You are on page 1of 12

Behavioural Brain Research 412 (2021) 113410

Contents lists available at ScienceDirect

Behavioural Brain Research


journal homepage: www.elsevier.com/locate/bbr

Stress, memory, and implications for major depression


Geovan Menezes de Sousa Júnior a, b, Hector David Quinones Vargas a, b,
Flávio Freitas Barbosa c, d, Nicole Leite Galvão-Coelho a, b, e, *
a
Laboratory of Hormone Measures, Department of Physiology & Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
b
Postgraduate Program of Psychobiology, Center for Biosciences, Federal University of Rio Grande do Norte, Natal, Brazil
c
Laboratory of Studies on Memory and Cognition, Department of Psychology, Federal University of Paraíba, João Pessoa, Brazil
d
Laboratory of Psychopharmacology, Federal University of Paraíba, João Pessoa, Brazil
e
National Science and Technology Institute for Translational Medicine, São Paulo, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: The stress response comprises a phylogenetically conserved set of cognitive, physiological, and behavioral re­
Memory systems sponses that evolved as a survival strategy. In this context, the memory of stressful events would be adaptive as it
Stress could avoid re-exposure to an adverse event, otherwise the event would be facilitated in positively stressful or
Depression
non-distressful conditions. However, the interaction between stress and memory comprises complex responses,
Cortisol
Catecholamines
some of them which are not yet completely understood, and which depend on several factors such as the memory
system that is recruited, the nature and duration of the stressful event, as well as the timing in which this
interaction takes place. In this narrative review, we briefly discuss the mechanisms of the stress response, the
main memory systems, and its neural correlates. Then, we show how stress, through the action of its biochemical
mediators, influences memory systems and mnemonic processes. Finally, we make use of major depressive
disorder to explore the possible implications of non-adaptive interactions between stress and memory to psy­
chiatric disorders, as well as possible roles for memory studies in the field of psychiatry.

1. Introduction memory formation, storage, and retrieval in several different ways,


depending on the duration and timing of stress, for instance, but also on
Both stress and memory are markedly present both in our phyloge­ the operational phase of memory [2]. Nevertheless, a long-lasting stress
netic history, as well as in our daily lives, guiding behavioral responses response can be harmful and may be involved in the development of
and aiding survival. These are complex physiological processes that psychopathological conditions that have an impact on memory pro­
depend on context, prior experiences, and the interaction between these. cesses, such as major depression [3]. Memory disruptions, primarily in
The stress response is mainly guided by the sympathoadrenalmedullary autobiographical and working memory, are a symptom of major
system and the hypothalamic-pituitary-adrenal axis, along with the depression commonly used for diagnosis that may remain as a residual
contribution of immune and metabolic systems. It promotes a physio­ symptom after remission [4,5]. In this narrative review, we aim to
logical, behavioral, and cognitive response which is highly adaptive [1]. outline the physiological basis of both the stress and memory systems,
The stress response influences the recruitment of memory systems, and to overview the main findings regarding their adaptive and

Abbreviations: ACTH, adrenocorticotropic hormone; AMPAR, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; BDNF, brain-derived neurotrophic
factor; BLA, basolateral amygdala; CA1,2,3, cornus amonis 1,2,3; CREB, cyclic AMP response element binding; CRH, corticotropin releasing hormone; DA, dopamine;
DG, dentate gyrus; dlCPF, dorsolateral prefrontal cortex; DNA, deoxyribonucleic acid; EC, entorhinal cortex; FKBP5, FK506 binding protein 51; GABA, γ-amino­
butyric acid; GC, glucocorticoid; GR, glucocorticoid receptor; GRE, glucocorticoid response element; HPA, hypothalamus-pituitary-adrenal axis; ilCPF, infralimbic
prefrontal cortex; LC, locus coeruleus; LTM, long-term memory; LTP, long-term potentiation; MR, mineralocorticoid receptor; NA, noradrenaline; NMDAR, N-methyl-
D-aspartate receptor; NTS, nucleus of the solitary tract; p75NTR, pan-neurotrophin receptor 75; PFC, prefrontal cortex; POMC, proopiomelanocortin; PVN, para­
ventricular nucleus of hypothalamus; sANS, sympathetic division of the autonomic nervous system; STM, short-term memory; TrkB, tropomyosin receptor kinase B;
TRD, treatment-resistant depression; vmCPF, ventromedial prefrontal cortex; VTA, ventral tegmental area; WM, working memory.
* Corresponding author at: Universidade Federal do Rio Grande do Norte, Departamento de Fisiologia & Comportamento, Caixa Postal, 1511, 59078-970, Natal,
RN, Brazil.
E-mail address: nicole.galvao@ufrn.br (N.L. Galvão-Coelho).

https://doi.org/10.1016/j.bbr.2021.113410
Received 12 October 2020; Received in revised form 4 June 2021; Accepted 6 June 2021
Available online 9 June 2021
0166-4328/© 2021 Elsevier B.V. All rights reserved.
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

disrupted interactions, considering the type and phase of memory, and strengthening [17,23] and the synthesis of neurotransmitters [17,24,25]
the duration of stress (acute or chronic). We also use major depressive in key areas for memory and other executive functions.
disorder to illustrate some of these maladaptive interactions. It is expected that the activation of the ANS and HPA axis promotes
an adaptive response that leads to the resolution of the stressor agent
2. Stress response while still in the resistance phase. Thus, the shutdown of the stress
response begins, by a cortisol-induced negative feedback on the HPA
When faced with a challenge or situation that disrupts homeostasis axis, specifically in the hypothalamus and anterior pituitary. Typically,
and/or threatens survival – that is, a stressor – a set of physiological, this negative feedback occurs through a slow genomic mechanism
behavioral and cognitive changes are triggered, in order to prepare the (delayed response). The intracellular hormone-receptor complex is
individual for an adequate performance regarding the resolution of the translocated from the cytoplasm to the nucleus, where it may, for
stressor and subsequently, reestablish homeostatic patterns [6,7]. The instance, decrease the transcription of crh and pomc genes, and increases
rapid response to stress, named by Hans Selye as the alarm phase, in­ the transcription of the fkbp5 gene. Fkbp5 regulates the transcription of
cludes the mobilization and targeting of energetic compounds to brain the FKBP5 protein (or FK506 binding protein 51), a class of chaperone
and skeletal muscles, which is essential to promote, respectively, that prevents the binding of GC to their receptors and, consequently, the
cognitive and physical readiness to the individual [6]. This is possible triggering of their response, comprising an ultrashort negative feedback
thanks to the sympathetic division of the autonomic nervous system loop on the HPA axis [26].
(ANS), which increases cardiorespiratory function and inhibits insulin Recently, a fast negative feedback, mediated by retrograde signaling
secretion, as well as trigger the release of catecholamines (adrenaline of endocannabinoids has been elucidated [27,28]. In the PVN, binding
and noradrenaline) by the adrenal glands, to promote a diffuse systemic of the GC to the membrane GR receptor induces the synthesis of endo­
physiological response [7]. In addition, catecholamines modulate the cannabinoids, which diffuse through the postsynaptic neuron membrane
immune system, stimulating the innate immune system’s readiness [8]. and act in inhibitory receptors at the presynaptic terminal to reduce CRH
At the central level, cognitive arousal is mainly mediated by the secretion, for instance [27]. In addition to the delayed and fast negative
noradrenergic activity of the brainstem nuclei locus coeruleus (LC) and feedback, the HPA axis is also modulated at the PVN level by cortico­
nucleus of the solitary tract (NTS). These nuclei stimulate the release of limbic and brainstem areas, through transsynaptic neural pathways. The
corticotropin-releasing hormone (CRH) by the parvocellular cells of the PVN is inhibited by the ventral hippocampus [29,30] and the prelimbic
paraventricular nucleus of the hypothalamus (PVN) into the hypophy­ (dorsomedial region) prefrontal cortex [31,32], while it is stimulated by
seal portal system. The CRH reaches the anterior pituitary and induces the amygdala [33], infralimbic (ventromedial region) prefrontal cortex
the release of adrenocorticotropic hormone (ACTH). ACTH is synthe­ (as it projects to sites of excitatory inputs to PVN [32,34]) and LC [35].
sized from pro-opiomelanocortin peptide (POMC) and secreted by cor­ The termination of the HPA axis after the stressful event is as
ticotropic cells in the systemic circulation, where it reaches the adrenal adaptive as its activation. The acute stress response (alarm and resis­
glands and stimulates the synthesis and release of glucocorticoids (GC) tance phases) is essential for survival [6]. However, the inability to
[9]. finish the stressor may lead to the third phase of the stress response: the
The main active glucocorticoid found in humans is cortisol, a exhaustion phase [6]. In this phase there is a dysregulation of the ac­
cholesterol-derived steroid hormone which rises in plasma approxi­ tivity or reactivity of the HPA axis, which may be associated with
mately 15 min after a stressful event, initiating the second phase of the pathological conditions such as cardiovascular, autoimmune diseases,
stress response - the resistance phase [6,10]. In the context stress type II diabetes mellitus, as well as some psychiatric disorders, such as
response, this neuroendocrine loop, the hypothalamus-pituitary-adrenal unipolar and bipolar depression, post-traumatic stress disorder and
axis (HPA), is responsible for the maintenance of the physiological anxiety disorders [1,36].
changes primarily activated by the ANS and modulates other body sys­
tems, including the adaptive immune system and the inhibition of the 3. Memory systems and stress
reproductive system [7,11,12]. Cortisol also crosses the blood-brain
barrier and acts on brain areas related to emotions and cognition, The knowledge obtained from the study of amnesic patients as a
such as the amygdala, the hippocampus, and the prefrontal cortex [13]. result of injuries in regions of the medial temporal lobe contributed to
The HPA axis is the main endocrine response to stress, since it sustains it the elucidation that memory is not unitary. Also, the use of animal
for an indefinite period, as long as the stressor is maintained [7,10]. models for the study of memory and the search for the engram (i.e., the
Cortisol acts by binding to two receptors: type I or MR (mineralo­ physical trace of memory in the brain) allowed the identification of
corticoid receptor) has a high affinity for cortisol and is mainly multiple memory systems and key structures regulating such systems
responsible for the circadian functions of GCs, and type II or GR [37,38]. However, memory should not be thought of as the confined
(glucocorticoid receptor) has less affinity and mediates the effects and activity of a brain region, but as neural circuits distributed across the
termination of the stress response [14,15]. Classically, these receptors brain, that allow different memory systems to coordinate the several
have been known to have an intracellular distribution and stimulate behavioral responses essential for daily life [39,40].
genomic responses, although nowadays the existence of G In this way, memory has been classified, in terms of its nature, into
protein-coupled versions of these receptors on the cell membrane is also two types: The first is explicit memory, the memory of facts and events,
known, mediating rapid non-genomic and indirect genomic responses which can be expressed by language and is a conscious process in
[16–18]. The membrane-bound receptors show low affinity for cortisol humans. In contrast, the second type, implicit memory, includes a set of
and participate in the stress response [19]. other types of memory in which consciousness is not needed for their
The canonical (intracellular) glucocorticoid receptors are transcrip­ expression, such as emotional memory, priming, conditioning, habits,
tion factors that, in the absence of a ligand, have their nuclear locali­ motor skills, habituation and sensitization [37,41].
zation sequence blocked by chaperone proteins [20]. The binding of the Another categorization of memory is made regarding its duration.
GC to the receptor induces the dissociation of the chaperones, so that the Short-term memory (STM) is responsible for storing information for
GC-receptor complex can be translocated to the nucleus, where it is able seconds to hours, allowing the efficient execution of activities or goals; it
to directly interact with DNA, – in regions responsive to glucocorticoids is conscious, limited and very vulnerable to interference [42]. In the
(GRE, glucocorticoid response element) [21] – or indirectly, through field of cognitive neuroscience, this terminology has been overwritten
protein-protein interactions (tethering), for instance, with transcription by the working memory model [43,44], which is the reason why we will
factors (see [22] for an in-depth discussion). Among the genes regulated address it in this review. Such a model is ahead of the passive storage of
by GC in the brain, some are related to plasticity and synaptic information in the short-term, as according to the concept of STM, to be

2
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

also able to actively manipulate and update such information [45]. results may be the time between the onset of stress and the assessment of
Long-term memories (LTM), on the other hand, are those from which we working memory (the longer the time, the greater the impairment), sex
can recall, consciously or not, the information retained for more than six (stronger impairment in men) and controllability over a stressor
hours [46,47]. (impairment when the stressor is uncontrollable) [59,64].
In an attempt to provide an alternative to conscious/non-conscious The phasic release of catecholamines during acute stress accentuates
retrieval-based classification, some other models have been proposed, glutamatergic neurotransmission in the PFC mediated by α2A adren­
although an in-depth discussion on this topic is outside the scope of this ergic receptors, which reduce membrane hyperpolarization [59,65].
review, we suggest ref [48] for a full review. Since the model that dis­ Then, the initial actions of glucocorticoids reinforce the release of PFC
tinguishes between explicit and implicit memory is well-established, and glutamate, potentiating AMPA (α-amino-3-hydroxy-5-methyl-4-isoxaz
studies of the influence of stress on mnemonic processes usually use olepropionic acid receptor) and NMDA receptors ionic currents, as well
terminologies from this model, we make use of this traditional model, as inducing the expression of AMPA subunits and its traffic to the
addressing different topics such as working memory (as a synonym for postsynaptic membrane, via genomic and non-genomic mechanisms
short-term memory) and long-term memories, as constituted by explicit [25,63,65]. Therefore, these initial actions induced by the stress
and implicit memories. response would improve WM by enhancing PFC functioning.
However, it is important to highlight that, according to nonhuman
3.1. Working memory animal studies, even acute stress, if uncontrollable, may weaken PFC
synapses, causing impairments to working memory. In this context, the
The term working memory (WM) is used to designate the conscious release of catecholamines becomes tonic in the PFC. Thus, NA in excess
maintenance and manipulation of relevant transient information during binds to low affinity receptors (α1), which weaken noradrenergic
the performance of a task that requires the use of complex executive neurotransmission due to the hyperpolarization of the postsynaptic
functions [43]. WM has a limited capacity for manipulating information membrane [66]. The noise inhibition promoted by DA becomes unre­
and can work independently of long-term memories (LTM), however, stricted, leading to the weakening of glutamatergic synapses, also due to
once its capacity is exceeded, the LTM contribute to the action that was hyperpolarization, which makes it difficult to propagate the post­
being performed by WM [49]. Animals whose prefrontal cortex (PFC) synaptic excitatory potential and therefore, impairs PFC-dependent ac­
has been injured or surgically cooled present losses in the performance tivities [59,67].
of delayed response tasks, placing the PFC as a possible region of These contrasting effects of acute stress are also expressed at WM
working memory processing [50]. The prefrontal cortex is a phyloge­ subsystems levels, where reports show both loss of visual and verbal
netically recent area of the neocortex, located in the anterior portion of working memory, mainly at high loads [61,68,69], but also improve­
the frontal lobe, and is related to executive functions, top-down control ments [70].
(visceral and emotional regulation), preparation in response to envi­ Regarding the effects of chronic stress on WM, studies are clear in
ronmental conditions (preparatory set), and inhibitory control [51]. pointing out the negative relationship between them, which is mediated
Working memory can be divided into four subsystems: visuospatial, by high levels of cortisol [68,71,72]. The PFC is overly sensitive to
verbal, episodic buffer, and central executive control [43]. Visuospatial hypercortisolemia, with morphological changes such as the reduction of
WM comprises the components of spatial knowledge and object recog­ dendrites and dendritic spines [73,74]. In this context, glutamatergic
nition, which depend on the connections between the PFC and the pa­ transmission is buffered by an increase in GABAergic transmission in the
rietal and temporal cortex, respectively [52]. The verbal subsystem, also rodent prefrontal infralimbic cortex (ilPFC) [72,75,76]. During acute
called phonological loop, maintains representations of spoken language; stress, the interneurons that reduce the activity of ilPFC are inhibited via
although its underlying circuitry is not well understood, it is known that the GR. In chronic stress such regulation is lost, due to high levels of
it depends on the temporal and prefrontal cortices [53,54]. The episodic glucocorticoids that result in the downregulation of GR in parvalbumi­
buffer combines sensory modalities and allows interaction with nergic interneurons. In this sense, there is greater interneuron activity,
perception or long-term memory [43,55]. The subsystems are regulated and consequently, inhibition of ilPFC pyramidal cells, which leads to
by an executive control center, also in the PFC, which allocates and prefrontal hypofunction not only in this region, but also in pre-limbic,
directs energy and attention to the subsystems [56]. lateral, and orbitofrontal regions [77–81]. Such changes are associated
PFC activity is mediated by glutamate and its receptors, mainly with a loss in working memory and other executive functions dependent
NMDAR (N-methyl-D-aspartate receptor), and modulated by diffuse on the PFC [75].
projections, especially dopamine (DA) and noradrenaline (NA) [57,58].
The noradrenergic activity under the PFC occurs in an inverted U-shape 3.2. Long-term memory
manner, mediated by the affinity to its receptors. At moderate levels, NA
binds to postsynaptic α2A receptors, preventing membrane hyperpo­ Long-term memory is commonly divided into explicit and implicit
larization; therefore, it enhances the transmission of the nerve impulse. memories. Explicit memory allows the retrieval of facts and events that,
However, in excess, NA binds to postsynaptic α1 receptors, inducing in humans, requires consciousness and can be expressed by language.
hyperpolarization and weakening the synapse. The PFC hypofunction The memory of facts is also called semantic memory, as it consists of
that results due to this weakening of the synapse may impair working knowledge about words and their meanings; while the event memory is
memory [57,59]. called episodic, as it represents personal experiences [44]. Implicit
The dopaminergic activity in the PFC is mediated mainly by the D1 long-term memory, on the other hand, is all non-explicit memory, which
receptors’ family (D1 and D5) and, just as NA, it has an inverted U-shape can be expressed through behavior, and is difficult to verbalize, such as
response [60]. DA acts mainly by inhibiting neurons whose transmitted emotional and motor memories. These can be either non-associative,
information is irrelevant to the main mnemonic information that must when learning results from a single stimulus (habituation and sensiti­
be maintained during the delay phase (when the mental representation zation), or associative, when learning results from the association of two
of a stimulus in its absence takes place) of a working memory task [60], stimuli (conditioning). Moreover, priming, which is the ability to un­
acting as an inhibitor of noise or distractors, helping to focus attention. consciously access items to which the individual was previously
exposed, is also a form of implicit memory. An important feature of
3.1.1. Stress and working memory implicit memories is their unconscious and automatic recall [44,82].
The evidence of the effects of acute stress on WM are mixed, with For LTM formation, three processes or operational phases are
studies pointing out either impairment [61,62] or increase in working required: acquisition, storage, and consolidation. Acquisition or
memory performance [25,63]. Possible determinants of such contrasting encoding consists of capturing, processing, and coding information into

3
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

neural signals. Memory storage happens through the association of new signaling impairs memory retention, as well as infusing it is able to
information with already existing ones by means of cell assemblies in a recover such impairments [46,98].
neural circuitry. The stored memory can then be consolidated (con­ Currently, it is known that there are two biologically active isoforms
nections are functionally and structurally strengthened) or forgotten of BDNF: pro-BDNF (molecule with the pro domain still attached) and
(connections are weakened or lost) [83]. mature BDNF (mBDNF, whose pro domain has been cleaved). Pro-BDNF
For a memory to be consolidated, retention in the form of short-term has higher affinity to the pan-neurotrophin receptor 75 (p75NTR), which
memory is not necessarily required, at least for some explicit memories, can lead to neuronal death, while mBDNF has higher affinity to the TrkB
as assessed by the inhibitory avoidance task [84]. Memory consolidation receptor, that induces neuronal survival [95]. TrkB-mediated signaling
is facilitated by repetition, by association with previously consolidated is required for LTP and hippocampal neurogenesis; consequently, it is
information, or according to the emotional content associated with the implicated in mnemonic processes [99–102]. Recently, it has been
mnemonic information in question. Although both types of LTM, explicit shown that after cleavage of proBDNF, the pro domain (or pro-peptide)
and implicit, are subject to forgetfulness, the latter is less flexible and, has biological activity, activating p75NTR receptors [103,104].
therefore, less vulnerable to modifications and forgetfulness [85]. Therefore, the repeated stimulation of a circuit by a given stimulus,
Finally, a fourth phase of LTM is retrieval, which consists of later strengthens its functioning through LTP, and with the persistence of the
access to the previously consolidated memories. Retrieving a memory stimulus, induces structural changes in the synapses of this circuit; a
makes it labile and susceptible to interference, requiring a new consol­ process that has been considered the cellular correlate of memory for­
idation of reactivated memory to stabilize it, a process called reconso­ mation [105].
lidation [86,87]. Habits are a motor memory based on a trial-and-error behavioral
response, and, like other motor memories, it requires repetition for
3.2.1. Explicit and implicit memories learning [44]. Habits are valuable in the interaction between stress and
The neural substrate of semantic memory, a long-term explicit memory and here they will be approached as an example of long-term
memory, is distributed throughout the cortex, including the lateral and implicit memory.
ventral temporal cortex, the inferior parietal lobe, in addition to the Initially, habits are susceptible to the value of the reward and the
interactions of these areas with the prefrontal cortex [88]. The biological effort-reinforcement, which is then developed when the balance is
substrate of episodic memory, in turn, depends on interactions between positive. This type of motor memory, like the others, is rigid, automatic,
the prefrontal cortex and the medial temporal lobe [89]. and repetitive [44,106]. Electrophysiological studies in animals show an
Much of the cellular and molecular mechanisms of explicit memory important and distinct participation of the striatum in the acquisition
are known thanks to the study of the hippocampus and anatomically and performance of habitual learning. Dopaminergic circuits between
adjacent areas. The hippocampal formation is located inside the medial the ventral striatum, the ventral tegmental area (VTA), and GABAergic
temporal lobe, and is composed of the hippocampus (CA1, CA2, CA3), nigroventral-tegmental circuits, are related to the assessment of the
dentate gyrus (DG), subicular complex and entorhinal cortex (EC) [89]. reward value of motivated (goal-directed) behaviors [106]. As the
The hippocampal formation is peculiar since it has a unidirectional tri­ behavior is repeated and the balance is positive, there is a transition in
synaptic pathway: the EC projects to the DG (perforant pathway), which the engagement of the ventral striatum to the dorsomedial striatum; at
in turn, projects to CA3 (mossy fibers) and from there to CA1 (Schaffer the post-learning (i.e., when habitual memory is learned) there is the
collateral). CA1, in turn, projects to the EC and subiculum. In addition, participation of the dorsolateral striatum [106], which acts simulta­
CA3 has intrinsic connections and projects back to the DG. The EC neously with the infralimbic prefrontal cortex, necessary for both
projects towards neocortical areas and the adjacent perirhinal and forming and performing a habit in real time [44]. In humans, however,
parahippocampal (postrhinal in rodents) cortices [89]. Therefore, it is the participation of striatum subregions is not well established [107,
possible to notice that the EC is both the main afferent and efferent 108].
hippocampal pathway [90]. Not only the hippocampus itself but other Moreover, the reciprocal communication between the cortex, basal
medial temporal lobe regions, such as perirhinal and parahippocampal ganglia, and cerebellum contributes to the formation of motor memory,
cortices, support different aspects of explicit memory [91]. However, we recruiting different neural networks according to the stages of sequential
will focus here on the proposed hippocampal cellular mechanisms sup­ learning [92]: initially learning based on exploration or trial and error
porting explicit memory since it has a solid underlying literature. (recruitment of vmPFC, ventral striatum and posterior cerebellum), then
Memory storage mediated by the hippocampus initially consists of recruiting the associative network (dlPFC, dorsomedial striatum and
changes in the hippocampal circuit’s synaptic activities through long- lateral posterior cerebellum). Finally, after the establishment of the
term potentiation (LTP). LTP is a special excitatory postsynaptic po­ habit or skill (formation of motor memory), the motor network (sup­
tential resulting from a high frequency presynaptic stimulation associ­ plementary motor area, putamen, and anterior cerebellum) is then
ated with a long-lasting postsynaptic depolarization, which is translated recruited [109,110].
into neural plasticity with associative and long-lasting characteristics Emotional memories are often studied in animals using a fear con­
[92]. LTP is well characterized in the hippocampal circuitry, where at ditioning paradigm, where an aversive stimulus (US, unconditioned
least two types of LTP are identified: one NMDAR-dependent (on stimulus) is regularly paired with a neutral one (CS, conditioned stim­
Schaffer collateral synapses), and another (on mossy fibers synapses) ulus), generating a fear of the CS even in the absence of the US (CR,
that is independent of this receptor, mediated by voltage-dependent conditioned response) [111], which may be expressed as the animal
Ca2+ channels and metabotropic glutamate receptors [93,94]. freezing response. The amygdala nuclei are core to the processing, for­
The concurrent activation of AMPAR and NMDAR leads to Ca2+ mation, and expression of fear conditioning, as reviewed elsewhere
influx. The downstream effect of Ca2+ will activate several intracellular [111–114]. In humans, fear conditioning relies on similar brain regions
signaling pathways, ultimately initiating the LTP’s protein synthesis [115], but it is frequently assessed by the skin conductance response and
phase, responsible for the functional strengthening and, later, the the startle reflex [116,117]. Central NA (via LC), and peripheral
structural synaptic transmission. One of the key genes in this process is adrenaline (via vagal-NTS signaling) [118], play a central role in
bdnf, that encodes the brain-derived neurotrophic factor (BDNF), a modulating fear conditioning, acting directly on the amygdala and on
neurotrophin involved in several aspects of synaptic plasticity, that brain areas related with amygdala regulation, such as PFC and hippo­
range from the potentiation of function and the modulation of excitatory campus [119]. Amygdala-dependent memory is also an important
neurotransmitters and receptors, to ultrastructural changes in the syn­ moderator for other memory systems, such as explicit memory. The
apse [95], making long-term changes on the synapse [96,97]. Therefore, processing of contents’ valence and arousal are amygdala-dependent
BDNF has a role on explicit memory consolidation, since blocking its and may change the direction of memory, mainly mediated by the

4
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

noradrenergic activity [111]. the acquisition of stressor-related contents would suppress the retrieval
of previous content [140] (Fig. 1).
3.2.2. Stress and the long-term memories While the interaction between acute stress and explicit LTM depends
The influence of stress on LTM is dependent on several factors, such on the several factors considered above, chronic stress often tends to
as: the timing when the stressor event is experienced (that is, before, impair explicit memory, whether it occurs before or after learning [102,
during or after the task/learning), the emotional valence of the event 141] (Fig. 1). The harmful effects of chronic stress on explicit memory
(neutral, positive, or negative), and the duration of the stressful event are explained based on the glucocorticoid cascade hypothesis, in which
(acute or chronic). chronic exposure to GC would downregulate GR and BDNF in the hip­
The acute stress can impair explicit LTM acquisition when the in­ pocampus, making it vulnerable and impairing the retrieval of explicit
terval between the stressor and the task is too long (that is, outside the memories [102,142]. Interestingly, inhibiting GC synthesis or endo­
context of learning): the longer the interval, the greater the impairment, cannabinoid metabolism prevents memory impairments [143,144].
as suggested by a meta-analysis of human studies [120] (Fig. 1). Regarding implicit memory, acute stress may increase acquisition as
Nevertheless, when acute stress is experienced in the context of learning, well as consolidation of fear conditioning both in humans [145] and
that is, in the acquisition of information and prior to the consolidation of animals [146] (for review, see [147]), by stimulating the GC-NA inter­
memory, these memory operations are facilitated [120] (Fig. 1). How­ action in the amygdala [148]. In fact, blocking BLA NA signaling with a
ever, we shall notice that this is true only when the items assessed by the β receptor antagonist prevents this memory enhancement, but no loss or
task show some relationship with the stressor event (i.e., they are enhancement effect is seen when GC is absent [149], suggesting only a
important for the response) [120]. permissive effect of GC on the noradrenergic activity in the BLA. Con­
Acute stress during acquisition or consolidation, free of the genomic cerning memory retrieval, there are some reports of impairment induced
effects of GC, enhances memory consolidation through catecholamines by acute stress on implicit memories both in rodents [150] and humans
actions, with a substantial role for β receptor-mediated noradrenergic [151]. Chronically stressed animals, in turn, seem to have an enhanced
activity in the BLA [121–124] (Fig. 1). Therefore, memories with fear memory [152–154], which may be due to the BDNF-induced tro­
emotionally arousal content, positive or negative, are easily consoli­ phic effects of chronic stress on amygdala, alongside the weakening in
dated, as compared to neutral ones, mediated by NA signaling the PFC [148,153–155].
[125–127]. Furthermore, the GR-activated intracellular pathways in Besides the previously mentioned moderator factors of stress effects
acute stress recruit and increase BDNF signaling, that also has a role on on memory, the observed heterogeneity may also be due to methodo­
memory formation [98]. However, the relationship between GC and logical issues, such as the diversity of tasks and unclear stressor timing
BDNF is in an inverted U-shape [128,129], brain region-dependent: in definition. In this sense, this research field would benefit of having
the PFC and in the hippocampus, chronic stress leads to a decrease in guidelines to provide some standardization regarding the methods and
BDNF [102,130], while in the amygdala, high levels of cortisol increase assessments.
BDNF expression, dendritic branching, and spinal growth [131–133]. It
is also suggested that BDNF-TrkB signaling is enhanced by an interaction 3.2.3. Stress modulation between explicit and implicit memory systems
between GR and TrkB. When excessive, however, glucocorticoids bind to Several memory systems operate cooperatively or competitively to
the GR receptor, decreasing the GR-TrkB interaction and reducing the guide the behavioral output responsible for our daily actions [156].
downstream BDNF signaling [128,129]. Stress is one of the factors that modulate the strategy used (i.e., the
When acute stress is experienced during retrieval, however, both recruited memory system) to perform a certain activity [157]. It is
genomic and non-genomic effects of GC, via MR, inhibit the enhance­ possible to evaluate this modulation using tasks with a double possibility
ment of explicit LTM [134–139]. A proposed explanation points out that of resolution - that is, through a cognitive or habit response -, for

Fig. 1. Stress influence on long-term memory operational phases is mainly dependent on stressor timing. When acute stress happens around learning (immediately
before acquisition or consolidation), noradrenergic (NA) and non-genomic glucocorticoids (GC) enhance explicit and implicit memory formation of the stressful
event, which is further maintained by genomic GC effects. The consolidation and retrieval of stress-unrelated items, however, is suppressed. Moreover, if the interval
of acute stress and learning is too long, the genomic GC effects impair the acquisition of the content. If the acute stress happens in the retrieval context, both NA/non-
genomic and genomic GC effects impair retrieval of explicit and implicit memory, since the brain is on the memory formation mode. Whatever happening before or
after learning, chronic stress often suppresses explicit memory while may enhance implicit memory.

5
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

instance, with the T-maze. memory (dependent on the dorsal striatum) (Fig. 2). At first glance, an
In this task, the rodent is placed in the center of a labyrinth with four inflexible, instinctive, and primitive behavioral response may seem
arms and must learn which arm the reward is on. During learning, a disadvantageous; however, such a strategy is less energetically expen­
cognitive strategy, mediated by explicit memory, can be recruited, in sive than the cognitive one and becomes highly adaptive in contexts
which the individual uses distal spatial clues (allocentric strategy) to where a rapid response is needed, such as in an ancestral environment,
solve the task (for example, memorizing the position of the arm con­ where the circuits and behaviors underlying the stress response evolved
taining the reward in relation to the other or still being guided by clues, [38].
such as objects positioned in the arms of the labyrinth) - or a habit
strategy - implicit memory in which the response is of the stimulus- 4. Stress, memory, and psychiatric implications: major
response type (for example, if whenever the animal turns right when depressive disorder
starting the task he gets the reward, the command “turn right” is then
associated with the reward; egocentric strategy). To assess which Despite the fact that the Diagnostic and Statistical Manual of Mental
memory system was recruited during learning, the rodent is placed in a Disorders (DSM-5) [5] presents a chapter dedicated to stress-related
new point in the labyrinth: if the strategy of navigating through the disorders, approaching, for instance, acute stress disorder and
labyrinth is used, learning was cognitive (hippocampus-dependent); if post-traumatic stress disorder (PTSD), the relationship between stress
the “turn to the right” strategy was used, learning was habitual (striatal- and mood disorders is well established [164–166]. Chronic stress is one
dependent) [157,158]. of the main etiologies of Major Depressive Disorder (MDD) [167], of
Exposure to either acute or chronic stress appears to modulate the which impaired memory is part of its diagnosis [5]. Therefore, a better
neural substrates of cognitive and habitual/emotional learning understanding of memory impairments in MDD and how they are
distinctly. In humans, the weakening of the PFC and the hippocampus, associated with the chronic stress response can open the doors for its use
induced by chronic stress, also weakens the recruitment of cognitive and as a prognostic and diagnostic tool, as well as in prophylactic and
instrumental learning to solve a task [159]. The effects of chronic stress therapeutic approaches, and treatment follow-up investigations.
seem to be reflected at the cellular level, in terms of PFC atrophy and The changes in memory presented by depressive patients are mainly
dorsal striatum hypertrophy, as depicted in animal studies [160]. In related to long-term explicit memory (autobiographical and episodic
addition, weakening of the PFC leads to a loss of top-down regulation, memories) and executive functions [168]. In depressed individuals,
where the activity of the amygdala and the striatum overlaps the PFC, mainly in the most severe cases, a hippocampal volumetric decrease and
inducing a rigid, quick, and instinctive behavioral response (lack of suppression of adult hippocampal neurogenesis may occur [169,170].
behavioral flexibility), interfering with decision making [160,161] Poor performance in tasks, such as the Autobiographical Memory Test
(Fig. 2). The activity of the amygdala, mainly the BLA, is increased by [171], and auditory and visual episodic memory tasks [172,173], is also
the permissive effect of GC on noradrenergic pathways; together, GC and found and confirmed by meta-analytic results [174]. Moreover, episodic
NA are essential to orient the action of the striatum [162,163]. When memory impairments were shown to be correlated with smaller hippo­
administered with propranolol, a β receptor antagonist-adrenergic, campal volume [175]. These structural changes are also observed in
recruitment of the striatum is prevented even when there is an infu­ nonhuman animals submitted to chronic stress [176,177] and are
sion of GC [162]. related to impairments on long-term memory [178].
Therefore, chronic stress favors a switch in the use of memory sys­ The impairment of executive function, which includes poor perfor­
tems, from a more flexible cognitive memory (dependent on the hip­ mance in working memory, attention, and recognition tasks [168,179],
pocampus and/or the prefrontal cortex), to a more rigid and instinctive suggests that working memory is impaired both in visuospatial [180,

Fig. 2. Stress duration differently influence the structure and function of key brain regions to processing of memory. Acute controllable stress may enhance synapses
within prefrontal cortex (PFC) as well as hippocampal (HIPP), therefore favoring working memory and long-term memory, respectively. In addition, the PFC top-
down control over the amygdala (AMY) suppresses instinctive and habitual responses mediated by amygdala and striatum (Str), favoring cognitive flexibility,
mediated by HIPP-PFC connection (upper left panel). When uncontrollable or intense, acute stress functionally impair PFC and hippocampal, as well as frontal-
hippocampal connectivity, weakening top-down control and enabling an amygdala-striatal-dependent instinctive response (upper right panel). The persistence of
chronic stress leads to different structural changes in these regions: atrophy in PFC and hippocampus, and hypertrophy in amygdala and striatum. In this context, the
top-down turns into an amygdala-striatal bottom-up control over frontal-hippocampal networks, shifting the memory system recruitment underlying learning to a
non-flexible, instinctive, and stimulus-response dependent habitual learning (lower left panel). Arrow’s thickness denotes connection strength.

6
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

181] and auditory [172,182] tasks. More importantly, executive effi­ symptoms [199]. In addition, after the treatment, salivary cortisol levels
ciency loss seems to be functionally associated with a decrease in PFC were similar to those of healthy controls [192], as well as the reduced
activity [181,183,184]. levels of systemic inflammation that were found [208]. These im­
In addition, in depressive patients, a stronger retrieval of negative provements enhance homeostatic balance and favor cognitive processes.
memories and attentional bias towards negative stimuli is found, while Moreover, ayahuasca has a broad range of action in the brain, modu­
the retrieval of positive memories is impoverished [168,178,185–187]. lating areas of the default mode network (implicated in ruminative
Evidence points to a greater activity of the amygdala in face of negative processes) [209], stimulating neurogenesis in vitro(through its active
emotional stimuli, and a blunted response to positive stimuli [151]. compounds), preventing glutamate-induced excitotoxicity, and
Therefore, changes in the processing of emotional memory by amygdala improving spatial memory [210,211].
dysfunction will mediate sensitization to negative stimuli, as well as the The use of memory as a tool for prevention, diagnosis and treatment
larger retrieval of negative memories, which are both observed in in psychiatry is an emerging field [212,213]. Some learning strategies in
depression [188]. a WM verbal task were used to differentiate psychotic from
Changes in the HPA axis are a frequent finding in depressed in­ non-psychotic depression helping to provide a more accurate diagnosis,
dividuals. Given the widespread location of the cortisol receptors in being a relatively passive strategy (serial clustering), the latter used by
important areas for memory, such as the PFC, hippocampus, and volunteers with psychotic depression, and being the one that best
amygdala, the HPA axis dysregulation may be one of the causes of differentiated the groups [214]. In addition, memory reconsolidation
cognitive impairments in depressed individuals [166,189,190]. Both has been a target for modifying traumatic memories, proposing alter­
cortisol levels and some genetic polymorphisms of their receptors are natives for both the prevention and treatment of psychopathologies
predictors of cognitive performance in depressed patients [166]. Inter­ [215,216]. The retrieval of positive memories works as a resilience
estingly, there seems to be a specificity between receptor variation and factor in adolescents at risk of developing depression (who experienced
the type of task, in which variations in GR are normally a predictor of stressful events in the beginning of life) [217], supporting the role of
executive function tasks, while variations in MR generally predict memory modulation as a prevention tool. In mice, behavioral and
acquisition and retrieval of long-term memories [166]. stress-induced hippocampal neurogenesis impairments were reversed by
Hypercortisolemia, the most frequent alteration of the HPA axis in optogenetically activating an engram related to a previous positive
MDD, probably occurs due to a failure in the HPA axis feedback, trig­ memory [218]. Therefore, plenty of approaches are currently being
gered by a loss of GR sensitivity, as suggested by studies with dexa­ developed to uncover the interplay between memory and various
methasone suppression tests [190,191]. However, a hypocortisolemic research fields in psychiatry, which is placing memory as a promising
profile has also been reported in depressive patients, particularly those tool for Psychiatry.
with severe depression, with comorbidities, or interned patients [192,
193]. Among the pathophysiological mechanisms proposed to explain 5. Conclusions
hypocortisolemia there is an uncompensated sensitivity in the feedback
of the HPA axis, and an insufficiency of the adrenal glands to synthesize There are several memory systems, classified generally based on
cortisol [192,193]. Both hypo- and hypercortisolemic profiles are shown their content or duration, whose neurobiology is not yet fully elucidated.
to impair memory. Hypocortisolemia was shown to be associated with Neuroendocrine mediators of the stress response are important modu­
low performance in tasks such as auditory and visual memory as well as lators of mnemonic processes, and their effects depend on certain
executive function [194,195]. Similarly, individuals with in a hyper­ characteristics of the stressor, such as: the duration/intensity and
cortisolemic condition often show impaired performance in verbal and controllability, and the timing in which the stressful event is experi­
visual memory tasks [196,197]. enced (before, during, or after the mnemonic context). In summary,
Although it does not occur consistently, changes in blood BDNF can evidence is mixed for the effects of acute stress on working memory, and
be reflected in the decrease in serum levels of depressed individuals points to contrasting effects on the acquisition, consolidation, and
[198,199]. Interestingly, studies have been pointing out that the effect retrieval of long-term memories, either explicit or implicit. Moreover, it
of antidepressants seems to require an increase in BDNF synthesis and also influences the recruited memory system; the transition from
hippocampal neurogenesis [200–202]. However, in a recent cognitive learning to habits. Chronic stress, on the other hand, impairs
meta-analysis, Semkovska and colleagues [4] found that even in­ both working memory and the operational phases of explicit long-term
dividuals who achieved remission after a depressive episode treated memories. Therefore, impairments in memory are common in psychi­
with commercial antidepressants perform poorly on most cognitive atric disorders, such as major depressive disorder, which is accompanied
variables when compared to controls, with a high effect size for variables by extensive changes in episodic memory and executive functions. Such
related to long-term memory [4]. impairments are usually due to biological changes underlying mne­
Therefore, having in mind the physiological relationship among monic processes. In this context, the study of the neurobiology of
cortisol, memory, and mood [166], new treatments for MDD should not memory may contribute both to the validation of biomarkers, as well as
only have the aim of decreasing depressed mood and anhedonia, but also to point out therapeutic targets.
focus on memory improvement and cortisol balance, in order to have a
better treatment response and less recurrent episodes. Studies with ke­ Source of funding
tamine, a psychotropic with rapid antidepressant action recently
approved for clinical use for the treatment resistant depression (TRD), This research did not receive any specific grant from funding
have shown a quick ketamine-induced increase on BDNF release asso­ agencies in the public, commercial, or not-for-profit sectors.
ciated with mood improvement, and decrease in negative mnemonic
associations [203]. Despite these reported benefits, impairment in
episodic memory associated with sub-anesthetic doses of ketamine in Declaration of Competing Interest
animal models of depression has also been reported [204].
Another promising new treatment for TRD is ayahuasca. Ayahuasca The authors report no declarations of interest.
is an infusion of Amazonian plants, used in indigenous and non-
indigenous religious rituals, with psychedelic properties [205–207]. In References
a study with hypocortisolemic patients with TRD, a single dose of
[1] S. Ramamoorthy, J.A. Cidlowski, Corticosteroids. Mechanisms of action in health
ayahuasca induced a rapid increase in serum BDNF levels (48 h after and disease, Rheum. Dis. Clin. North Am. 42 (2016) 15–31, https://doi.org/
drinking the brew), which were correlated with reductions in depressive 10.1016/j.rdc.2015.08.002.

7
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

[2] L. Schwabe, O.T. Wolf, M.S. Oitzl, Memory formation under stress: quantity and mechanism, J. Neurosci. 23 (2003) 4850–4857, https://doi.org/10.1523/
quality, Neurosci. Biobehav. Rev. 34 (2010) 584–591, https://doi.org/10.1016/j. JNEUROSCI.23-12-04850.2003.
neubiorev.2009.11.015. [28] N.K. Evanson, J.G. Tasker, M.N. Hill, C.J. Hillard, J.P. Herman, Fast feedback
[3] R.S. Duman, G.K. Aghajanian, G. Sanacora, J.H. Krystal, Synaptic plasticity and inhibition of the HPA axis by glucocorticoids is mediated by endocannabinoid
depression: new insights from stress and rapid-acting antidepressants, Nat. Med. signaling, Endocrinology 151 (2010) 4811–4819, https://doi.org/10.1210/
22 (2016) 238–249, https://doi.org/10.1038/nm.4050. en.2010-0285.
[4] M. Semkovska, L. Quinlivan, T. O’Grady, R. Johnson, A. Collins, J. O’Connor, [29] W.E. Cullinan, J.P. Herman, S.J. Watson, Ventral subicular interaction with the
H. Knittle, E. Ahern, T. Gload, Cognitive function following a major depressive hypothalamic paraventricular nucleus: evidence for a relay in the bed nucleus of
episode: a systematic review and meta-analysis, Lancet Psychiatry 0366 (2019) the stria terminalis, J. Comp. Neurol. 332 (1993) 1–20, https://doi.org/10.1002/
1–11, https://doi.org/10.1016/S2215-0366(19)30291-3. cne.903320102.
[5] American Psychiatric Association, Manual diagnóstico e estatístico de transtornos [30] J.P. Herman, C.M. Dolgas, S.L. Carlson, Ventral subiculum regulates
mentais: DSM-5-5a Edição, 5th ed., Artmed, Porto Alegre, 2015. hypothalamo-pituitary-adrenocortical and behavioural responses to cognitive
[6] H. Selye, The general adaptation syndrome and the diseases of adaptation, J. Clin. stressors, Neuroscience 86 (1998) 449–459, https://doi.org/10.1016/S0306-
Endocrinol. Metab. 6 (1946) 117–230, https://doi.org/10.1210/jcem-6-2-117. 4522(98)00055-4.
[7] M.B.C. de Sousa, H.P.A. Silva, N.L. Galvão-Coelho, Resposta ao estresse: I. [31] J.J. Radley, K.L. Gosselink, P.E. Sawchenko, A discrete GABAergic relay mediates
Homeostase e teoria da alostase, Estud. Psicol. 20 (2015) 1–10, https://doi.org/ medial prefrontal cortical inhibition of the neuroendocrine stress response,
10.5935/1678-4669.20150002. J. Neurosci. 29 (2009) 7330–7340, https://doi.org/10.1523/JNEUROSCI.5924-
[8] S.C. Segerstrom, Resources, stress, and immunity: an ecological perspective on 08.2009.
human psychoneuroimmunology, Ann. Behav. Med. 40 (2010) 114–125, https:// [32] J.J. Radley, C.M. Arias, P.E. Sawchenko, Regional differentiation of the medial
doi.org/10.1007/s12160-010-9195-3. prefrontal cortex in regulating adaptive responses to acute emotional stress,
[9] J.P. Herman, J.M. McKlveen, S. Ghosal, B. Kopp, A. Wulsin, R. Makinson, J. Neurosci. 26 (2006) 12967–12976, https://doi.org/10.1523/
J. Scheimann, B. Myers, Regulation of the hypothalamic-pituitary- adrenocortical JNEUROSCI.4297-06.2006.
stress response, Compr. Physiol. 6 (2016) 603–621, https://doi.org/10.1002/ [33] C.V. Dayas, K.M. Buller, T.A. Day, Neuroendocrine responses to an emotional
cphy.c150015. stressor: evidence for involvement of the medial but not the central amygdala,
[10] R.M. Sapolsky, L.M. Romero, A.U. Munck, How Do Glucocorticoids Influence Eur. J. Neurosci. 11 (1999) 2312–2322, https://doi.org/10.1046/j.1460-
Stress Responses? Integrating Permissive, Suppressive, Stimulatory, and 9568.1999.00645.x.
Preparative Actions*, 2000. https://academic.oup.com/edrv/article-abstract [34] R.P. Vertes, Differential projections of the infralimbic and prelimbic cortex in the
/21/1/55/2423840. rat, Synapse 51 (2004) 32–58, https://doi.org/10.1002/syn.10279.
[11] B. Liu, L. Zhao, C. Yue, M. Qian, M. Xie, Changes in gonadal function at different [35] Cass Ziegler, Herman, excitatory influence of the locus coeruleus in
stages of chronic restraint stress-induced depression animals, Physiol. Behav. hypothalamic-pituitary-adrenocortical axis responses to stress,
(2019), https://doi.org/10.1016/j.physbeh.2019.112656. J. Neuroendocrinol. 11 (1999) 361–369, https://doi.org/10.1046/j.1365-
[12] H.J. Yuan, Z. Bin Li, X.Y. Zhao, G.Y. Sun, G.L. Wang, Y.Q. Zhao, M. Zhang, J. 2826.1999.00337.x.
H. Tan, Glucocorticoids impair oocyte competence and trigger apoptosis of [36] M. Kadmiel, J.A. Cidlowski, Glucocorticoid receptor signaling in health and
ovarian cells via activating the TNF-α system, Reproduction (2020), https://doi. disease, Trends Pharmacol. Sci. 34 (2013) 518–530, https://doi.org/10.1016/j.
org/10.1530/REP-20-0025. tips.2013.07.003.
[13] J.L. Gerlach, B.S. McEwen, Rat brain binds adrenal steroid hormone: [37] L.R. Squire, J.T. Wixted, The cognitive neuroscience of human memory since H.
radioautography of Hippocampus with corticosterone, Science (80-) 175 (1972) M, Annu. Rev. Neurosci. 34 (2011) 259–288, https://doi.org/10.1146/annurev-
1133–1136, https://doi.org/10.1126/science.175.4026.1133. neuro-061010-113720.
[14] J.M.H.M. REUL, E.R. DE KLOET, Two receptor systems for corticosterone in rat [38] L. Schwabe, Stress and the engagement of multiple memory systems: integration
brain: microdistribution and differential occupation, Endocrinology 117 (1985) of animal and human studies, Hippocampus 23 (2013) 1035–1043, https://doi.
2505–2511, https://doi.org/10.1210/endo-117-6-2505. org/10.1002/hipo.22175.
[15] J.M.H.M. Reul, E.R. De Kloet, Anatomical resolution of two types of [39] T. Kitamura, S.K. Ogawa, D.S. Roy, T. Okuyama, M.D. Morrissey, L.M. Smith, R.
corticosterone receptor sites in rat brain with in vitro autoradiography and L. Redondo, S. Tonegawa, Engrams and circuits crucial for systems consolidation
computerized image analysis, J. Steroid Biochem. 24 (1986) 269–272, https:// of a memory, Science (80-.) 356 (2017) 73–78, https://doi.org/10.1126/science.
doi.org/10.1016/0022-4731(86)90063-4. aam6808.
[16] N.K. Evanson, J.P. Herman, Role of paraventricular nucleus glutamate signaling [40] D.M. Amodio, Social cognition 2.0: an interactive memory systems account,
in regulation of HPA axis stress responses, Interdiscip. Inf. Sci. 21 (2015) Trends Cogn. Sci. 23 (2019) 21–33, https://doi.org/10.1016/j.tics.2018.10.002.
253–260, https://doi.org/10.4036/iis.2015.B.10. [41] E.R. Kandel, Y. Dudai, M.R. Mayford, The molecular and systems biology of
[17] M. Joëls, R. Angela Sarabdjitsingh, H. Karst, Unraveling the time domains of memory, Cell 157 (2014) 163–186, https://doi.org/10.1016/j.cell.2014.03.001.
corticosteroid hormone influences on brain activity: rapid, slow, and chronic [42] N. Cowan, The magical number 4 in short-term memory: a reconsideration of
modes, Pharmacol. Rev. 64 (2012) 901–938, https://doi.org/10.1124/ mental storage capacity, Behav. Brain Sci. 24 (2001) 87–114, https://doi.org/
pr.112.005892. 10.1017/S0140525X01003922.
[18] M. Joëls, T.Z. Baram, The neuro-symphony of stress, Nat. Rev. Neurosci. 10 [43] A. Baddeley, Working memory, Curr. Biol. 20 (2010) R136–R140, https://doi.
(2009) 459–466, https://doi.org/10.1038/nrn2632. org/10.1016/j.cub.2009.12.014.
[19] J. Nahar, J.R. Rainville, G.P. Dohanich, J.G. Tasker, Further evidence for a [44] L.R. Squire, A.J.O. Dede, Conscious and unconscious memory systems, cold spring
membrane receptor that binds glucocorticoids in the rodent hypothalamus, Harb, Perspect. Biol. Med. 7 (2015) a021667, https://doi.org/10.1101/
Steroids 114 (2016) 33–40, https://doi.org/10.1016/j.steroids.2016.05.013. cshperspect.a021667.
[20] D. Picard, B. Khursheed, M.J. Garabedian, M.G. Fortin, S. Lindquist, K. [45] D. Norris, Short-term memory and long-term memory are still different, Psychol.
R. Yamamoto, Reduced levels of hsp90 compromise steroid receptor action in Bull. 143 (2017) 992–1009, https://doi.org/10.1037/bul0000108.
vivo, Nature 348 (1990) 166–168, https://doi.org/10.1038/348166a0. [46] P. Bekinschtein, M. Cammarota, C. Katche, L. Slipczuk, J.I. Rossato, A. Goldin,
[21] V.L. Chandler, B.A. Maler, K.R. Yamamoto, DNA sequences bound specifically by I. Izquierdo, J.H. Medina, BDNF is essential to promote persistence of long-term
glucocorticoid receptor in vitro render a heterologous promoter hormone memory storage, Proc. Natl. Acad. Sci. U. S. A. (2008), https://doi.org/10.1073/
responsive in vivo, Cell 33 (1983) 489–499, https://doi.org/10.1016/0092-8674 pnas.0711863105.
(83)90430-0. [47] I. Izquierdo, J.L. McGaugh, Behavioural pharmacology and its contribution to the
[22] E.R. Weikum, M.T. Knuesel, E.A. Ortlund, K.R. Yamamoto, Glucocorticoid molecular basis of memory consolidation, Behav. Pharmacol. (2000), https://doi.
receptor control of transcription: precision and plasticity via allostery, Nat. Rev. org/10.1097/00008877-200011000-00001.
Mol. Cell Biol. 18 (2017) 159–174, https://doi.org/10.1038/nrm.2016.152. [48] J. Ferbinteanu, Memory systems 2018 – towards a new paradigm, Neurobiol.
[23] C. Finsterwald, C.M. Alberini, Stress and glucocorticoid receptor-dependent Learn. Mem. 157 (2019) 61–78, https://doi.org/10.1016/j.nlm.2018.11.005.
mechanisms in long-term memory: from adaptive responses to [49] A. Jeneson, K.N. Mauldin, L.R. Squire, Intact working memory for relational
psychopathologies, Neurobiol. Learn. Mem. 112 (2014) 17–29, https://doi.org/ information after medial temporal lobe damage, J. Neurosci. 30 (2010)
10.1016/j.nlm.2013.09.017. 13624–13629, https://doi.org/10.1523/JNEUROSCI.2895-10.2010.
[24] H. Karst, S. Berger, M. Turiault, F. Tronche, G. Schütz, M. Joëls, Mineralocorticoid [50] B. Plakke, J. Hwang, L.M. Romanski, Inactivation of primate prefrontal cortex
receptors are indispensable for nongenomic modulation of hippocampal impairs auditory and audiovisual working memory, J. Neurosci. 35 (2015)
glutamate transmission by corticosterone, Proc. Natl. Acad. Sci. U. S. A. 102 9666–9675, https://doi.org/10.1523/JNEUROSCI.1218-15.2015.
(2005) 19204–19207, https://doi.org/10.1073/pnas.0507572102. [51] M. Carlén, What constitutes the prefrontal cortex? Science (80-.) 358 (2017)
[25] E.Y. Yuen, W. Liu, I.N. Karatsoreos, Y. Ren, J. Feng, B.S. McEwen, Z. Yan, 478–482, https://doi.org/10.1126/science.aan8868.
Mechanisms for acute stress-induced enhancement of glutamatergic transmission [52] E.F. Ester, T.C. Sprague, J.T. Serences, Parietal and frontal cortex encode
and working memory, Mol. Psychiatry 16 (2011) 156–170, https://doi.org/ stimulus-specific mnemonic representations during visual working memory,
10.1038/mp.2010.50. Neuron 87 (2015) 893–905, https://doi.org/10.1016/j.neuron.2015.07.013.
[26] G.M. Wochnik, J. Rüegg, G.A. Abel, U. Schmidt, F. Holsboer, T. Rein, FK506- [53] J. Bigelow, B. Rossi, A. Poremba, Neural correlates of short-term memory in
binding proteins 51 and 52 differentially regulate dynein interaction and nuclear primate auditory cortex, Front. Neurosci. 8 (2014) 250, https://doi.org/10.3389/
translocation of the glucocorticoid receptor in mammalian cells, J. Biol. Chem. fnins.2014.00250.
280 (2005) 4609–4616, https://doi.org/10.1074/jbc.M407498200. [54] B. Plakke, C.W. Ng, A. Poremba, Neural correlates of auditory recognition
[27] S. Di, R. Malcher-Lopes, K.C. Halmos, J.G. Tasker, Nongenomic glucocorticoid memory in primate lateral prefrontal cortex, Neuroscience 244 (2013) 62–76,
inhibition via endocannabinoid release in the hypothalamus: a fast feedback https://doi.org/10.1016/j.neuroscience.2013.04.002.

8
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

[55] A. Baddeley, The episodic buffer: a new component of working memory? Trends brain function in adulthood, Proc. Natl. Acad. Sci. U. S. A. (2013), https://doi.
Cogn. Sci. 4 (2000) 417–423, https://doi.org/10.1016/S1364-6613(00)01538-2. org/10.1073/pnas.1308240110.
[56] E.K. Miller, J.D. Cohen, An integrative theory of prefrontal cortex function, Annu. [81] C. Liston, B.S. McEwen, B.J. Casey, Psychosocial stress reversibly disrupts
Rev. Neurosci. 24 (2001) 167–202, https://doi.org/10.1146/annurev. prefrontal processing and attentional control, Proc. Natl. Acad. Sci. U. S. A.
neuro.24.1.167. (2009), https://doi.org/10.1073/pnas.0807041106.
[57] C.W. Berridge, R.C. Spencer, Differential cognitive actions of norepinephrine a2 [82] P.J. Bayley, J.C. Frascino, L.R. Squire, Robust habit learning in the absence of
and a1 receptor signaling in the prefrontal cortex, Brain Res. 1641 (2016) awareness and independent of the medial temporal lobe, Nature 436 (2005)
189–196, https://doi.org/10.1016/j.brainres.2015.11.024. 550–553, https://doi.org/10.1038/nature03857.
[58] B. Xing, Y.C. Li, W.J. Gao, Norepinephrine versus dopamine and their interaction [83] T. Abel, K.M. Lattal, Molecular mechanisms of memory acquisition, consolidation
in modulating synaptic function in the prefrontal cortex, Brain Res. (2016), and retrieval, Curr. Opin. Neurobiol. 11 (2001) 180–187, https://doi.org/
https://doi.org/10.1016/j.brainres.2016.01.005. 10.1016/S0959-4388(00)00194-X.
[59] A.F.T. Arnsten, Stress weakens prefrontal networks: molecular insults to higher [84] I. Izquierdo, D.M. Barros, T. Mello e Souza, M.M. de Souza, L.A. Izquierdo, J.
cognition, Nat. Neurosci. 18 (2015) 1376–1385, https://doi.org/10.1038/ H. Medina, Mechanisms for memory types differ, Nature 393 (1998) 635–636,
nn.4087. https://doi.org/10.1038/31371.
[60] S. Vijayraghavan, M. Wang, S.G. Birnbaum, G.V. Williams, A.F.T. Arnsten, [85] P. Graf, D.L. Schacter, Selective effects of interference on implicit and explicit
Inverted-U dopamine D1 receptor actions on prefrontal neurons engaged in memory for new associations, J. Exp. Psychol. Learn. Mem. Cogn. 13 (1987)
working memory, Nat. Neurosci. 10 (2007) 376–384, https://doi.org/10.1038/ 45–53, https://doi.org/10.1037/0278-7393.13.1.45.
nn1846. [86] J.L.C. Lee, K. Nader, D. Schiller, An update on memory reconsolidation updating,
[61] C. Jiang, P.L.P. Rau, Working memory performance impaired after exposure to Trends Cogn. Sci. (2017), https://doi.org/10.1016/j.tics.2017.04.006.
acute social stress: the evidence comes from ERPs, Neurosci. Lett. 658 (2017) [87] M.H. Milekic, C.M. Alberini, Temporally graded requirement for protein synthesis
137–141, https://doi.org/10.1016/j.neulet.2017.08.054. following memory reactivation, Neuron 36 (2002) 521–525, https://doi.org/
[62] J. Passecker, S. Barlow, S.M. O’Mara, Dissociating effects of acute photic stress on 10.1016/S0896-6273(02)00976-5.
spatial, episodic-like and working memory in the rat, Behav. Brain Res. (2014), [88] J.R. Binder, R.H. Desai, The neurobiology of semantic memory, Trends Cogn. Sci.
https://doi.org/10.1016/j.bbr.2014.07.007. (2011), https://doi.org/10.1016/j.tics.2011.10.001.
[63] E.Y. Yuen, W. Liu, I.N. Karatsoreos, J. Feng, B.S. McEwen, Z. Yan, Acute stress [89] P. Andersen, R. Morris, D. Amaral, T. Bliss, J.O.’ Keefe, The Hippocampus Book,
enhances glutamatergic transmission in prefrontal cortex and facilitates working Oxford University Press, 2009, https://doi.org/10.1093/acprof:oso/
memory, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 14075–14079, https://doi. 9780195100273.001.0001.
org/10.1073/pnas.0906791106. [90] N.M. Van Strien, N.L.M. Cappaert, M.P. Witter, The anatomy of memory: an
[64] G.S. Shields, M.A. Sazma, A.P. Yonelinas, The effects of acute stress on core interactive overview of the parahippocampal- hippocampal network, Nat. Rev.
executive functions: a meta-analysis and comparison with cortisol, Neurosci. Neurosci. (2009), https://doi.org/10.1038/nrn2614.
Biobehav. Rev. 68 (2016) 651–668, https://doi.org/10.1016/j. [91] L. Davachi, Item, context and relational episodic encoding in humans, Curr. Opin.
neubiorev.2016.06.038. Neurobiol. 16 (2006) 693–700, https://doi.org/10.1016/j.conb.2006.10.012.
[65] M. Popoli, Z. Yan, B.S. McEwen, G. Sanacora, The stressed synapse: the impact of [92] C. Lüscher, R.C. Malenka, NMDA receptor-dependent long-term potentiation and
stress and glucocorticoids on glutamate transmission, Nat. Rev. Neurosci. 13 long-term depression (LTP/LTD), Cold Spring Harb. Perspect. Biol. 4 (2012)
(2012) 22–37, https://doi.org/10.1038/nrn3138. 1–15, https://doi.org/10.1101/cshperspect.a005710.
[66] S.G. Birnbaum, P.X. Yuan, M. Wang, S. Vijayraghavan, A.K. Bloom, D.J. Davis, K. [93] T. Manabe, NMDA receptor-independent long-term potentiation in hippocampal
T. Gobeske, J.D. Sweatt, H.K. Manji, A.F.T. Arnsten, Protein kinase C overactivity interneurons, J. Physiol. 595 (2017) 3263–3264, https://doi.org/10.1113/
impairs prefrontal cortical regulation of working memory, Science (80-.) 306 JP274093.
(2004) 882–884, https://doi.org/10.1126/science.1100021. [94] H. Wang, A.O. Ardiles, S. Yang, T. Tran, R. Posada-Duque, G. Valdivia, M. Baek,
[67] A.F.T. Arnsten, Stress signalling pathways that impair prefrontal cortex structure Y.A. Chuang, A.G. Palacios, M. Gallagher, P. Worley, A. Kirkwood, Metabotropic
and function, Nat. Rev. Neurosci. 10 (2009) 410–422, https://doi.org/10.1038/ glutamate receptors induce a form of LTP controlled by translation and arc
nrn2648. signaling in the hippocampus, J. Neurosci. 36 (2016) 1723–1729, https://doi.
[68] N.Y.L. Oei, W.T.A.M. Everaerd, B.M. Elzinga, S. Van Well, B. Bermond, org/10.1523/JNEUROSCI.0878-15.2016.
Psychosocial stress impairs working memory at high loads: an association with [95] M. Sasi, B. Vignoli, M. Canossa, R. Blum, Neurobiology of local and intercellular
cortisol levels and memory retrieval, Stress 9 (2006) 133–141, https://doi.org/ BDNF signaling, Pflugers Arch. 469 (2017) 593–610, https://doi.org/10.1007/
10.1080/10253890600965773. s00424-017-1964-4.
[69] M. Luethi, B. Meier, C. Sandi, Stress effects on working memory, explicit memory, [96] S.C. Harward, N.G. Hedrick, C.E. Hall, P. Parra-Bueno, T.A. Milner, E. Pan,
and implicit memory for neutral and emotional stimuli in healthy men, Front. T. Laviv, B.L. Hempstead, R. Yasuda, J.O. McNamara, Autocrine BDNF-TrkB
Behav. Neurosci. 3 (2009), https://doi.org/10.3389/neuro.08.005.2008. signalling within a single dendritic spine, Nature 538 (2016) 99–103, https://doi.
[70] R. Weerda, M. Muehlhan, O.T. Wolf, C.M. Thiel, Effects of acute psychosocial org/10.1038/nature19766.
stress on working memory related brain activity in men, Hum. Brain Mapp. [97] G. Leal, D. Comprido, C.B. Duarte, BDNF-induced local protein synthesis and
(2010), https://doi.org/10.1002/hbm.20945. synaptic plasticity, Neuropharmacology 76 (2014) 639–656, https://doi.org/
[71] G.W. Evans, M.A. Schamberg, Childhood poverty, chronic stress, and adult 10.1016/j.neuropharm.2013.04.005.
working memory, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 6545–6549, https:// [98] D.Y. Chen, D. Bambah-Mukku, G. Pollonini, C.M. Alberini, Glucocorticoid
doi.org/10.1073/pnas.0811910106. receptors recruit the CaMKIIα-BDNF-CREB pathways to mediate memory
[72] A. Mika, G.J. Mazur, A.N. Hoffman, J.S. Talboom, H.A. Bimonte-Nelson, consolidation, Nat. Neurosci. 15 (2012) 1707–1714, https://doi.org/10.1038/
F. Sanabria, C.D. Conrad, Chronic stress impairs prefrontal cortex-dependent nn.3266.
response inhibition and spatial working memory, Behav. Neurosci. 126 (2012) [99] Y. Li, B.W. Luikart, S. Birnbaum, J. Chen, C.H. Kwon, S.G. Kernie, R. Bassel-Duby,
605–619, https://doi.org/10.1037/a0029642. L.F. Parada, TrkB regulates hippocampal neurogenesis and governs sensitivity to
[73] B.S. McEwen, J.H. Morrison, The brain on stress: vulnerability and plasticity of antidepressive treatment, Neuron 59 (2008) 399–412, https://doi.org/10.1016/j.
the prefrontal cortex over the life course, Neuron 79 (2013) 16–29, https://doi. neuron.2008.06.023.
org/10.1016/j.neuron.2013.06.028. [100] P.T. Pang, G. Nagappan, W. Guo, B. Lu, Extracellular and intracellular cleavages
[74] L. Musazzi, G. Treccani, M. Popoli, Functional and structural remodeling of of proBDNF required at two distinct stages of late-phase LTP, NPJ Sci. Learn. 1
glutamate synapses in prefrontal and frontal cortex induced by behavioral stress, (2016) 16003, https://doi.org/10.1038/npjscilearn.2016.3.
Front. Psychiatry 6 (2015), https://doi.org/10.3389/fpsyt.2015.00060. [101] D. Panja, C.R. Bramham, BDNF mechanisms in late LTP formation: a synthesis and
[75] S. Ghosal, B.D. Hare, R.S. Duman, Prefrontal cortex GABAergic deficits and circuit breakdown, Neuropharmacology 76 (2014) 664–676, https://doi.org/10.1016/j.
dysfunction in the pathophysiology and treatment of chronic stress and neuropharm.2013.06.024.
depression, Curr. Opin. Behav. Sci. 14 (2017) 1–8, https://doi.org/10.1016/j. [102] M. Radahmadi, H. Alaei, M.R. Sharifi, N. Hosseini, Effects of different timing of
cobeha.2016.09.012. stress on corticosterone, BDNF and memory in male rats, Physiol. Behav. 139
[76] J.M. McKlveen, R.L. Morano, M. Fitzgerald, S. Zoubovsky, S.N. Cassella, J. (2015) 459–467, https://doi.org/10.1016/j.physbeh.2014.12.004.
R. Scheimann, S. Ghosal, P. Mahbod, B.A. Packard, B. Myers, M.L. Baccei, J. [103] M. Kojima, T. Mizui, BDNF Propeptide: A Novel Modulator of Synaptic Plasticity,
P. Herman, Chronic stress increases prefrontal inhibition: a mechanism for stress- 1st ed., Elsevier Inc., 2017 https://doi.org/10.1016/bs.vh.2016.11.006.
induced prefrontal dysfunction, Biol. Psychiatry 80 (2016) 754–764, https://doi. [104] M. Kojima, K. Matsui, T. Mizui, BDNF pro-peptide: physiological mechanisms and
org/10.1016/j.biopsych.2016.03.2101. implications for depression, Cell Tissue Res. (2019), https://doi.org/10.1007/
[77] I. Negrón-Oyarzo, F. Aboitiz, P. Fuentealba, Impaired functional connectivity in s00441-019-03034-6.
the prefrontal cortex: a mechanism for chronic stress-induced neuropsychiatric [105] K.P. Giese, K. Mizuno, The roles of protein kinases in learning and memory, Learn.
disorders, Neural Plast. 2016 (2016), https://doi.org/10.1155/2016/7539065. Mem. 20 (2013) 540–552, https://doi.org/10.1101/lm.028449.112.
[78] Y. Fu, S. Yu, X. Guo, X. Li, T. Li, H. Li, Y. Dong, Fluvoxamine increased glutamate [106] A.M. Graybiel, S.T. Grafton, The striatum: where skills and habits meet, Cold
release by activating both 5-HT 3 and sigma-1 receptors in prelimbic cortex of Spring Harb. Perspect. Biol. 7 (2015), https://doi.org/10.1101/cshperspect.
chronic restraint stress C57BL/6 mice, Biochim. Biophys. Acta - Mol. Cell Res. a021691.
(2012), https://doi.org/10.1016/j.bbamcr.2012.01.008. [107] K. Foerde, What are habits and do they depend on the striatum? A view from the
[79] S.L. Gourley, A.M. Swanson, A.J. Koleske, Corticosteroid-induced neural study of neuropsychological populations, Curr. Opin. Behav. Sci. 20 (2018)
remodeling predicts behavioral vulnerability and resilience, J. Neurosci. (2013), 17–24, https://doi.org/10.1016/j.cobeha.2017.08.011.
https://doi.org/10.1523/JNEUROSCI.2138-12.2013. [108] T.K. Patterson, B.J. Knowlton, Subregional specificity in human striatal habit
[80] P. Kim, G.W. Evans, M. Angstadt, S.S. Ho, C.S. Sripada, J.E. Swain, I. Liberzon, K. learning: a meta-analytic review of the fMRI literature, Curr. Opin. Behav. Sci. 20
L. Phan, Effects of childhood poverty and chronic stress on emotion regulatory (2018) 75–82, https://doi.org/10.1016/j.cobeha.2017.10.005.

9
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

[109] A.C. Bostan, P.L. Strick, The basal ganglia and the cerebellum: nodes in an [134] R. Dorey, C. Piérard, S. Shinkaruk, C. Tronche, F. Chauveau, M. Baudonnat,
integrated network, Nat. Rev. Neurosci. 19 (2018) 338–350, https://doi.org/ D. Béracochéa, Membrane mineralocorticoid but not glucocorticoid receptors of
10.1038/s41583-018-0002-7. the dorsal hippocampus mediate the rapid effects of corticosterone on memory
[110] A.S.R. Fermin, T. Yoshida, J. Yoshimoto, M. Ito, S.C. Tanaka, K. Doya, Model- retrieval, Neuropsychopharmacology 36 (2011) 2639–2649, https://doi.org/
based action planning involves cortico-cerebellar and basal ganglia networks, Sci. 10.1038/npp.2011.152.
Rep. 6 (2016), https://doi.org/10.1038/srep31378. [135] T.M. Schilling, M. Kölsch, M.F. Larra, C.M. Zech, T.D. Blumenthal, C. Frings,
[111] K.S. LaBar, R. Cabeza, Cognitive neuroscience of emotional memory, Nat. Rev. H. Schächinger, For whom the bell (curve) tolls: cortisol rapidly affects memory
Neurosci. 7 (2006) 54–64, https://doi.org/10.1038/nrn1825. retrieval by an inverted U-shaped dose-response relationship,
[112] B. Li, Central amygdala cells for learning and expressing aversive emotional Psychoneuroendocrinology 38 (2013) 1565–1572, https://doi.org/10.1016/j.
memories, Curr. Opin. Behav. Sci. 26 (2019) 40–45, https://doi.org/10.1016/j. psyneuen.2013.01.001.
cobeha.2018.09.012. [136] L. Schwabe, M. Joëls, B. Roozendaal, O.T. Wolf, M.S. Oitzl, Stress effects on
[113] P. Sah, Y. Sun, H. Gooch, Fear conditioning and the basolateral amygdala, memory: an update and integration, Neurosci. Biobehav. Rev. 36 (2012)
F1000Research 9 (2020) 1–8, https://doi.org/10.12688/f1000research.21201.1. 1740–1749, https://doi.org/10.1016/j.neubiorev.2011.07.002.
[114] R.L. Ressler, S. Maren, Synaptic encoding of fear memories in the amygdala, Curr. [137] L. Schwabe, O.T. Wolf, Timing matters: temporal dynamics of stress effects on
Opin. Neurobiol. 54 (2019) 54–59, https://doi.org/10.1016/j.conb.2018.08.012. memory retrieval, Cogn. Affect. Behav. Neurosci. 14 (2014) 1041–1048, https://
[115] K.S. Labar, J.C. Gatenby, J.C. Gore, J.E. Ledoux, E.A. Phelps, Human amygdala doi.org/10.3758/s13415-014-0256-0.
activation during conditioned fear acquisition and extinction: a mixed-trial fMRI [138] U. Rimmele, L. Besedovsky, T. Lange, J. Born, Blocking mineralocorticoid
study to investigate amygdala function in human populations have produced receptors impairs, blocking glucocorticoid receptors enhances memory retrieval
inconsistent results across techniques. Whereas neuropsychological studies have in humans, Neuropsychopharmacology 38 (2013) 884–894, https://doi.org/
repo, Neuron 20 (1998) 937–945. 10.1038/npp.2012.254.
[116] C. Büchel, J. Morris, R.J. Dolan, K.J. Friston, Brain systems mediating aversive [139] S.A. Gagnon, A.D. Wagner, Acute stress and episodic memory retrieval:
conditioning: an event-related fMRI study, Neuron 20 (1998) 947–957, https:// neurobiological mechanisms and behavioral consequences, Ann. N. Y. Acad. Sci.
doi.org/10.1016/S0896-6273(00)80476-6. 1369 (2016) 55–75, https://doi.org/10.1111/nyas.12996.
[117] G. Mertens, J. De Houwer, Potentiation of the startle reflex is in line with [140] M. Lindau, O. Almkvist, A.H. Mohammed, Effects of Stress on Learning and
contingency reversal instructions rather than the conditioning history, Biol. Memory, in: Stress Concepts, Cogn. Emot. Behav. Handb. Stress, Elsevier, 2016,
Psychol. 113 (2016) 91–99, https://doi.org/10.1016/j.biopsycho.2015.11.014. pp. 153–160, https://doi.org/10.1016/B978-0-12-800951-2.00018-2.
[118] S.O. King, C.L. Williams, Novelty-induced arousal enhances memory for cued [141] C. Sandi, M.T. Pinelo-Nava, Stress and memory: Behavioral effects and
classical fear conditioning: interactions between peripheral adrenergic and neurobiological mechanisms, Neural Plast. 2007 (2007) 20, https://doi.org/
brainstem glutamatergic systems, Learn. Mem. 16 (2009) 625–634, https://doi. 10.1155/2007/78970.
org/10.1101/lm.1513109. [142] A. Colciago, L. Casati, P. Negri-Cesi, F. Celotti, Learning and memory: Steroids
[119] T.F. Giustino, S. Maren, Noradrenergic modulation of fear conditioning and and epigenetics, J. Steroid Biochem. Mol. Biol. 150 (2015) 64–85, https://doi.
extinction, Front. Behav. Neurosci. 12 (2018) 1–20, https://doi.org/10.3389/ org/10.1016/j.jsbmb.2015.02.008.
fnbeh.2018.00043. [143] O.T. Wolf, P. Atsak, D.J. de Quervain, B. Roozendaal, K. Wingenfeld, Stress and
[120] G.S. Shields, M.A. Sazma, A.M. McCullough, A.P. Yonelinas, The effects of acute memory: a selective review on recent developments in the understanding of stress
stress on episodic memory: a meta-analysis and integrative review, Psychol. Bull. hormone effects on memory and their clinical relevance, J. Neuroendocrinol. 28
143 (2017) 636–675, https://doi.org/10.1037/bul0000100. (2016), https://doi.org/10.1111/jne.12353.
[121] B. Roozendaal, E.J. Hermans, Norepinephrine effects on the encoding and [144] A. Santori, P. Colucci, G.F. Mancini, M. Morena, M. Palmery, V. Trezza, S. Puglisi-
consolidation of emotional memory: improving synergy between animal and Allegra, M.N. Hill, P. Campolongo, Anandamide modulation of circadian- and
human studies, Curr. Opin. Behav. Sci. 14 (2017) 115–122, https://doi.org/ stress-dependent effects on rat short-term memory, Psychoneuroendocrinology
10.1016/j.cobeha.2017.02.001. 108 (2019) 155–162, https://doi.org/10.1016/j.psyneuen.2019.06.018.
[122] H.C. Schiff, J.P. Johansen, M. Hou, D.E.A. Bush, E.K. Smith, J.A.E. Klein, J. [145] K. Simon-Kutscher, N. Wanke, C. Hiller, L. Schwabe, Fear without context: acute
E. LeDoux, R.M. Sears, В-adrenergic receptors regulate the acquisition and stress modulates the balance of cue-dependent and contextual fear learning,
consolidation phases of aversive memory formation through distinct, temporally Psychol. Sci. 30 (2019) 1123–1135, https://doi.org/10.1177/
regulated signaling pathways, Neuropsychopharmacology (2017), https://doi. 0956797619852027.
org/10.1038/npp.2016.238. [146] T.J. Shors, Acute stress rapidly and persistently enhances memory formation in
[123] H. Villain, A. Benkahoul, A. Drougard, M. Lafragette, E. Muzotte, S. Pech, E. Bui, the male rat, Neurobiol. Learn. Mem. 75 (2001) 10–29, https://doi.org/10.1006/
A. Brunet, P. Birmes, P. Roullet, Effects of propranolol, a β-noradrenergic nlme.1999.3956.
antagonist, on memory consolidation and reconsolidation in mice, Front. Behav. [147] C.M. Raio, E.A. Phelps, The influence of acute stress on the regulation of
Neurosci. (2016), https://doi.org/10.3389/fnbeh.2016.00049. conditioned fear, Neurobiol. Stress 1 (2015) 134–146, https://doi.org/10.1016/j.
[124] B. Roozendaal, Stress and memory: Opposing effects of glucocorticoids on ynstr.2014.11.004.
memory consolidation and memory retrieval, Neurobiol. Learn. Mem. 78 (2002) [148] A.V. Aubry, P.A. Serrano, N.S. Burghardt, Molecular mechanisms of stress-
578–595, https://doi.org/10.1006/nlme.2002.4080. induced increases in fear memory consolidation within the amygdala, Front.
[125] T. Hatfield, J.L. McGaugh, Norepinephrine infused into the basolateral amygdala Behav. Neurosci. 10 (2016) 191, https://doi.org/10.3389/fnbeh.2016.00191.
posttraining enhances retention in a spatial water maze task, Neurobiol. Learn. [149] B. Roozendaal, S. Okuda, E.A. Van Der Zee, J.L. McGaugh, Glucocorticoid
Mem. 71 (1999) 232–239, https://doi.org/10.1006/nlme.1998.3875. enhancement of memory requires arousal-induced noradrenergic activation in the
[126] B. Roozendaal, N.A. Castello, G. Vedana, A. Barsegyan, J.L. McGaugh, basolateral amygdala, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 6741–6746,
Noradrenergic activation of the basolateral amygdala modulates consolidation of https://doi.org/10.1073/pnas.0601874103.
object recognition memory⋆, Neurobiol. Learn. Mem. 90 (2008) 576–579, [150] Atsaka, Friederike M. Guenzela, Deniz Kantar-Goka, Ioannis Zalachorase,
https://doi.org/10.1016/j.nlm.2008.06.010. Piraye Yargicoglud, Onno C. Meijere, Gina L. Quirartef, Oliver T. Wolfc,
[127] F.S. Maheu, R. Joober, S. Beaulieu, S.J. Lupien, Differential effects of adrenergic Lars Schwabeg, Benno Roozendaal, P. Atsak, F.M. Guenzel, D. Kantar-Gok,
and corticosteroid hormonal systems on human short- and long-term declarative I. Zalachoras, P. Yargicoglu, O.C. Meijer, G.L. Quirarte, O.T. Wolf, L. Schwabe,
memory for emotionally arousing material, Behav. Neurosci. (2004), https://doi. B. Roozendaal, Glucocorticoids mediate stress-induced impairment of retrieval of
org/10.1037/0735-7044.118.2.420. stimulus-response memory, Psychoneuroendocrinology 67 (2016) 207–215,
[128] H. Kunugi, H. Hori, N. Adachi, T. Numakawa, Interface between hypothalamic- https://doi.org/10.1016/j.psyneuen.2016.02.0060306-4530.
pituitary-adrenal axis and brain-derived neurotrophic factor in depression, [151] F.M. Guenzel, O.T. Wolf, L. Schwabe, Stress disrupts response memory retrieval,
Psychiatry Clin. Neurosci. 64 (2010) 447–459, https://doi.org/10.1111/j.1440- Psychoneuroendocrinology 38 (2013) 1460–1465, https://doi.org/10.1016/j.
1819.2010.02135.x. psyneuen.2012.12.010.
[129] T. Numakawa, E. Kumamaru, N. Adachi, Y. Yagasaki, A. Izumi, H. Kunugi, [152] Y. Zu, Q. Shan, X. Qin, Y. Sun, H. Fang, J. Zhou, Acute and chronic Stress exert
Glucocorticoid receptor interaction with TrkB promotes BDNF-triggered PLC-γ opposite effects on formation and contextualrelated fear conditioninq in rats,
signaling for glutamate release via a glutamate transporter, Proc. Natl. Acad. Sci. Neuroendocrinol. Lett. 40 (2019) 59–67.
U. S. A. 106 (2009) 647–652, https://doi.org/10.1073/pnas.0800888106. [153] A. Suvrathan, S. Bennur, S. Ghosh, A. Tomar, S. Anilkumar, S. Chattarji, Stress
[130] V. Di Liberto, M. Frinchi, V. Verdi, A. Vitale, F. Plescia, C. Cannizzaro, M. enhances fear by forming new synapses with greater capacity for long-term
F. Massenti, N. Belluardo, G. Mudò, Anxiolytic effects of muscarinic acetylcholine potentiation in the amygdala, Philos. Trans. R. Soc. B Biol. Sci. 369 (2014) 60–65,
receptors agonist oxotremorine in chronically stressed rats and related changes in https://doi.org/10.1098/rstb.2013.0151.
BDNF and FGF2 levels in the hippocampus and prefrontal cortex, [154] A.N. Hoffman, A. Parga, P.R. Paode, L.R. Watterson, E.M. Nikulina, R.P. Hammer,
Psychopharmacology (Berl.) 234 (2017) 559–573, https://doi.org/10.1007/ C.D. Conrad, Chronic stress enhanced fear memories are associated with
s00213-016-4498-0. increased amygdala zif268 mRNA expression and are resistant to reconsolidation,
[131] H. Lakshminarasimhan, S. Chattarji, Stress leads to contrasting effects on the Neurobiol. Learn. Mem. 120 (2015) 61–68, https://doi.org/10.1016/j.
levels of brain derived neurotrophic factor in the hippocampus and amygdala, nlm.2015.02.004.
PLoS One (2012), https://doi.org/10.1371/journal.pone.0030481. [155] P. Chakraborty, S. Chattarji, Timing is everything: differential effects of chronic
[132] W. Liu, T. Ge, Y. Leng, Z. Pan, J. Fan, W. Yang, R. Cui, The role of neural plasticity stress on fear extinction, Psychopharmacology (Berl.) 236 (2019) 73–86, https://
in depression: from Hippocampus to prefrontal cortex, Neural Plast. 2017 (2017), doi.org/10.1007/s00213-018-5053-y.
https://doi.org/10.1155/2017/6871089. [156] T. Hartley, N. Burgess, Complementary memory systems: competition,
[133] H. Qiao, M.X. Li, C. Xu, H. Bin Chen, S.C. An, X.M. Ma, Dendritic spines in cooperation and compensation, Trends Neurosci. (2005), https://doi.org/
depression: what we learned from animal models, Neural Plast. 2016 (2016) 10.1016/j.tins.2005.02.004.
20–24, https://doi.org/10.1155/2016/8056370.

10
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

[157] E.V. Goldfarb, E.A. Phelps, Stress and the trade-off between hippocampal and mental rotation, J. Affect. Disord. 235 (2018) 277–284, https://doi.org/10.1016/
striatal memory, Curr. Opin. Behav. Sci. 14 (2017) 47–53, https://doi.org/ j.jad.2018.04.006.
10.1016/j.cobeha.2016.11.017. [181] M. Schecklmann, T. Dresler, S. Beck, J.T. Jay, R. Febres, J. Haeusler, T.A. Jarczok,
[158] R.A. Poldrack, M.G. Packard, Competition among multiple memory systems: A. Reif, M.M. Plichta, A.C. Ehlis, A.J. Fallgatter, Reduced prefrontal oxygenation
converging evidence from animal and human brain studies, Neuropsychologia during object and spatial visual working memory in unpolar and bipolar
(2003), https://doi.org/10.1016/S0028-3932(02)00157-4. depression, Psychiatry Res. - Neuroimaging. 194 (2011) 378–384, https://doi.
[159] L. Schwabe, O.T. Wolf, Stress-induced modulation of instrumental behavior: from org/10.1016/j.pscychresns.2011.01.016.
goal-directed to habitual control of action, Behav. Brain Res. 219 (2011) [182] Y. Chen, J. Liu, Z. Li, B. Liu, Y. Ji, Y. Ju, H. Fang, Q. Zheng, M. Wang, W. Guo,
321–328, https://doi.org/10.1016/j.bbr.2010.12.038. H. Li, X. Lu, L. Li, The tendency of modified electroconvulsive therapy-related
[160] E. Dias-Ferreira, J.C. Sousa, I. Melo, P. Morgado, A.R. Mesquita, J.J. Cerqueira, R. working memory and subjective memory deficits in depression: a prospective
M. Costa, N. Sousa, Chronic stress causes frontostriatal reorganization and affects follow-up study, J. ECT 36 (2020) 198–204, https://doi.org/10.1097/
decision-making, Science (80-.) 325 (2009) 621–625, https://doi.org/10.1126/ YCT.0000000000000668.
science.1171203. [183] D. Downey, S. Brigadoi, L. Trevithick, R. Elliott, C. Elwell, R.H. McAllister-
[161] A. Friedman, D. Homma, B. Bloem, L.G. Gibb, K. ichi Amemori, D. Hu, Williams, I.M. Anderson, Frontal haemodynamic responses in depression and the
S. Delcasso, T.F. Truong, J. Yang, A.S. Hood, K.A. Mikofalvy, D.W. Beck, effect of electroconvulsive therapy, J. Psychopharmacol. 33 (2019) 1003–1014,
N. Nguyen, E.D. Nelson, S.E. Toro Arana, R.H. Vorder Bruegge, K.A. Goosens, A. https://doi.org/10.1177/0269881119858313.
M. Graybiel, Chronic stress alters striosome-circuit dynamics, leading to aberrant [184] T.M. Le, J.A. Borghi, A.J. Kujawa, D.N. Klein, H.C. Leung, Alterations in visual
decision-making, Cell 171 (2017), https://doi.org/10.1016/j.cell.2017.10.017, cortical activation and connectivity with prefrontal cortex during working
1191.e28-1192. memory updating in major depressive disorder, Neuroimage Clin. 14 (2017)
[162] J. Goodman, K.C. Leong, M.G. Packard, Glucocorticoid enhancement of 43–53, https://doi.org/10.1016/j.nicl.2017.01.004.
dorsolateral striatum-dependent habit memory requires concurrent noradrenergic [185] E. Begovic, V. Panaite, L.M. Bylsma, C. George, M. Kovacs, I. Yaroslavsky, I. Baji,
activity, Neuroscience 311 (2015) 1–8, https://doi.org/10.1016/j. I. Benák, R. Dochnal, E. Kiss, Á. Vetró, K. Kapornai, J. Rottenberg, Positive
neuroscience.2015.10.014. autobiographical memory deficits in youth with depression histories and their
[163] B. Roozendaal, B.S. McEwen, S. Chattarji, Stress, memory and the amygdala, Nat. never-depressed siblings, Br. J. Clin. Psychol. 56 (2017) 329–346, https://doi.
Rev. Neurosci. 10 (2009) 423–433, https://doi.org/10.1038/nrn2651. org/10.1111/bjc.12141.
[164] K. Sriram, M. Rodriguez-Fernandez, F.J. Doyle, Modeling cortisol dynamics in the [186] D.G. Dillon, D.A. Pizzagalli, Mechanisms of memory disruption in depression,
neuro-endocrine axis distinguishes normal, depression, and post-traumatic stress Trends Neurosci. 41 (2018) 137–149, https://doi.org/10.1016/j.
disorder (PTSD) in humans, PLoS Comput. Biol. 8 (2012), https://doi.org/ tins.2017.12.006.
10.1371/journal.pcbi.1002379. [187] I.B.M. Bento de Souza, F.F. Barbosa, A.M. Lacerda, N.A. dos Santos, N. Torro-
[165] P.W. Gold, The organization of the stress system and its dysregulation in Alves, Evaluation of facial expressions in women with major depression: is there a
depressive illness, Mol. Psychiatry 20 (2015) 32–47, https://doi.org/10.1038/ negative bias? Psychol. Neurosci. (2014) https://doi.org/10.3922/j.
mp.2014.163. psns.2014.4.10.
[166] J. Keller, R. Gomez, G. Williams, A. Lembke, L. Lazzeroni, G.M. Murphy, A. [188] K.D. Young, G.J. Siegle, J. Bodurka, W.C. Drevets, Amygdala activity during
F. Schatzberg, HPA axis in major depression: cortisol, clinical symptomatology autobiographical memory recall in depressed and vulnerable individuals:
and genetic variation predict cognition, Mol. Psychiatry 22 (2017) 527–536, association with symptom severity and autobiographical overgenerality, Am. J.
https://doi.org/10.1038/mp.2016.120. Psychiatry 173 (2016) 78–89, https://doi.org/10.1176/appi.ajp.2015.15010119.
[167] K.S. Kendler, L.M. Karkowski, C.A. Prescott, Causal relationship between stressful [189] S. Boku, S. Nakagawa, H. Toda, A. Hishimoto, Neural basis of major depressive
life events and the onset of major depression, Am. J. Psychiatry (1999), https:// disorder: beyond monoamine hypothesis, Comput. Graph. Forum 37 (2018) 3–12,
doi.org/10.1176/ajp.156.6.837. https://doi.org/10.1111/pcn.12604.
[168] P.L. Rock, J.P. Roiser, W.J. Riedel, A.D. Blackwell, Cognitive impairment in [190] N. Nikkheslat, C.M. Pariante, P.A. Zunszain, Neuroendocrine Abnormalities in
depression: a systematic review and meta-analysis, Psychol. Med. 44 (2014) Major Depression: An Insight Into Glucocorticoids, Cytokines, and the Kynurenine
2029–2040, https://doi.org/10.1017/S0033291713002535. Pathway, Chapter 3, Elsevier Inc., 2018, https://doi.org/10.1016/B978-0-12-
[169] P.J. Lucassen, C.A. Oomen, M. Schouten, J.M. Encinas, C.P. Fitzsimons, Adult 811073-7.00003-9.
Neurogenesis, Chronic Stress and Depression, Elsevier Inc., 2016, https://doi.org/ [191] S. Kinoshita, T. Kanazawa, H. Kikuyama, H. Yoneda, Clinical application of DEX/
10.1016/B978-0-12-801977-1.00008-8. CRH test and multi-channel NIRS in patients with depression, Behav. Brain Funct.
[170] T. Toda, S.L. Parylak, S.B. Linker, F.H. Gage, The role of adult hippocampal (2016), https://doi.org/10.1186/s12993-016-0108-x.
neurogenesis in brain health and disease, Mol. Psychiatry (2018) 1–21, https:// [192] A.C. de, M. Galvão, R.N. de Almeida, E.A. do, S. Silva, F.A.M. Freire, F. Palhano-
doi.org/10.1038/s41380-018-0036-2. Fontes, H. Onias, E. Arcoverde, J.P. Maia-de-Oliveira, D.B. de Araújo, B. Lobão-
[171] N. Ridout, B. Dritschel, K. Matthews, R. O’Carroll, Autobiographical memory Soares, N.L. Galvão-Coelho, Cortisol modulation by ayahuasca in patients with
specificity in response to verbal and pictorial cues in clinical depression, J. Behav. treatment resistant depression and healthy controls, Front. Psychiatry 9 (2018)
Ther. Exp. Psychiatry 51 (2016) 109–115, https://doi.org/10.1016/j. 1–10, https://doi.org/10.3389/fpsyt.2018.00185.
jbtep.2016.01.002. [193] H. Kunugi, H. Hori, S. Ogawa, Biochemical markers subtyping major depressive
[172] M. Talarowska, A. Florkowski, K. Zboralski, D. Berent, P. Wierzbiński, P. Gałecki, disorder, Psychiatry Clin. Neurosci. 69 (2015) 597–608, https://doi.org/
Auditory-verbal declarative and operating memory among patients suffering from 10.1111/pcn.12299.
depressive disorders - preliminary study, Adv. Med. Sci. 55 (2010) 317–327, [194] D. Cicchetti, F.A. Rogosch, M.L. Howe, S.L. Toth, The effects of maltreatment and
https://doi.org/10.2478/v10039-010-0053-0. neuroendocrine regulation on memory performance, Child Dev. 81 (2010)
[173] F. Pauls, F. Petermann, A.C. Lepach, Episodic memory and executive functioning 1504–1519, https://doi.org/10.1111/j.1467-8624.2010.01488.x.
in currently depressed patients compared to healthy controls, Cogn. Emot. 29 [195] J. Tiemensma, C.D. Andela, N.R. Biermasz, J.A. Romijn, A.M. Pereira, Mild
(2015) 383–400, https://doi.org/10.1080/02699931.2014.915208. cognitive deficits in patients with primary adrenal insufficiency,
[174] E. Ahern, M. Semkovska, Cognitive functioning in the first-episode of major Psychoneuroendocrinology 63 (2016) 170–177, https://doi.org/10.1016/j.
depressive disorder: a systematic review and meta-analysis, Neuropsychology 31 psyneuen.2015.09.029.
(2017) 52–72, https://doi.org/10.1037/neu0000319. [196] E. Resmini, A. Santos, B. Gómez-Anson, Y. Vives, P. Pires, I. Crespo, M.J. Portella,
[175] D.M. Barch, M.P. Harms, R. Tillman, E. Hawkey, J.L. Luby, Early childhood M. De Juan-Delago, M.J. Barahona, S.M. Webb, Verbal and visual memory
depression, emotion regulation, episodic memory, and hippocampal performance and hippocampal volumes, measured by 3-tesla magnetic resonance
development, J. Abnorm. Psychol. 128 (2019) 81–95, https://doi.org/10.1037/ imaging, in patients with Cushing’s syndrome, J. Clin. Endocrinol. Metab. 97
abn0000392. (2012) 663–671, https://doi.org/10.1210/jc.2011-2231.
[176] S.M. Cinini, G.F. Barnabe, N.L. Galvão-Coelho, M.A. de Medeiros, P. Perez- [197] M. Piasecka, E. Papakokkinou, E. Valassi, A. Santos, S.M. Webb, F. de Vries, A.
Mendes, M.B.C. de Sousa, L. Covolan, L.E. Mello, Social isolation disrupts M. Pereira, O. Ragnarsson, Psychiatric and neurocognitive consequences of
hippocampal neurogenesis in young non-human primates, Front. Neurosci. endogenous hypercortisolism, J. Intern. Med. 288 (2020) 168–182, https://doi.
(2014), https://doi.org/10.3389/fnins.2014.00045. org/10.1111/joim.13056.
[177] J. Radley, D. Morilak, V. Viau, S. Campeau, Chronic stress and brain plasticity: [198] M.L. Molendijk, P. Spinhoven, M. Polak, B.A.A. Bus, B.W.J.H. Penninx, B.
mechanisms underlying adaptive and maladaptive changes and implications for M. Elzinga, Serum BDNF concentrations as peripheral manifestations of
stress-related CNS disorders, Neurosci. Biobehav. Rev. 58 (2015) 79–91, https:// depression: evidence from a systematic review and meta-analyses on 179
doi.org/10.1016/j.neubiorev.2015.06.018. associations (N=9484), Mol. Psychiatry (2014), https://doi.org/10.1038/
[178] K.D. Young, P.S.F. Bellgowan, J. Bodurka, W.C. Drevets, Autobiographical deficits mp.2013.105.
correlate with gray matter volume in depressed and high risk participants, Soc. [199] R.N. de Almeida, A.C. de, M. Galvão, F.S. da Silva, E.A. dos, S. Silva, F. Palhano-
Cogn. Affect. Neurosci. 10 (2014) 1588–1595, https://doi.org/10.1093/scan/ Fontes, J.P. Maia-de-Oliveira, D.B. de Araújo, B. Lobão-Soares, N.L. Galvão-
nsv047. Coelho, Modulation of serum brain-derived neurotrophic factor by a single dose
[179] R.S. Høifødt, K. Waterloo, C.E.A. Wang, M. Eisemann, Y. Figenschau, of ayahuasca: observation from a randomized controlled trial, Front. Psychol.
M. Halvorsen, Cortisol levels and cognitive profile in major depression: a (2019), https://doi.org/10.3389/fpsyg.2019.01234.
comparison of currently and previously depressed patients, [200] C. Björkholm, L.M. Monteggia, BDNF - A key transducer of antidepressant effects,
Psychoneuroendocrinology 99 (2019) 57–65, https://doi.org/10.1016/j. Neuropharmacology 102 (2016) 72–79, https://doi.org/10.1016/j.
psyneuen.2018.08.024. neuropharm.2015.10.034.
[180] C. Oshiyama, C. Sutoh, H. Miwa, S. Okabayashi, H. Hamada, D. Matsuzawa, [201] C.J. Harmer, R.S. Duman, P.J. Cowen, How do antidepressants work? New
Y. Hirano, T. Takahashi, S. ichi Niwa, M. Honda, K. Sakatsume, T. Nishimura, perspectives for refining future treatment approaches, Lancet Psychiatry 4 (2017)
E. Shimizu, Gender-specific associations of depression and anxiety symptoms with 409–418, https://doi.org/10.1016/S2215-0366(17)30015-9.

11
G.M. Sousa Júnior et al. Behavioural Brain Research 412 (2021) 113410

[202] A. Piccinni, D. Marazziti, M. Catena, L. Domenici, A. Del Debbio, C. Bianchi, [209] F. Palhano-Fontes, K.C. Andrade, L.F. Tofoli, A.C.S. Jose, A.S. Crippa, J.E.
C. Mannari, C. Martini, E. Da Pozzo, E. Schiavi, A. Mariotti, I. Roncaglia, A. Palla, C. Hallak, S. Ribeiro, D.B. de Araújo, The psychedelic state induced by Ayahuasca
G. Consoli, L. Giovannini, G. Massimetti, L. Dell’Osso, Plasma and serum brain- modulates the activity and connectivity of the default mode network, PLoS One
derived neurotrophic factor (BDNF) in depressed patients during 1 year of 10 (2015) 1–13, https://doi.org/10.1371/journal.pone.0118143.
antidepressant treatments, J. Affect. Disord. 105 (2008) 279–283, https://doi. [210] R.G. dos Santos, J.E.C. Hallak, Effects of the natural β-carboline alkaloid harmine,
org/10.1016/j.jad.2007.05.005. a main constituent of Ayahuasca, in memory and in the Hippocampus: a
[203] S.A. Stuart, P. Butler, M.R. Munafò, D.J. Nutt, E.S.J. Robinson, Distinct systematic literature review of preclinical studies, J. Psychoactive Drugs 49
neuropsychological mechanisms may explain delayed-versus rapid-onset (2017) 1–10, https://doi.org/10.1080/02791072.2016.1260189.
antidepressant efficacy, Neuropsychopharmacology (2015), https://doi.org/ [211] J.A. Morales-García, M. De La Fuente Revenga, S. Alonso-Gil, M.I. Rodríguez-
10.1038/npp.2015.59. Franco, A. Feilding, A. Perez-Castillo, J. Riba, The alkaloids of Banisteriopsis
[204] I.B.M.B. de Souza, Y. da, S.R. Meurer, P.M. Tavares, K.C. Pugliane, R.H. Lima, R. caapi, the plant source of the Amazonian hallucinogen Ayahuasca, stimulate adult
H. Silva, F.F. Barbosa, Episodic-like memory impairment induced by sub- neurogenesis in vitro, Sci. Rep. 7 (2017) 1–13, https://doi.org/10.1038/s41598-
anaesthetic doses of ketamine, Behav. Brain Res. 359 (2019) 165–171, https:// 017-05407-9.
doi.org/10.1016/j.bbr.2018.10.031. [212] M. Kargar, S. Askari, A. Khoshaman, A. Mohammadi, Differential diagnosis of
[205] F. de, L. Osório, L. Ribeiro, H. De Macedo, J. Quevedo, J. Alexandre, D.S. Crippa, schizophrenia and schizoaffective disorder from normal subjects using virtual
The therapeutic potential of harmine and ayahuasca in depression : evidence from reality, Psychiatry Res. 273 (2019) 378–386, https://doi.org/10.1016/j.
exploratory animal and human studies, Ethnopharmacol. Ayahuasca 661 (2011) psychres.2019.01.037.
75–85. http://srv1.iceers.org/docs/science/ayahuasca/Rafael_Guimares_TheEthn [213] A. Mohammadi, E. Hesami, M. Kargar, J. Shams, Detecting allocentric and
opharmacology_of_Ayahuasca/EthnopharmAyahuasca_Chapter5.pdf. egocentric navigation deficits in patients with schizophrenia and bipolar disorder
[206] R.F. Sanches, F. De Lima Osório, R.G. Santos, L.R.H. Macedo, J.P. Maia-De- using virtual reality, Neuropsychol. Rehabil. 28 (2018) 398–415, https://doi.org/
Oliveira, L. Wichert-Ana, D.B. de Araújo, J. Riba, J.A.S. Crippa, J.E.C. Hallak, 10.1080/09602011.2017.1369888.
Antidepressant effects of a single dose of ayahuasca in patients with recurrent [214] S.K. Gill, R.G. Gomez, J. Keller, A.F. Schatzberg, Diagnostic differences in verbal
depression a SPECT study, J. Clin. Psychopharmacol. 36 (2016) 77–81, https:// learning strategies and verbal memory in patients with mood disorders and
doi.org/10.1097/JCP.0000000000000436. psychotic disorders, Psychiatry Res. 269 (2018) 733–739, https://doi.org/
[207] F. Palhano-Fontes, D. Barreto, H. Onias, K.C. Andrade, M.M. Novaes, J.A. Pessoa, 10.1016/j.psychres.2018.09.003.
S.A. Mota-Rolim, F.L. Osório, R. Sanches, R.G. dos Santos, L.F. Tófoli, G. de [215] C.R. Brewin, Memory and forgetting, Curr. Psychiatry Rep. 20 (2018), https://
Oliveira Silveira, M. Yonamine, J. Riba, F.R. Santos, A.A. Silva-Junior, J. doi.org/10.1007/s11920-018-0950-7.
C. Alchieri, N.L. Galvão-Coelho, B. Lobão-Soares, J.E.C. Hallak, E. Arcoverde, J. [216] S. Kida, Reconsolidation/destabilization, extinction and forgetting of fear
P. Maia-de-Oliveira, D.B. Araújo, Rapid antidepressant effects of the psychedelic memory as therapeutic targets for PTSD, Psychopharmacology (Berl.) 236 (2019)
ayahuasca in treatment-resistant depression: a randomized placebo-controlled 49–57, https://doi.org/10.1007/s00213-018-5086-2.
trial, Psychol. Med. 49 (2019) 655–663, https://doi.org/10.1017/ [217] A.D. Askelund, S. Schweizer, I.M. Goodyer, A.L. van Harmelen, Positive memory
S0033291718001356. specificity is associated with reduced vulnerability to depression, Nat. Hum.
[208] N.L. Galvão-Coelho, A.C. de, M. Galvão, R.N. de Almeida, F. Palhano-Fontes, Behav. 3 (2019) 265–273, https://doi.org/10.1038/s41562-018-0504-3.
I. Campos Braga, B. Lobão Soares, J.P. Maia-de-Oliveira, D. Perkins, J. Sarris, D. [218] S. Ramirez, X. Liu, C.J. MacDonald, A. Moffa, J. Zhou, R.L. Redondo,
B. de Araújo, Changes in inflammatory biomarkers are related to the S. Tonegawa, Activating positive memory engrams suppresses depression-like
antidepressant effects of Ayahuasca, J. Psychopharmacol. (2020), https://doi. behaviour, Nature 522 (2015) 335–339, https://doi.org/10.1038/nature14514.
org/10.1177/0269881120936486, 026988112093648.

12

You might also like