohort of older men and women. Psychol. Med. 2015;45(9):1931–1944. doi: 10.1017/S0033291714003055. [http:// dx.doi.org/10.1017/S0033291714003055]. [PMID: 25896060]. [DOI] [PubMed] [Google Scholar]
  • 272.Boldrini M., Hen R., Underwood M.D., Rosoklija G.B., Dwork A.J., Mann J.J., Arango V. Hippocampal angiogenesis and progenitor cell proliferation are increased with antidepressant use in major depression. Biol. Psychiatry. 2012;72(7):562–571. doi: 10.1016/j.biopsych.2012.04.024. [http:// dx.doi.org/10.1016/j.biopsych.2012.04.024]. [PMID: 22652019]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Boldrini M., Santiago A.N., Hen R., Dwork A.J., Rosoklija G.B., Tamir H., Arango V., John M.J. Hippocampal granule neuron number and dentate gyrus volume in antidepressant-treated and untreated major depression. Neuropsychopharmacology. 2013;38(6):1068–1077. doi: 10.1038/npp.2013.5. [http://dx.doi.org/10.1038/npp.2013.5]. [PMID: 23303074]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Nashiro K., Sakaki M., Mather M. Age differences in brain activity during emotion processing: reflections of age-related decline or increased emotion regulation? Gerontology. 2012;58(2):156–163. doi: 10.1159/000328465. [http://dx.doi.org/10.1159/000328465]. [PMID: 21691052]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Tadayonnejad R., Ajilore O. Brain network dysfunction in late-life depression: a literature review. J. Geriatr. Psychiatry Neurol. 2014;27(1):5–12. doi: 10.1177/0891988713516539. [http://dx.doi.org/10.1177/0891988713516539]. [PMID: 24381233]. [DOI] [PubMed] [Google Scholar]
  • 276.Brydges N.M., Wood E.R., Holmes M.C., Hall J. Prepubertal stress and hippocampal function: sex-specific effects. Hippocampus. 2014;24(6):684–692. doi: 10.1002/hipo.22259. [http://dx.doi.org/10.1002/hipo.22259]. [PMID: 24677338]. [DOI] [PubMed] [Google Scholar]
  • 277.Ege M.A., Messias E., Thapa P.B., Krain L.P. Adverse childhood experiences and geriatric depression: results from the 2010 BRFSS. Am. J. Geriatr. Psychiatry. 2015;23(1):110–114. doi: 10.1016/j.jagp.2014.08.014. [http:// dx.doi.org/10.1016/j.jagp.2014.08.014]. [PMID: 25306195]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Schalinski I., Teicher M.H., Nischk D., Hinderer E., Müller O., Rockstroh B. Type and timing of adverse childhood experiences differentially affect severity of PTSD, dissociative and depressive symptoms in adult inpatients. BMC Psychiatry. 2016;16:295. doi: 10.1186/s12888-016-1004-5. [http://dx.doi.org/10.1186/s12888-016-1004-5]. [PMID: 27543114]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Danese A., McEwen B.S. Adverse childhood experiences, allostasis, allostatic load, and age-related disease. Physiol. Behav. 2012;106(1):29–39. doi: 10.1016/j.physbeh.2011.08.019. [http://dx.doi.org/10.1016/j.physbeh.2011.08.019]. [PMID: 21888923]. [DOI] [PubMed] [Google Scholar]
  • 280.Cole S.W. Social regulation of human gene expression: mechanisms and implications for public health. Am. J. Public Health. 2013;103(Suppl. 1):S84–S92. doi: 10.2105/AJPH.2012.301183. [http://dx.doi.org/10.2105/AJPH. 2012.301183]. [PMID: 23927506]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Juruena M.F. Early-life stress and HPA axis trigger recurrent adulthood depression. Epilepsy Behav. 2014;38:148–159. doi: 10.1016/j.yebeh.2013.10.020. [http://dx.doi.org/10.1016/j.yebeh.2013.10.020]. [PMID: 24269030]. [DOI] [PubMed] [Google Scholar]
  • 282.Tyrka A.R., Price L.H., Kao H.T., Porton B., Marsella S.A., Carpenter L.L. Childhood maltreatment and telomere shortening: preliminary support for an effect of early stress on cellular aging. Biol. Psychiatry. 2010;67(6):531–534. doi: 10.1016/j.biopsych.2009.08.014. [http://dx.doi.org/10.1016/ j.biopsych.2009.08.014]. [PMID: 19828140]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Gingrich J.A., Ansorge M.S., Merker R., Weisstaub N., Zhou M. New lessons from knockout mice: The role of serotonin during development and its possible contribution to the origins of neuropsychiatric disorders. CNS Spectr. 2003;8(8):572–577. doi: 10.1017/s1092852900018848. [http://dx.doi.org/10.1017/S1092852900018848]. [PMID: 12907920]. [DOI] [PubMed] [Google Scholar]
  • 284.Ansorge M.S., Zhou M., Lira A., Hen R., Gingrich J.A. Early-life blockade of the 5-HT transporter alters emotional behavior in adult mice. Science. 2004;306(5697):879–881. doi: 10.1126/science.1101678. [http://dx.doi.org/ 10.1126/science.1101678]. [PMID: 15514160]. [DOI] [PubMed] [Google Scholar]
  • 285.Austad S.N., Bartke A. Sex Differences in Longevity and in Responses to Anti-Aging Interventions: A Mini-Review. Gerontology. 2015;62(1):40–46. doi: 10.1159/000381472. [http://dx.doi.org/10.1159/000381472]. [PMID: 25968226]. [DOI] [PubMed] [Google Scholar]
  • 286.Barrett E.L., Richardson D.S. Sex differences in telomeres and lifespan. Aging Cell. 2011;10(6):913–921. doi: 10.1111/j.1474-9726.2011.00741.x. [http://dx.doi.org/10. 1111/j.1474-9726.2011.00741.x]. [PMID: 21902801]. [DOI] [PubMed] [Google Scholar]
  • 287.Scheinost D., Finn E.S., Tokoglu F., Shen X., Papademetris X., Hampson M., Constable R.T. Sex differences in normal age trajectories of functional brain networks. Hum. Brain Mapp. 2015;36(4):1524–1535. doi: 10.1002/hbm.22720. [http://dx.doi.org/10.1002/hbm.22720]. [PMID: 25523617]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Murtagh K.N., Hubert H.B. Gender differences in physical disability among an elderly cohort. Am. J. Public Health. 2004;94(8):1406–1411. doi: 10.2105/ajph.94.8.1406. [http://dx.doi.org/10.2105/AJPH.94.8.1406]. [PMID: 15284051]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Dalla C., Antoniou K., Drossopoulou G., Xagoraris M., Kokras N., Sfikakis A., Papadopoulou-Daifoti Z. Chronic mild stress impact: are females more vulnerable? Neuroscience. 2005;135(3):703–714. doi: 10.1016/j.neuroscience.2005.06.068. [http://dx.doi.org/10.1016/j.neuroscience.2005.06.068]. [PMID: 16125862]. [DOI] [PubMed] [Google Scholar]
  • 290.Bale T.L., Epperson C.N. Sex differences and stress across the lifespan. Nat. Neurosci. 2015;18(10):1413–1420. doi: 10.1038/nn.4112. [http://dx.doi. org/10.1038/nn.4112]. [PMID: 26404716]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Breslau N., Wilcox H.C., Storr C.L., Lucia V.C., Anthony J.C. Trauma exposure and posttraumatic stress disorder: a study of youths in urban America. J. Urban Health. 2004;81(4):530–544. doi: 10.1093/jurban/jth138. [http://dx.doi.org/10.1093/jurban/jth138]. [PMID: 15466836]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Wang Q., Joels M., Swaab D.F., Lucassen P.J. Hippocampal GR expression is increased in elderly depressed females. Neuropharmacology. 2012;62(1):527–533. doi: 10.1016/j.neuropharm.2011.09.014. [http://dx.doi.org/10.1016/ j.neuropharm.2011.09.014]. [PMID: 21945289]. [DOI] [PubMed] [Google Scholar]
  • 293.Qureshi I.A., Mehler M.F. Genetic and epigenetic underpinnings of sex differences in the brain and in neurological and psychiatric disease susceptibility. 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Siegel A.M., Mathews S.B. Diagnosis and Treatment of Anxiety in the Aging Woman. Curr. Psychiatry Rep. 2015;17(12):93. doi: 10.1007/s11920-015-0636-3. [http://dx.doi.org/10.1007/s11920-015-0636-3]. [PMID: 26458819]. [DOI] [PubMed] [Google Scholar]
  • 295.Bromberger J.T., Assmann S.F., Avis N.E., Schocken M., Kravitz H.M., Cordal A. Persistent mood symptoms in a multiethnic community cohort of pre- and perimenopausal women. Am. J. Epidemiol. 2003;158(4):347–356. doi: 10.1093/aje/kwg155. [http://dx.doi.org/10.1093/ aje/kwg155]. [PMID: 12915500]. [DOI] [PubMed] [Google Scholar]
  • 296.Sagsöz N., Oguztürk O., Bayram M., Kamaci M. Anxiety and depression before and after the menopause. Arch. Gynecol. Obstet. 2001;264(4):199–202. doi: 10.1007/s004040000108. [http://dx.doi.org/10.1007/s004040000108]. [PMID: 11205708]. [DOI] [PubMed] [Google Scholar]
  • 297.Woods N.F., Mariella A., Mitchell E.S. Depressed mood symptoms during the menopausal transition: observations from the Seattle Midlife Women’s Health Study. Climacteric. 2006;9(3):195–203. doi: 10.1080/13697130600730663. [http://dx.doi.org/10.1080/13697130600730663]. [PMID: 16766433]. [DOI] [PubMed] [Google Scholar]
  • 298.Freeman E.W., Sammel M.D., Liu L., Gracia C.R., Nelson D.B., Hollander L. Hormones and menopausal status as predictors of depression in women in transition to menopause. Arch. Gen. Psychiatry. 2004;61(1):62–70. doi: 10.1001/archpsyc.61.1.62. [http://dx.doi.org/10.1001/archpsyc. 61.1.62]. [PMID: 14706945]. [DOI] [PubMed] [Google Scholar]
  • 299.Nemeroff C.B. Stress, menopause and vulnerability for psychiatric illness. Expert Rev. Neurother. 2007;7(11) Suppl.:S11–S13. doi: 10.1586/14737175.7.11s.S11. [http://dx.doi.org/10.1586/14737175.7.11s.S11]. [PMID: 18039060]. [DOI] [PubMed] [Google Scholar]
  • 300.Freeman E.W., Sammel M.D., Boorman D.W., Zhang R. Longitudinal pattern of depressive symptoms around natural menopause. JAMA Psychiatry. 2014;71(1):36–43. doi: 10.1001/jamapsychiatry.2013.2819. [http://dx.doi.org/10.1001/ jamapsychiatry.2013.2819]. [PMID: 24227182]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Mebes I., Graf M., Kellner M., Keck C., Segerer S.E. High estradiol levels during postmenopause - pitfalls in laboratory analysis. Geburtshilfe Frauenheilkd. 2015;75(9):941–944. doi: 10.1055/s-0035-1557815. [http://dx.doi.org/10.1055/s-0035-1557815]. [PMID: 26500371]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Dumas J.A., Albert K.M., Naylor M.R., Sites C.K., Benkelfat C., Newhouse P.A. The effects of age and estrogen on stress responsivity in older women. Am. J. Geriatr. Psychiatry. 2012;20(9):734–743. doi: 10.1097/JGP.0b013e31825c0a14. [http://dx.doi.org/10.1097/JGP.0b013e31825c0a14]. [PMID: 22832417]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Newhouse P.A., Dumas J., Wilkins H., Coderre E., Sites C.K., Naylor M., Benkelfat C., Young S.N. Estrogen treatment impairs cognitive performance after psychosocial stress and monoamine depletion in postmenopausal women. Menopause. 2010;17(4):860–873. doi: 10.1097/gme.0b013e3181e15df4. [http://dx.doi.org/10.1097/gme.0b013e3181e15df4]. [PMID: 20616673]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Albert K., Pruessner J., Newhouse P. Estradiol levels modulate brain activity and negative responses to psychosocial stress across the menstrual cycle. Psychoneuroendocrinology. 2015;59:14–24. doi: 10.1016/j.psyneuen.2015.04.022. [http://dx.doi.org/10.1016/j.psyneuen.2015.04.022]. [PMID: 26123902]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Kelly S.D., Harrell C.S., Neigh G.N. Chronic stress modulates regional cerebral glucose transporter expression in an age-specific and sexually-dimorphic manner. Physiol. Behav. 2014;126:39–49. doi: 10.1016/j.physbeh.2013.12.002. [http://dx.doi.org/10.1016/j.physbeh.2013.12.002]. [PMID: 24382486]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Newman R. APA’s Resilience Initiative. Prof. Psychol. Res. Pr. 2005;36(3):227–229. [http://dx.doi.org/10.1037/0735-7028.36.3.227]. [Google Scholar]
  • 307.Shors T.J., Olson R.L., Bates M.E., Selby E.A., Alderman B.L. Mental and physical (MAP) Training: a neurogenesis-inspired intervention that enhances health in humans. Neurobiol. Learn. Mem. 2014;115:3–9. doi: 10.1016/j.nlm.2014.08.012. [http://dx.doi.org/10.1016/j.nlm.2014.08.012]. [PMID: 25219804]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Denning Laura Aiuppa, Meisnere Marc, Warner Kenneth E. 2014.
  • 309.Leppin A.L., Bora P.R., Tilburt J.C., Gionfriddo M.R., Zeballos-Palacios C., Dulohery M.M., Sood A., Erwin P.J., Brito J.P., Boehmer K.R., Montori V.M. The efficacy of resiliency training programs: a systematic review and meta-analysis of randomized trials. PLoS One. 2014;9(10):e111420. doi: 10.1371/journal.pone.0111420. [http://dx.doi.org/10.1371/ journal.pone.0111420]. [PMID: 25347713]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Loving R.T., Kripke D.F., Elliott J.A., Knickerbocker N.C., Grandner M.A. Bright light treatment of depression for older adults. BMC Psychiatry. 2005;5:41. doi: 10.1186/1471-244X-5-41. [ISRCTN55452501]. [ISRCTN55452501]. [http://dx.doi.org/10.1186/1471-244X-5-41]. [PMID: 16283925]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Lieverse R., Van Someren E.J., Nielen M.M., Uitdehaag B.M., Smit J.H., Hoogendijk W.J. Bright light treatment in elderly patients with nonseasonal major depressive disorder: a randomized placebo-controlled trial. Arch. Gen. Psychiatry. 2011;68(1):61–70. doi: 10.1001/archgenpsychiatry.2010.183. [http://dx.doi.org/10.1001/archgenpsychiatry.2010.183]. [PMID: 21199966]. [DOI] [PubMed] [Google Scholar]
  • 312.Lapierre S., Erlangsen A., Waern M., De Leo D., Oyama H., Scocco P., Gallo J., Szanto K., Conwell Y., Draper B., Quinnett P. A systematic review of elderly suicide prevention programs. Crisis. 2011;32(2):88–98. doi: 10.1027/0227-5910/a000076. [http://dx.doi.org/10.1027/0227-5910/a000076]. [PMID: 21602163]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Smith J.L., Hollinger-Smith L. Savoring, resilience, and psychological well-being in older adults. Aging Ment. Health. 2015;19(3):192–200. doi: 10.1080/13607863.2014.986647. [http://dx.doi.org/10.1080/13607863.2014.986647]. [PMID: 25471325]. [DOI] [PubMed] [Google Scholar]
  • 314.Lyubomirsky S., Sousa L., Dickerhoof R. The costs and benefits of writing, talking, and thinking about life’s triumphs and defeats. J. Pers. Soc. Psychol. 2006;90(4):692–708. doi: 10.1037/0022-3514.90.4.692. [http://dx.doi.org/10. 1037/0022-3514.90.4.692]. [PMID: 16649864]. [DOI] [PubMed] [Google Scholar]
  • 315.Bretherton S.J., McLean L.A. Interrelations of stress, optimism and control in older people’s psychological adjustment. Australas. J. Ageing. 2015;34(2):103–108. doi: 10.1111/ajag.12138. [http://dx.doi.org/10.1111/ajag. 12138]. [PMID: 24629026]. [DOI] [PubMed] [Google Scholar]
  • 316.Mancini A.D., Bonanno G.A. Resilience in the face of potential trauma: clinical practices and illustrations. J. Clin. Psychol. 2006;62(8):971–985. doi: 10.1002/jclp.20283. [http://dx.doi.org/10.1002/jclp.20283]. [PMID: 16700017]. [DOI] [PubMed] [Google Scholar]
  • 317.Mancini A.D., Bonanno G.A. Predictors and parameters of resilience to loss: toward an individual differences model. J. Pers. 2009;77(6):1805–1832. doi: 10.1111/j.1467-6494.2009.00601.x. [http://dx.doi.org/10.1111/j.1467-6494. 2009.00601.x]. [PMID: 19807863]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Yoshikawa E., Nishi D., Matsuoka Y.J. Association between regular physical exercise and depressive symptoms mediated through social support and resilience in Japanese company workers: a cross-sectional study. BMC Public Health. 2016;16:553. doi: 10.1186/s12889-016-3251-2. [http://dx.doi.org/10.1186/s12889-016-3251-2]. [PMID: 27405459]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Deuster P.A., Silverman M.N. Physical fitness: a pathway to health and resilience. U.S. Army Med. Dep. J. 2013;•••:24–35. [PMID: 24146240]. [PubMed] [Google Scholar]
  • 320.Cho M.S. Verification of the mediation effect of recovery resilience according to the relation between elderly users’ participation in exercise rehabilitation program and their successful aging. J. Exerc. Rehabil. 2014;10(5):319–325. doi: 10.12965/jer.140164. [http://dx.doi.org/10.12965/jer. 140164]. [PMID: 25426471]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Fleg J.L. Aerobic exercise in the elderly: a key to successful aging. Discov. Med. 2012;13(70):223–228. [PMID: 22463798]. [PubMed] [Google Scholar]
  • 322.Resnick B.A., Inguito P.L. The Resilience Scale: psychometric properties and clinical applicability in older adults. Arch. Psychiatr. Nurs. 2011;25(1):11–20. doi: 10.1016/j.apnu.2010.05.001. [http://dx.doi.org/10.1016/j.apnu.2010. 05.001]. [PMID: 21251597]. [DOI] [PubMed] [Google Scholar]
  • 323.Rantanen T., Masaki K., He Q., Ross G.W., Willcox B.J., White L. Midlife muscle strength and human longevity up to age 100 years: a 44-year prospective study among a decedent cohort. Age (Dordr.) 2012;34(3):563–570. doi: 10.1007/s11357-011-9256-y. [http://dx.doi.org/10.1007/ s11357-011-9256-y]. [PMID: 21541735]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Roh J., Rhee J., Chaudhari V., Rosenzweig A. The role of exercise in cardiac aging: from physiology to molecular mechanisms. Circ. Res. 2016;118(2):279–295. doi: 10.1161/CIRCRESAHA.115.305250. [http://dx.doi.org/10.1161/ CIRCRESAHA.115.305250]. [PMID: 26838314]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Saxena S., Jané-Llopis E., Hosman C. Prevention of mental and behavioural disorders: implications for policy and practice. World Psychiatry. 2006;5(1):5–14. [PMID: 16757984]. [PMC free article] [PubMed] [Google Scholar]
  • 326.Harmell A.L., Jeste D., Depp C. Strategies for successful aging: a research update. Curr. Psychiatry Rep. 2014;16(10):476. doi: 10.1007/s11920-014-0476-6. [http://dx.doi.org/10.1007/s11920-014-0476-6]. [PMID: 25135776]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Garatachea N., Pareja-Galeano H., Sanchis-Gomar F., Santos-Lozano A., Fiuza-Luces C., Morán M., Emanuele E., Joyner M.J., Lucia A. Exercise attenuates the major hallmarks of aging. Rejuvenation Res. 2015;18(1):57–89. doi: 10.1089/rej.2014.1623. [http://dx.doi.org/10. 1089/rej.2014.1623]. [PMID: 25431878]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Sallam N., Laher I. Exercise modulates oxidative stress and inflammation in aging and cardiovascular diseases. Oxid. Med. Cell. Longev. 2016;2016:7239639. doi: 10.1155/2016/7239639. [http://dx.doi.org/10.1155/ 2016/7239639]. [PMID: 26823952]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Mercken E.M., Carboneau B.A., Krzysik-Walker S.M., de Cabo R. Of mice and men: the benefits of caloric restriction, exercise, and mimetics. Ageing Res. Rev. 2012;11(3):390–398. doi: 10.1016/j.arr.2011.11.005. [http://dx. doi.org/10.1016/j.arr.2011.11.005]. [PMID: 22210414]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Weindruch R. The retardation of aging by caloric restriction: studies in rodents and primates. Toxicol. Pathol. 1996;24(6):742–745. doi: 10.1177/019262339602400618. [http://dx.doi.org/10.1177/019262339602400618]. [PMID: 8994305]. [DOI] [PubMed] [Google Scholar]
  • 331.Masoro E.J. Dietary restriction and aging. J. Am. Geriatr. Soc. 1993;41(9):994–999. doi: 10.1111/j.1532-5415.1993.tb06767.x. [http://dx.doi.org/10.1111/j.1532-5415.1993. tb06767.x]. [PMID: 8409187]. [DOI] [PubMed] [Google Scholar]
  • 332.Barger J.L., Walford R.L., Weindruch R. The retardation of aging by caloric restriction: its significance in the transgenic era. Exp. Gerontol. 2003;38(11-12):1343–1351. doi: 10.1016/j.exger.2003.10.017. [http://dx.doi.org/10. 1016/j.exger.2003.10.017]. [PMID: 14698815]. [DOI] [PubMed] [Google Scholar]
  • 333.Hauck S.J., Aaron J.M., Wright C., Kopchick J.J., Bartke A. Antioxidant enzymes, free-radical damage, and response to paraquat in liver and kidney of long-living growth hormone receptor/binding protein gene-disrupted mice. Horm. Metab. Res. 2002;34(9):481–486. doi: 10.1055/s-2002-34787. [http://dx.doi.org/10.1055/s-2002-34787]. [PMID: 12384824]. [DOI] [PubMed] [Google Scholar]
  • 334.Harper J.M., Salmon A.B., Chang Y., Bonkowski M., Bartke A., Miller R.A. Stress resistance and aging: influence of genes and nutrition. Mech. Ageing Dev. 2006;127(8):687–694. doi: 10.1016/j.mad.2006.04.002. [http://dx. doi.org/10.1016/j.mad.2006.04.002]. [PMID: 16713617]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Yu B.P. Why calorie restriction would work for human longevity. Biogerontology. 2006;7(3):179–182. doi: 10.1007/s10522-006-9009-y. [http://dx.doi.org/10.1007/ s10522-006-9009-y]. [PMID: 16676136]. [DOI] [PubMed] [Google Scholar]
  • 336.Hussin N.M., Shahar S., Teng N.I., Ngah W.Z., Das S.K. Efficacy of fasting and calorie restriction (FCR) on mood and depression among ageing men. J. Nutr. Health Aging. 2013;17(8):674–680. doi: 10.1007/s12603-013-0344-9. [http://dx.doi.org/10.1007/s12603-013-0344-9]. [PMID: 24097021]. [DOI] [PubMed] [Google Scholar]
  • 337.Teng N.I., Shahar S., Manaf Z.A., Das S.K., Taha C.S., Ngah W.Z. Efficacy of fasting calorie restriction on quality of life among aging men. Physiol. Behav. 2011;104(5):1059–1064. doi: 10.1016/j.physbeh.2011.07.007. [http://dx. doi.org/10.1016/j.physbeh.2011.07.007]. [PMID: 21781980]. [DOI] [PubMed] [Google Scholar]
  • 338.Rea S.L., Wu D., Cypser J.R., Vaupel J.W., Johnson T.E. A stress-sensitive reporter predicts longevity in isogenic populations of Caenorhabditis elegans. Nat. Genet. 2005;37(8):894–898. doi: 10.1038/ng1608. [http://dx.doi.org/10.1038/ng1608]. [PMID: 16041374]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Harper J.M., Salmon A.B., Leiser S.F., Galecki A.T., Miller R.A. Skin-derived fibroblasts from long-lived species are resistant to some, but not all, lethal stresses and to the mitochondrial inhibitor rotenone. Aging Cell. 2007;6(1):1–13. doi: 10.1111/j.1474-9726.2006.00255.x. [http://dx.doi.org/ 10.1111/j.1474-9726.2006.00255.x]. [PMID: 17156084]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Kapahi P., Boulton M.E., Kirkwood T.B. Positive correlation between mammalian life span and cellular resistance to stress. Free Radic. Biol. Med. 1999;26(5-6):495–500. doi: 10.1016/s0891-5849(98)00323-2. [http://dx.doi.org/10. 1016/S0891-5849(98)00323-2]. [PMID: 10218637]. [DOI] [PubMed] [Google Scholar]
  • 341.Anisimov V.N., Zabezhinski M.A., Popovich I.G., Piskunova T.S., Semenchenko A.V., Tyndyk M.L., Yurova M.N., Rosenfeld S.V., Blagosklonny M.V. Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice. Cell Cycle. 2011;10(24):4230–4236. doi: 10.4161/cc.10.24.18486. [http://dx.doi.org/10.4161/cc.10.24. 18486]. [PMID: 22107964]. [DOI] [PubMed] [Google Scholar]
  • 342.Spong A., Bartke A. Rapamycin slows aging in mice. Cell Cycle. 2012;11(5):845. doi: 10.4161/cc.11.5.19607. [http://dx.doi.org/10.4161/cc.11.5.19607]. [PMID: 22356747]. [DOI] [PubMed] [Google Scholar]
  • 343.Hurez V., Dao V., Liu A., Pandeswara S., Gelfond J., Sun L., Bergman M., Orihuela C.J., Galvan V., Padrón Á., Drerup J., Liu Y., Hasty P., Sharp Z.D., Curiel T.J. Chronic mTOR inhibition in mice with rapamycin alters T, B, myeloid, and innate lymphoid cells and gut flora and prolongs life of immune-deficient mice. Aging Cell. 2015;14(6):945–956. doi: 10.1111/acel.12380. [http://dx.doi.org/ 10.1111/acel.12380]. [PMID: 26315673]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Kitada M., Koya D. The use of calorie restriction mimetics to study aging. Methods Mol. Biol. 2013;1048:95–107. doi: 10.1007/978-1-62703-556-9_8. [http://dx. doi.org/10.1007/978-1-62703-556-9_8]. [PMID: 23929100]. [DOI] [PubMed] [Google Scholar]
  • 345.Markus M.A., Morris B.J. Resveratrol in prevention and treatment of common clinical conditions of aging. Clin. Interv. Aging. 2008;3(2):331–339. [PMID: 18686754]. [PMC free article] [PubMed] [Google Scholar]
  • 346.Longo V.D., Antebi A., Bartke A., Barzilai N., Brown-Borg H.M., Caruso C., Curiel T.J., de Cabo R., Franceschi C., Gems D., Ingram D.K., Johnson T.E., Kennedy B.K., Kenyon C., Klein S., Kopchick J.J., Lepperdinger G., Madeo F., Mirisola M.G., Mitchell J.R., Passarino G., Rudolph K.L., Sedivy J.M., Shadel G.S., Sinclair D.A., Spindler S.R., Suh Y., Vijg J., Vinciguerra M., Fontana L. Interventions to slow aging in humans: are we ready? Aging Cell. 2015;14(4):497–510. doi: 10.1111/acel.12338. [http://dx.doi.org/ 10.1111/acel.12338]. [PMID: 25902704]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Hildon Z., Montgomery S.M., Blane D., Wiggins R.D., Netuveli G. Examining resilience of quality of life in the face of health-related and psychosocial adversity at older ages: what is “right” about the way we age? Gerontologist. 2010;50(1):36–47. doi: 10.1093/geront/gnp067. [http:// dx.doi.org/10.1093/geront/gnp067]. [PMID: 19549715]. [DOI] [PubMed] [Google Scholar]
  • 348.Tugade M.M., Fredrickson B.L. Resilient individuals use positive emotions to bounce back from negative emotional experiences. J. Pers. Soc. Psychol. 2004;86(2):320–333. doi: 10.1037/0022-3514.86.2.320. [http://dx.doi.org/10. 1037/0022-3514.86.2.320]. [PMID: 14769087]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Windle G., Bennett K.M., Noyes J. A methodological review of resilience measurement scales. Health Qual. Life Outcomes. 2011;9:8. doi: 10.1186/1477-7525-9-8. [http://dx.doi.org/10.1186/1477-7525-9-8]. [PMID: 21294858]. [DOI] [PMC free article] [PubMed] [Google Scholar]

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

RESOURCES