Skip to main content
Neural Plasticity logoLink to Neural Plasticity
. 2016 Jan 5;2016:4928081. doi: 10.1155/2016/4928081

Seeding Stress Resilience through Inoculation

Archana Ashokan 1, Meenalochani Sivasubramanian 1, Rupshi Mitra 1,*
PMCID: PMC4736400  PMID: 26881112

Abstract

Stress is a generalized set of physiological and psychological responses observed when an organism is placed under challenging circumstances. The stress response allows organisms to reattain the equilibrium in face of perturbations. Unfortunately, chronic and/or traumatic exposure to stress frequently overwhelms coping ability of an individual. This is manifested as symptoms affecting emotions and cognition in stress-related mental disorders. Thus environmental interventions that promote resilience in face of stress have much clinical relevance. Focus of the bulk of relevant neurobiological research at present remains on negative aspects of health and psychological outcomes of stress exposure. Yet exposure to the stress itself can promote resilience to subsequent stressful episodes later in the life. This is especially true if the prior stress occurs early in life, is mild in its magnitude, and is controllable by the individual. This articulation has been referred to as “stress inoculation,” reminiscent of resilience to the pathology generated through vaccination by attenuated pathogen itself. Using experimental evidence from animal models, this review explores relationship between nature of the “inoculum” stress and subsequent psychological resilience.

1. Stress and Stress Inoculation

Stress is a nonspecific response of the body to any demand placed by external environment or internal metabolic milieu [1]. The stress response itself cannot be eradicated or evaded for very long because these challenges are inevitable aspects of life. The concept of stress was first introduced in the work of Selye [1], who observed that individual animals exhibited a “general adaptation syndrome” when confronted with a variety of perturbations. He argued for a common bodily mechanism that was invoked during these challenging episodes and termed it “stress,” further remarking that the stress “suffers from the mixed blessing of being too well known and too little understood” [1]. In due course of time psychological dimensions were included in the repertoire of stress syndrome, including emotional and cognitive facets. In recent decades, relationship between stress and predisposition to mental disorders has gained mounting importance [2]. This renewed focus is borne out by reclassification of stress and trauma related disorders as an identifiable mental disorder in recent diagnostic manual [3]. The clinical interest is paralleled by increasing knowledge about neural and endocrine underpinnings of this process. In this regard, feedback loops between adrenal hormones and discrete brain regions have been extensively described and analysed (succinctly reviewed in [47]).

Despite its negative connotations, stress has an adaptive value in that it promotes homeostasis. Exposure to stressful events that are not devastating, yet challenging enough to provoke emotional instigation and cognitive processing, might nurture successful coping with subsequent stressors. Thus exposure to prolonged unpredictable and uncontrollable stress induces long-term neurological impairment, but exposure to moderately stressful and controllable events seems to increase efficacy of regulating future stress response. This phenomenon has been referred to as “stress inoculation” [8]. Rhetorically, stress inoculation is reminiscent of protection from a pathology afforded by prior inoculation with attenuated pathological agent. Just as vaccination by a dead or weakened pathogen enables the body to mount a long-lasting immune response, exposure to moderate amount of stress enables organisms to effectively cope with future stressors. Although this hypothesis has great potential, it remains relatively understudied at present.

The effects of stress on health outcomes often vary in a nonlinear manner with severity of the stress, roughly corresponding to an inverted U shaped reaction norm [911]. When starting from a low baseline, moderate amount of stressful challenge enhances both short-term and long-term health outcomes. With successive increase in severity, stress leads to diminished health leading to an inverted U relationship between stress and health parameters. The idea of inverted U shaped reaction norm was first formalized to explain relationship between arousal levels and strength of discrimination based learning [12]. The concept has been subsequently used to empirically explain variation in behaviour with respect to different kinds and strengths of stressor. A few studies have also demonstrated this relationship within a single experimental design. For example, rats swimming in colder water experience greater secretion of stress hormones (16°C > 19°C > 25°C) when undergoing training for spatial learning in radial arm water maze. Animals trained at moderate stress levels of 19°C perform fewer errors compared to those trained at either 16°C or 25°C [13]. Similarly direction of the corticosterone influence on hippocampal primed burst potentiation is dependent upon the concentration of this stress hormone [14]. Briefly, primed burst potentiation utilizes electrical stimulation that mimics pattern of endogenous activity of hippocampal neurons, leading to long-lasting increase in synaptic strength [15]. Low to medium corticosterone levels are positively correlated with primed burst potentiation, representing ascending part of the inverse U. At higher concentration, corticosterone levels become negatively correlated with electrophysiological potentiation, reflecting descending part of the curve.

Various factors influence the shape and inflexion point of such inverted U curve. These include age, sex, and predictability of the stress [1619]. Very importantly, these factors also include degree, nature, and developmental timing of the historical stress exposure [20]. These factors are capable of shifting the curve from its general course towards either stress pathologies or stress resilience [9]. The essence of stress inoculation thus lies in the idea that prior stress exposure can induce resilience to later stress, if the prior stress is of optimal degree and provided at crucial stage of life.

2. State of Current Animal Models

Animals models are primarily used for stress research due to the methodological and ethical limitations involved in human studies [21]. A good animal model of stress inoculation will ideally comprise face validity, construct validity, and predictive validity (e.g., [22, 23]). In other words, an animal model of stress inoculation must contain elements that are analogous to human stress inoculation. It must comprise measurable endpoint that accurately reflects the unmeasurable theoretical construct of stress inoculation. Additionally, animal model in question must be able to prospectively predict strength of the inoculation. Beyond these, the animal models should be consistent and reproducible and have internal controls to measure influence of confound like locomotion or nutrition. Unfortunately, a consensus about the animal model that meets these criteria for stress inoculation remains elusive at present. A variety of inoculum stressors and correspondingly varied subsequent stressors to test the inoculation have been used. Further work is required to refine animal models with respect to both external and internal validity.

Within the limits of current animal models, researches on rodents and primates support the stress inoculation hypothesis and provide insight into its neurobiological mechanisms. Broadly, early life stress inoculation triggers broad developmental cascades that increase adaptation. For example, studies on male and female squirrel monkeys show that a brief intermittent maternal separation during early childhood enhances long-lasting and trait-like transformation in the multiple domains of adaptive functioning [24]. Young male and female monkeys presented with a moderate stressor in the form of periodic short maternal separation from postnatal week 17 to postnatal week 27 experienced acute distress during the separation periods manifested by agitation and temporary elevation in the stress hormone levels [25]. However later in life, at nine months of age, the same set of monkeys demonstrated lower anxiety and decreased stress hormone levels when compared to the control animals. Further these inoculated monkeys showed higher cognitive control when accessed at 1.5 years of age, higher curiosity when accessed at 2.5 years, and larger prefrontal cortex volume at 3.3 years of age, compared to the age matched noninoculated controls [25, 26]. These results suggest that engagement in new situations that require challenging but not overwhelmingly stressful experiences results in enduring effects that stimulate adaptation in cognitive, motivational, and socioemotional aspect of behaviour in primates.

Early developmental stages of an individual asymmetrically contribute to the shaping of resilience in later life. Several studies have demonstrated entrainment of adult behaviour as a consequence of stress during early development. For example, squirrels change their growth trajectories based on in utero exposure to stress [27], and early childhood stress results in earlier menarche in human females [28] and aversive conditioning during infancy blunts the strength of further conditioning in adulthood [29]. Congruently, majority of studies pertaining to stress inoculation provide initial stress in early life [3034]. For instance, maternal separation in infant rodents since birth (continuously for 2 weeks, 3 hours per day) gives rise to hyperactive stress responses in the form of heightened stress hormone release [34]. However, intermittent brief separation of these pups from the mother results in adaptive endocrine responses characterizing resilient features [32]. Similarly in primates, when four-month-old squirrel monkeys are exposed to intermittent levels of the same form of maternal separation (ten sessions per week), it leads to emotionally stable responses under stressful situations and lowered release of stress hormones accompanied by more exploration of novel settings [24]. This presents an exemplary case of stress inoculation which is dependent on the developmental stage of an individual.

In should be noted that resilience in models involving maternal separation could result from behavioural change in either mother or the offspring. In other words, it is possible that early life stress promotes resilience because the separation changes maternal behaviour towards pups rather than stress inoculation of the pups themselves. For example, brief intermittent exposure to foot shock during infancy augments resilience. This is due to the increased maternal stimulation received after the rat pups are returned to their nest. This increased maternal stimulation has been shown to enhance stress regulation in pups that endure into their adulthood [30]. In contrast to rats, similar effects have not been observed in primates. For example, differences in the maternal behaviour did not correspond with differences in the development of arousal regulation in young monkeys [33]. Thus both the locus of initial behavioural change and the outcome can be idiosyncratically specific to the species being studied. This creates further challenge to create an animal model for studying stress inoculation in humans.

While early life has a long-lasting influence on the stress inoculation, several papers have also reported protective effects of preceding stress in adulthood. Thus stress inoculation in male mice by exposure to mild stress (noncontact interaction via resident intrusion) in adulthood leads to more emotionally stable response, lowered depressive like symptoms, and enhanced exploratory behaviour [35]. The same study also showed reduced secretion of stress hormones in response to repeated restraint. Similarly, exposure to three or more mild restraints before inescapable shock or three sessions of inescapable tail-shocks with intervening rest days attenuates development of learned helplessness in male rats [36]. In adult female squirrel monkeys, intermittent separation from group and introduction of novel group partners create a stress inoculation against future stressor [37]. This manifests as reduction in anhedonia and reduced activation of stress hormone axis when inoculated animals are exposed to a subsequent social separation. This generality of inoculation models across developmental stages, if reinforced by further studies, creates greater opportunity for use of this paradigm in the adulthood.

Gender presents an important consideration when interpreting effects of stress inoculation. Sexually dimorphic gonadal hormones robustly interact with brain and behaviour, including the stress response (reviewed in [3841]). For example, major depression and anxiety disorder are more prevalent in women of reproductive age than corresponding male population [42]. In rats, chronic stress causes lesser anxiogenesis in females compared to males [4346]. Biological substrates of gender dimorphism pertaining to stress remain understudied. Similarly, reasons for discordant direction of stress effects on humans and rodents are unclear at present.

3. Environmental Manipulation: A Potential Regulator for Hypothalamus-Pituitary-Adrenal Axis Tone

The degree of control that an animal has on a specific stressor plays a key role in defining whether the event will lead to ensuing vulnerability or resilience to the stress. Animals administered with unavoidable and unpredictable shock tend to develop exaggerated fear response, heightened anxiety, and deficits in active coping when faced with subsequent stressor [47], a phenomenon often referred to as learned helplessness [48]. However, animals that are given shock and are concomitantly given the ability to avoid them by modifying their behaviour do not develop learned helplessness [49, 50]. Similar effects have been observed in humans, whereby individuals previously inoculated by a controllable stress acquire resilience to a broader range of other subsequent stressors [51].

In terms of the endocrine activation, stress inoculation results in lower responsiveness and earlier termination of stress hormone secretion, while severe stress results in the opposite effect. For example, repeated maternal separation of rat pups results in lower expression of glucocorticoid receptors (GRs) in the hippocampus when these pups reach adulthood [52]. The hippocampal glucocorticoid receptors bind to circulating corticosterone (CORT), a stress hormone secreted by adrenal glands. The occupancy of these GRs then sends a negative feedback to the hypothalamus-pituitary-adrenal axis, thus terminating further stress hormone release. A reduction in glucocorticoid receptors leads to reduced efficacy of this negative feedback which then blunts the ability to terminate ongoing stress response. This example demonstrates that early environment can have long-lasting implications for future stress response.

CORT, the primary ligand for GR, is often measured to reflect ongoing stress response [53, 54]. Amount of circulating CORT in rodents (cortisol in primates) exhibits robust sensitivity to the environment [55]. For example, introduction of novelty in the environment causes increase in CORT, leading to an emotional arousal in the individual [56]. Modulations in the environment on an intermittent basis might lead to increase or decrease of CORT and when this is done in moderation, it can cause a shift in the threshold of the HPA activity for an individual [55, 57]. The resultant effect would reflect the shift in the inverted U shaped curve of stress response which corresponds to stress inoculation [9]. In brief, stress inoculation can shift dose response curve between future stress and performance leftward (greater performance at lower levels of future stress) and/or rightward (greater tolerance to higher level of stress) [9].

Thus both stress response and CORT are exquisitely responsive to the degree and nature of environmental changes. Interestingly, CORT itself causes differential neural plasticity in different brain regions. For example, chronic stress which raises the circulating CORT leads to neuronal atrophy in the hippocampus but hypertrophy in the basolateral amygdala (BLA) [5860]. These contrasting effects are suggested to cause reduced memory performance due to chronic stress mediated by its hippocampal effects and increased anxiety mediated by its amygdalar effects. The dorsal region of the hippocampus is necessary for spatial learning and is directly linked to stress-induced memory deficits [61, 62]. Likewise, intra-BLA experimental manipulation suggests necessity and sufficiency of BLA changes for stress-induced anxiogenesis. For example, decreasing excitability of BLA neurons by overexpression of SK2 K+ channels simultaneously reduces stress-induced stress hormone secretion, anxiety, and BLA hypertrophy [63]. Similarly, rerouting of stress hormone signalling away from glucocorticoid receptors within BLA also reduces anxiety [6466].

Several studies described above have used CORT levels as a proxy for HPA tone and implicitly as a proxy for stress responsiveness. Yet the relationship between CORT levels and HPA responsiveness is not linear. For example, effects of CORT depend on expression of GR and relative expression of GR/MR, in addition to specific brain regions expressing these receptors. It is noteworthy that an experimental change in ratio between MR and GR expression in the hippocampus can drastically change memory [67, 68]. Similar manipulation in the amygdala reduces anxiety and future endogenous CORT release [64]. This suggests that effects of CORT on brain and behaviour are dependent on expression level of receptors and type of the central receptors (GR and MR) available. The importance of the central receptors is further supported by the observations that stress in rodents and monkeys can downregulate hippocampal GRs and thus is secondarily leading to loss of negative feedback of CORT secretion [69, 70]. In this context, it is interesting that hippocampus and amygdala exhibit differential expression of GR and MR [52, 71].

Cognitive decline associated with hippocampus has been extensively studied in respect to effects of stress (reviewed in [60, 72, 73]). However BLA, which is critical for generation and maintenance of fear and anxiety [74], has been relatively understudied in this regard [75, 76]. Future studies to bridge this gap will hopefully bring more clarity to biological mechanisms of stress inoculation.

Acknowledgments

The authors were supported by Ministry of Education, Singapore. The authors thank Dr. Ajai Vyas for assistance in editing the final draft.

Conflict of Interests

The authors declare that there is no conflict of interests regarding the publication of this paper.

References

  • 1.Selye H. The evolution of the stress concept: the originator of the concept traces its development from the discovery in 1936 of the alarm reaction to modern therapeutic applications of syntoxic and catatoxic hormones. American Scientist. 1973;61(6):692–699. [PubMed] [Google Scholar]
  • 2.Karatsoreos I. N., McEwen B. S. Psychobiological allostasis: resistance, resilience and vulnerability. Trends in Cognitive Sciences. 2011;15(12):576–584. doi: 10.1016/j.tics.2011.10.005. [DOI] [PubMed] [Google Scholar]
  • 3.Association A. P. Diagnostic and Statistical Manual of Mental Disorders: DSM-5. ManMag; 2003. [Google Scholar]
  • 4.Hunter R. G., McEwen B. S. Stress and anxiety across the lifespan: structural plasticity and epigenetic regulation. Epigenomics. 2013;5(2):177–194. doi: 10.2217/epi.13.8. [DOI] [PubMed] [Google Scholar]
  • 5.McEwen B. S. Brain on stress: how the social environment gets under the skin. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(supplement 2):17180–17185. doi: 10.1073/pnas.1121254109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.McEwen B. S., Getz L. Lifetime experiences, the brain and personalized medicine: an integrative perspective. Metabolism. 2013;62(supplement 1):S20–S26. doi: 10.1016/j.metabol.2012.08.020. [DOI] [PubMed] [Google Scholar]
  • 7.Popoli M., Yan Z., McEwen B. S., Sanacora G. The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nature Reviews Neuroscience. 2012;13(1):22–37. doi: 10.1038/nrn3138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Meichenbaum D., Cameron R. Stress inoculation training. In: Meichenbaum D., Jaremko M., editors. Stress Reduction and Prevention. New York, NY, USA: Springer; 1989. pp. 115–154. [Google Scholar]
  • 9.Russo S. J., Murrough J. W., Han M.-H., Charney D. S., Nestler E. J. Neurobiology of resilience. Nature Neuroscience. 2012;15(11):1475–1484. doi: 10.1038/nn.3234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sapolsky R. M. Stress and the brain: individual variability and the inverted-U. Nature Neuroscience. 2015;18(10):1344–1346. doi: 10.1038/nn.4109. [DOI] [PubMed] [Google Scholar]
  • 11.Mendl M. Performing under pressure: stress and cognitive function. Applied Animal Behaviour Science. 1999;65(3):221–244. doi: 10.1016/s0168-1591(99)00088-x. [DOI] [Google Scholar]
  • 12.Yerkes R. M., Dodson J. D. The relation of strength of stimulus to rapidity of habit-formation. Journal of Comparative Neurology and Psychology. 1908;18(5):459–482. doi: 10.1002/cne.920180503. [DOI] [Google Scholar]
  • 13.Salehi B., Cordero M. I., Sandi C. Learning under stress: the inverted-U-shape function revisited. Learning and Memory. 2010;17(10):522–530. doi: 10.1101/lm.1914110. [DOI] [PubMed] [Google Scholar]
  • 14.Diamond D. M., Bennett M. C., Fleshner M., Rose G. M. Inverted-U relationship between the level of peripheral corticosterone and the magnitude of hippocampal primed burst potentiation. Hippocampus. 1992;2(4):421–430. doi: 10.1002/hipo.450020409. [DOI] [PubMed] [Google Scholar]
  • 15.Rose G., Diamond D. M., Pang K., Dunwiddie T. V. Synaptic Plasticity in the Hippocampus. Berlin, Germany: Springer; 1988. Primed burst potentiation: lasting synaptic plasticity invoked by physiologically patterned stimulation; pp. 96–98. [DOI] [Google Scholar]
  • 16.Lupien S. J., McEwen B. S., Gunnar M. R., Heim C. Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nature Reviews Neuroscience. 2009;10(6):434–445. doi: 10.1038/nrn2639. [DOI] [PubMed] [Google Scholar]
  • 17.Luine V. Sex differences in chronic stress effects on memory in rats. Stress. 2002;5(3):205–216. doi: 10.1080/1025389021000010549. [DOI] [PubMed] [Google Scholar]
  • 18.Martins A. Q., McIntyre D., Ring C. Aversive event unpredictability causes stress-induced hypoalgesia. Psychophysiology. 2015;52(8):1066–1070. doi: 10.1111/psyp.12427. [DOI] [PubMed] [Google Scholar]
  • 19.Andersen S. L. Exposure to early adversity: points of cross-species translation that can lead to improved understanding of depression. Development and Psychopathology. 2015;27(2):477–491. doi: 10.1017/s0954579415000103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lyons D. M., Parker K. J., Schatzberg A. F. Animal models of early life stress: implications for understanding resilience. Developmental Psychobiology. 2010;52(7):616–624. doi: 10.1002/dev.20500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Campos A. C., Fogaça M. V., Aguiar D. C., Guimarães F. S. Animal models of anxiety disorders and stress. Revista Brasileira de Psiquiatria. 2013;35(supplement 2):S101–S111. doi: 10.1590/1516-4446-2013-1139. [DOI] [PubMed] [Google Scholar]
  • 22.Belzung C., Lemoine M. Criteria of validity for animal models of psychiatric disorders: focus on anxiety disorders and depression. Biology of Mood & Anxiety Disorders. 2011;1(1):p. 9. doi: 10.1186/2045-5380-1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Walf A. A., Frye C. A. The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nature Protocols. 2007;2(2):322–328. doi: 10.1038/nprot.2007.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Parker K. J., Buckmaster C. L., Schatzberg A. F., Lyons D. M. Prospective investigation of stress inoculation in young monkeys. Archives of General Psychiatry. 2004;61(9):933–941. doi: 10.1001/archpsyc.61.9.933. [DOI] [PubMed] [Google Scholar]
  • 25.Parker K. J., Buckmaster C. L., Justus K. R., Schatzberg A. F., Lyons D. M. Mild early life stress enhances prefrontal-dependent response inhibition in monkeys. Biological Psychiatry. 2005;57(8):848–855. doi: 10.1016/j.biopsych.2004.12.024. [DOI] [PubMed] [Google Scholar]
  • 26.Lyons D. M., Parker K. J. Stress inoculation-induced indications of resilience in monkeys. Journal of Traumatic Stress. 2007;20(4):423–433. doi: 10.1002/jts.20265. [DOI] [PubMed] [Google Scholar]
  • 27.Dantzer B., Newman A. E. M., Boonstra R., et al. Density triggers maternal hormones that increase adaptive offspring growth in a wild mammal. Science. 2013;340(6137):1215–1217. doi: 10.1126/science.1235765. [DOI] [PubMed] [Google Scholar]
  • 28.Chisholm J. S., Quinlivan J. A., Petersen R. W., Coall D. A. Early stress predicts age at menarche and first birth, adult attachment, and expected lifespan. Human Nature. 2005;16(3):233–265. doi: 10.1007/s12110-005-1009-0. [DOI] [PubMed] [Google Scholar]
  • 29.Sevelinges Y., Moriceau S., Holman P., et al. Enduring effects of infant memories: infant odor-shock conditioning attenuates amygdala activity and adult fear conditioning. Biological Psychiatry. 2007;62(10):1070–1079. doi: 10.1016/j.biopsych.2007.04.025. [DOI] [PubMed] [Google Scholar]
  • 30.Cameron N. M., Champagne F. A., Parent C., Fish E. W., Ozaki-Kuroda K., Meaney M. J. The programming of individual differences in defensive responses and reproductive strategies in the rat through variations in maternal care. Neuroscience and Biobehavioral Reviews. 2005;29(4-5):843–865. doi: 10.1016/j.neubiorev.2005.03.022. [DOI] [PubMed] [Google Scholar]
  • 31.Coe C. L., Glass J. C., Wiener S. G., Levine S. Behavioral, but not physiological, adaptation to repeated separation in mother and infant primates. Psychoneuroendocrinology. 1983;8(4):401–409. doi: 10.1016/0306-4530(83)90019-7. [DOI] [PubMed] [Google Scholar]
  • 32.Denenberg V. H. Commentary: is maternal stimulation the mediator of the handling effect in infancy? Developmental Psychobiology. 1999;34(1):1–3. doi: 10.1002/(sici)1098-2302(199901)34:160;1::aid-dev262;3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  • 33.Jordan T. C., Hennessy M. B., Gonzalez C. A., Levine S. Social and environmental factors influencing mother-infant separation-reunion in squirrel monkeys. Physiology and Behavior. 1985;34(4):489–493. doi: 10.1016/0031-9384(85)90038-1. [DOI] [PubMed] [Google Scholar]
  • 34.Plotsky P. M., Meaney M. J. Early, postnatal experience alters hypothalamic corticotropin-releasing factor (CRF) mRNA, median eminence CRF content and stress-induced release in adult rats. Molecular Brain Research. 1993;18(3):195–200. doi: 10.1016/0169-328X(93)90189-V. [DOI] [PubMed] [Google Scholar]
  • 35.Brockhurst J., Cheleuitte-Nieves C., Buckmaster C. L., Schatzberg A. F., Lyons D. M. Stress inoculation modeled in mice. Translational Psychiatry. 2015;5(3, article e537) doi: 10.1038/tp.2015.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Plumb T. N., Cullen P. K., Minor T. R. Parameters of hormetic stress and resilience to trauma in rats. Stress. 2015;18(1):88–95. doi: 10.3109/10253890.2014.974154. [DOI] [PubMed] [Google Scholar]
  • 37.Lee A. G., Buckmaster C. L., Yi E., Schatzberg A. F., Lyons D. M. Coping and glucocorticoid receptor regulation by stress inoculation. Psychoneuroendocrinology. 2014;49(1):272–279. doi: 10.1016/j.psyneuen.2014.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Curtis K. S., Krause E. G. Sex differences in physiology and behavior: focus on central actions of ovarian hormones. Physiology and Behavior. 2009;97(2):141–142. doi: 10.1016/j.physbeh.2009.02.026. [DOI] [PubMed] [Google Scholar]
  • 39.Kajantie E., Phillips D. I. W. The effects of sex and hormonal status on the physiological response to acute psychosocial stress. Psychoneuroendocrinology. 2006;31(2):151–178. doi: 10.1016/j.psyneuen.2005.07.002. [DOI] [PubMed] [Google Scholar]
  • 40.Shepard K. N., Michopoulos V., Toufexis D. J., Wilson M. E. Genetic, epigenetic and environmental impact on sex differences in social behavior. Physiology & Behavior. 2009;97(2):157–170. doi: 10.1016/j.physbeh.2009.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ter Horst G. J., Wichmann R., Gerrits M., Westenbroek C., Lin Y. Sex differences in stress responses: focus on ovarian hormones. Physiology and Behavior. 2009;97(2):239–249. doi: 10.1016/j.physbeh.2009.02.036. [DOI] [PubMed] [Google Scholar]
  • 42.Kessler R. C., McGonagle K. A., Swartz M., Blazer D. G., Nelson C. B. Sex and depression in the National Comorbidity Survey. I: lifetime prevalence, chronicity and recurrence. Journal of Affective Disorders. 1993;29(2-3):85–96. doi: 10.1016/0165-0327(93)90026-g. [DOI] [PubMed] [Google Scholar]
  • 43.Vyas A., Chattarji S. Modulation of different states of anxiety-like behavior by chronic stress. Behavioral Neuroscience. 2004;118(6):1450–1454. doi: 10.1037/0735-7044.118.6.1450. [DOI] [PubMed] [Google Scholar]
  • 44.Mitra R., Vyas A., Chatterjee G., Chattarji S. Chronic-stress induced modulation of different states of anxiety-like behavior in female rats. Neuroscience Letters. 2005;383(3):278–283. doi: 10.1016/j.neulet.2005.04.037. [DOI] [PubMed] [Google Scholar]
  • 45.Westenbroek C., Den Boer J. A., Ter Horst G. J. Gender-specific effects of social housing on chronic stress-induced limbic FOS expression. Neuroscience. 2003;121(1):189–199. doi: 10.1016/s0306-4522(03)00367-1. [DOI] [PubMed] [Google Scholar]
  • 46.Westenbroek C., Snijders T. A. B., Den Boer J. A., Gerrits M., Fokkema D. S., Ter Horst G. J. Pair-housing of male and female rats during chronic stress exposure results in gender-specific behavioral responses. Hormones and Behavior. 2005;47(5):620–628. doi: 10.1016/j.yhbeh.2005.01.004. [DOI] [PubMed] [Google Scholar]
  • 47.Krishnan V., Han M.-H., Graham D. L., et al. 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. [DOI] [PubMed] [Google Scholar]
  • 48.Fleshner M., Maier S. F., Lyons D. M., Raskind M. A. The neurobiology of the stress-resistant brain. Stress. 2011;14(5):498–502. doi: 10.3109/10253890.2011.596865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Greenwood B. N., Fleshner M. Exercise, learned helplessness, and the stress-resistant brain. NeuroMolecular Medicine. 2008;10(2):81–98. doi: 10.1007/s12017-008-8029-y. [DOI] [PubMed] [Google Scholar]
  • 50.Berton O., Covington H. E., III, Ebner K., et al. Induction of ΔFosB in the periaqueductal gray by stress promotes active coping responses. Neuron. 2007;55(2):289–300. doi: 10.1016/j.neuron.2007.06.033. [DOI] [PubMed] [Google Scholar]
  • 51.Kerr D. L., McLaren D. G., Mathy R. M., Nitschke J. B. Controllability modulates the anticipatory response in the human ventromedial prefrontal cortex. Frontiers in Psychology. 2012;3, article 557 doi: 10.3389/fpsyg.2012.00557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Joëls M., Sarabdjitsingh R. A., Karst H. Unraveling the time domains of corticosteroid hormone influences on brain activity: rapid, slow, and chronic modes. Pharmacological Reviews. 2012;64(4):901–938. doi: 10.1124/pr.112.005892. [DOI] [PubMed] [Google Scholar]
  • 53.Smith S. M., Vale W. W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues in Clinical Neuroscience. 2006;8(4):383–395. doi: 10.31887/DCNS.2006.8.4/ssmith. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Herman J. P., Cullinan W. E. Neurocircuitry of stress: central control of the hypothalamo-pituitary-adrenocortical axis. Trends in Neurosciences. 1997;20(2):78–84. doi: 10.1016/s0166-2236(96)10069-2. [DOI] [PubMed] [Google Scholar]
  • 55.Crofton E. J., Zhang Y., Green T. A. Inoculation stress hypothesis of environmental enrichment. Neuroscience and Biobehavioral Reviews. 2015;49:19–31. doi: 10.1016/j.neubiorev.2014.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Okuda S., Roozendaal B., McGaugh J. L. Glucocorticoid effects on object recognition memory require training-associated emotional arousal. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(3):853–858. doi: 10.1073/pnas.0307803100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.McEwen B. S., Gianaros P. J. Stress- and allostasis-induced brain plasticity. Annual Review of Medicine. 2011;62:431–445. doi: 10.1146/annurev-med-052209-100430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mitra R., Jadhav S., McEwen B. S., Vyas A., Chattarji S. Stress duration modulates the spatiotemporal patterns of spine formation in the basolateral amygdala. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(26):9371–9376. doi: 10.1073/pnas.0504011102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Vyas A., Mitra R., Shankaranarayana Rao B. S., Chattarji S. Chronic stress induces contrasting patterns of dendritic remodeling in hippocampal and amygdaloid neurons. Journal of Neuroscience. 2002;22(15):6810–6818. doi: 10.1523/JNEUROSCI.22-15-06810.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.McEwen B. S. Stress and hippocampal plasticity. Annual Review of Neuroscience. 1999;22:105–122. doi: 10.1146/annurev.neuro.22.1.105. [DOI] [PubMed] [Google Scholar]
  • 61.Moser E., Moser M.-B., Andersen P. Spatial learning impairment parallels the magnitude of dorsal hippocampal lesions, but is hardly present following ventral lesions. The Journal of Neuroscience. 1993;13(9):3916–3925. doi: 10.1523/JNEUROSCI.13-09-03916.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Handelmann G. E., Olton D. S. Spatial memory following damage to hippocampal CA3 pyramidal cells with kainic acid: impairment and recovery with preoperative training. Brain Research. 1981;217(1):41–58. doi: 10.1016/0006-8993(81)90183-9. [DOI] [PubMed] [Google Scholar]
  • 63.Mitra R., Ferguson D., Sapolsky R. M. SK2 potassium channel overexpression in basolateral amygdala reduces anxiety, stress-induced corticosterone secretion and dendritic arborization. Molecular Psychiatry. 2009;14(9):847–855. doi: 10.1038/mp.2009.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mitra R., Ferguson D., Sapolsky R. M. Mineralocorticoid receptor overexpression in basolateral amygdala reduces corticosterone secretion and anxiety. Biological Psychiatry. 2009;66(7):686–690. doi: 10.1016/j.biopsych.2009.04.016. [DOI] [PubMed] [Google Scholar]
  • 65.Mitra R., Sapolsky R. M. Expression of chimeric estrogen-glucocorticoid-receptor in the amygdala reduces anxiety. Brain Research. 2010;1342:33–38. doi: 10.1016/j.brainres.2010.03.092. [DOI] [PubMed] [Google Scholar]
  • 66.Mitra R., Sapolsky R. M. Gene therapy in rodent amygdala against fear disorders. Expert Opinion on Biological Therapy. 2010;10(9):1289–1303. doi: 10.1517/14712598.2010.509341. [DOI] [PubMed] [Google Scholar]
  • 67.Ferguson D., Sapolsky R. Overexpression of mineralocorticoid and transdominant glucocorticoid receptor blocks the impairing effects of glucocorticoids on memory. Hippocampus. 2008;18(11):1103–1111. doi: 10.1002/hipo.20467. [DOI] [PubMed] [Google Scholar]
  • 68.Ferguson D., Sapolsky R. Mineralocorticoid receptor overexpression differentially modulates specific phases of spatial and nonspatial memory. The Journal of Neuroscience. 2007;27(30):8046–8052. doi: 10.1523/jneurosci.1187-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brooke S. M., De Haas-Johnson A. M., Kaplan J. R., Manuck S. B., Sapolsky R. M. Dexamethasone resistance among nonhuman primates associated with a selective decrease of glucocorticoid receptors in the hippocampus and a history of social instability. Neuroendocrinology. 1994;60(2):134–140. doi: 10.1159/000126743. [DOI] [PubMed] [Google Scholar]
  • 70.Mizoguchi K., Ishige A., Aburada M., Tabira T. Chronic stress attenuates glucocorticoid negative feedback: involvement of the prefrontal cortex and hippocampus. Neuroscience. 2003;119(3):887–897. doi: 10.1016/s0306-4522(03)00105-2. [DOI] [PubMed] [Google Scholar]
  • 71.Reul J. M. H. M., de Kloet E. R. Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation. Endocrinology. 1985;117(6):2505–2511. doi: 10.1210/endo-117-6-2505. [DOI] [PubMed] [Google Scholar]
  • 72.McEwen B. S. Plasticity of the hippocampus: adaptation to chronic stress and allostatic load. Annals of the New York Academy of Sciences. 2001;933:265–277. doi: 10.1111/j.1749-6632.2001.tb05830.x. [DOI] [PubMed] [Google Scholar]
  • 73.McEwen B. S. Sex, stress and the hippocampus: allostasis, allostatic load and the aging process. Neurobiology of Aging. 2002;23(5):921–939. doi: 10.1016/s0197-4580(02)00027-1. [DOI] [PubMed] [Google Scholar]
  • 74.Davis M., Rainnie D., Cassell M. Neurotransmission in the rat amygdala related to fear and anxiety. Trends in Neurosciences. 1994;17(5):208–214. doi: 10.1016/0166-2236(94)90106-6. [DOI] [PubMed] [Google Scholar]
  • 75.Boyle L. M. A neuroplasticity hypothesis of chronic stress in the basolateral amygdala. Yale Journal of Biology and Medicine. 2013;86(2):117–125. [PMC free article] [PubMed] [Google Scholar]
  • 76.McEwen B. S., Nasca C., Gray J. D. Stress effects on neuronal structure: hippocampus, amygdala, and prefrontal cortex. Neuropsychopharmacology. 2015 doi: 10.1038/npp.2015.171. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neural Plasticity are provided here courtesy of Wiley

RESOURCES