Abstract
Exposure to chronic stress contributes considerably to the development of cognitive impairments in psychiatric disorders such as depression, generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), post-traumatic stress disorder (PTSD), and addictive behavior. Unfortunately, unlike mood-related symptoms, cognitive impairments are not effectively treated by available therapies, a situation in part resulting from a still incomplete knowledge of the neurobiological substrates that underly cognitive domains and the difficulty in generating interventions that are both efficacious and safe.
In this review, we will present an overview of the cognitive domains affected by stress with a specific focus on cognitive flexibility, behavioral inhibition, and working memory. We will then consider the effects of stress on neuronal correlates of cognitive function and the factors which may modulate the interaction of stress and cognition. Finally, we will discuss intervention strategies for treatment of stress-related disorders and gaps in knowledge with emerging new treatments under development.
Understanding how cognitive impairment occurs during exposure to chronic stress is crucial to make progress towards the development of new and effective therapeutic approaches.
Keywords: Executive function, Stress, Therapy
List of abbreviations
- HPA axis
hypothalamic-pituitary-adrenal axis
- GCs
Glucocorticoids
- CORT
corticosterone, cortisol
- CRH/CRF
corticotropin-releasing hormone/factor
- ACTH
adrenocorticotropic hormone
- GR
glucocorticoid receptor
- MR
mineralocorticoid receptor
- E
epinephrine
- NE
norepinephrine
- CNTF
ciliary neurotrophic factor
- JAK
Janus kinase
- SSRIs
elective serotonin reuptake inhibitors
- PCP
phencyclidine
- MDMA
3,4-methylenedioxymethamphetamine
- ACC
anterior cingulate cortex
- PVN
paraventricular nucleus of the hypothalamus
- LC
locus coeruleus
- RVLM
rostral ventrolateral medulla
- NTS
nucleus of the solitary tract
- IML
dorsal intermediolateral cellular column
- mPFC
medial prefrontal cortex
- DLPFC
dorsolateral prefrontal cortex
- OFC
orbitofrontal cortex
- NAC
nucleus accumbens
- BLA
basolateral amygdala
- PH
posterior hypothalamus
- STN
subthalamic nucleus
- MDT
mediodorsal thalamus
- CIC stress
chronic intermittent cold stress
- CMS
chronic mild stress
- CUS
chronic unpredictable stress
- PTSD
post-traumatic stress disorder
- AST
attentional set-shifting test
- ED
extra-dimensional
- SSRTT
stop signal reaction time test
- 5-CSRTT
5-choice serial reaction time test
- CANTAB
Cambridge Neuropsychological Test Automated Battery
- WCST
Wisconsin Card test
- CBT
Cognitive Behavior Therapy
- TMS
Transcranial Magnetic Stimulation
- rTMS
repetitive TMS
- tDCS
Transcranial Direct Current Stimulation
- TRD
Treatment Resistant Depression
- DREADD
designer receptor exclusively activated by designer drugs
- SERT
serotonin transporter
- LTP
long-term potentiation
- LTD
long-term depression
- NF1
neurofibromatosis 1
- OCD
obsessive-compulsive disorder
- PTSD
post-traumatic stress disorder
- GAD
generalized anxiety disorder
1. Introduction
Stress is a prominent part of modern life. Humans, like other species, have evolved adaptive mechanisms to limit the physiological or psychological impact of stress. However, exposure to traumatic or cumulative stressors can considerably contribute to the development of psychiatric disorders such as, depression, generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), post-traumatic stress disorder (PTSD), and addictive behavior (Adams et al., 2018; Maeng and Milad 2017; Patriquin and Mathew 2017; Siegrist 2008; Sinha 2008). Indeed, a recent article reported that cases of depression alone were 3-fold higher during the COVID-19 pandemic compared to pre-pandemic data (Ettman et al., 2020).
Cognitive impairment is a transdiagnostic domain shared by multiple psychiatric disorders, including stress-related disorders (Millan et al., 2012); for example, abnormalities in various cognitive functions such as cognitive flexibility, working memory and behavioral inhibition have been documented in depression, PTSD and substance use disorder (Dossi et al., 2020; Ramey and Regier 2019; Snyder 2013), and a large body of evidence suggests that stress exacerbates cognitive impairments (Lupien et al., 2009). Unfortunately, unlike mood-related symptoms, cognitive impairments are not effectively treated by currently available therapies, and large gaps still exist in both identifying the neurobiological substrates that underly cognitive domains and in developing effective interventions.
In this review, we will present an overview of the cognitive domains affected by stress with a specific focus on cognitive flexibility, behavioral inhibition, and working memory. We will then specify how these cognitive domains are affected by acute and chronic stress (see Table 1 for a summary of relevant literature). Sex differences will be discussed if reported in the original paper, however, for an in-depth analysis of the interaction of sex and stress on cognitive function we refer the reader to recent focused reviews (Bangasser and Kawasumi 2015; Orsini et al., 2022; Swaab and Bao 2020; Wellman et al., 2020). Finally, we will discuss intervention strategies for treatment of stress-related disorders and gaps in knowledge with emerging new treatments under development.
Table 1.
List of studies describing effects of stress on cognitive domains examined in this review.
Rodent studies | ||||||
---|---|---|---|---|---|---|
Cognitive domain | Stress type | Stress paradigm | Sex/species | Stress effect | Sex difference | References |
Reversal learning | Acute | 30 min restraint | Male rats | Facilitation | Bryce and Howland, 2015 | |
Acute | 10 min swim, 3d | Male mice | Facilitation | Graybeal et al., 2011 | ||
Acute | 30 min restraint | Male rats | Facilitation | Thai et al., 2013 | ||
Early life | Fragmented maternal care | Male and female rats | No effect in males, deficit in females | Y | Goodwill et al., 2018 | |
Chronic | 6h/14d cold stress | Male rats | Deficit | Danet et al., 2010, Donegan et al., 2014, Furr et al., 2012, Lapiz-Bluhm et al., 2009, Patton et al., 2017, Wallace et al., 2014 | ||
Chronic | 14-21d unpredictable mild stress | Male rats | Deficit | Bondi et al., 2007, Bondi et al., 2010, Jett et al., 2013, Jett et al., 2015, Hill et al., 2005,Quan et al., 2011, Yu et al., 2016 | ||
Set-shifting | Acute | 15 min tail pinch | Male rats | Deficit | Butts et al., 2013 | |
Acute | 30 min restraint | Male rats | No effect | Thai et al., 2013 | ||
Acute | Single prolonged stress | Male rats | Deficit | George et al., 2015 | ||
Chronic | 6h/14d cold stress | Male rats | No effect | Lapiz-Bluhm et al., 2009 | ||
Chronic | 21d repeated restraint | Male rats | Deficit | Liston et al., 2006 | ||
Chronic | 14d unpredictable mild stress | Male rats | Deficit | Bondi et al., 2008, Fucich et al., 2016, Jett et al., 2013, Morilak et al., 2005, | ||
Behavioral inhibition | Acute | Footshock on 2 consecutive days | Male rats | No effect on impulsive action | Girotti et al., 2022 | |
Early life | 5d variable stress | Male and female rats | Increased cocaine-induced impulsive action in females | Y | Paine et al., 2021 | |
CORT exposure in early life | 20 d corticosterone administration | Male rats | Reduced impulsive action but increased impulsive choice | Torregrossa et al., 2012 | ||
Chronic | 6h/14d cold stress | Male rats | Increase impulsive action in well trained task | Girotti et al., 2022 | ||
Chronic | 14d unpredictable mild stress | Male rats | Increase impulsive action in well trained and novel tasks | Girotti et al., 2022 | ||
Working memory | Acute | 60 min restraint | Male and female rats | Deficit in females | Y | Shansky et al., 2006 |
Acute | 20 min swim stress | Male rats | Enhancement | Yuen et al., 2009, Yuen et al., 2011 | ||
Acute | 40 min intermittent footshock | Male rats | Enhancement soon after stress, deficit at later times | Musazzi et al., 2019 | ||
Early life | Maternal separation | Male rats | Deficit in adults | Banqueri et al., 2021 | ||
Early life | Maternal separation | Male rats | Deficit in adolescence | Brenhouse and Andersen, 2011 | ||
Early life | Maternal separation | Male rats | Deficit in adults | Sanchez et al., 2021 | ||
Early life | Maternal separation | Male mice | Deficit in adults | Viola et al., 2019 | ||
Early life | Maternal separation | Male and female rats | No effect in males, slight deficit in females | Y | Sun et al., 2020 | |
Chronic | 4 weeks unpredictable mild stress | Male rats | Deficit | Cerqueira et al., 2007a | ||
Chronic | 21d repeated restraint | Male rats | Deficit | Wright and Conrad, 2008 | ||
Chronic | 28d repeated restraint | Male rats | Deficit | Mika et al., 2012 | ||
Chronic | 7d repeated restraint | Male and female rats | Deficits in males, no effect in females | Y | Wei et al., 2014 |
Human studies | ||||||
---|---|---|---|---|---|---|
Cognitive domain | Stress type | Stress paradigm | Sex examined | Stress effect | Sex difference | References |
Reversal learning | Acute | TSST | Males | Facilitation | Wieland et al., 2023 | |
Acute | CPT | Males and females | Impairment | Not reported | Raio et al., 2017 | |
Early + Acute | Early life stress + TSST | Males and females | Increased perseveration | Not reported | Franco and Knowlton, 2023 | |
Early life | History of medical and mental problems | Males and females adolescents | Deficit | Not reported | Harms et al., 2018 | |
Chronic | Perceived chronic stress | Females | Deficit | Monni et al., 2023 | ||
Set-shifting | Acute | TSST | Males | No effect | Hendrawan et al., 2012 | |
Acute | "Virtual" TSST | Males and females | Improved flexibility | Not reported | Delahaye et al., 2015 | |
Acute | TSST | Males and females | Deficit in males, no effect in females | Y | Shields et al., 2016 | |
Acute | CPT | Males and females | Deficit in males, no effect in females | Y | Kalia et al., 2018 | |
Acute | TSST | Males and females | Deficit | Not reported | Alexander et al., 2007 | |
Acute | Noise stress | Males and females | Deficit | Not reported | Hillier et al., 2006 | |
Chronic | Perceived chronic stress | Males and females | Deficit | Not reported | Orem et al., 2008 | |
Early life + Chronic | Perceived chronic stress | Males and females | Deficit | Not reported | Kalia and Knauft, 2020 | |
Early life + Chronic | Perceived chronic stress | Males and females | Deficit | Not reported | Kalia et al., 2021 | |
Behavioral inhibition | Acute | CPT | Males and females | Increase gambling as a function of trait impulsivity | Not reported | Canale et al., 2017 |
Acute | CPT | Males and females | Increase decision speed | Not reported | Raio et al., 2020 | |
Acute | Anticipate a videotape speech | Males | Increase choice for later, larger rewards | Lempert et al., 2012 | ||
Chronic | Lifetime stress exposure questionnaire | Males and females | Higher stress correlated with higher implsivity and food addiction | Not reported | McMullin et al., 2021 | |
Chronic | Lifetime stress exposure questionnaire | Males and females | Higher impulsivity and lifetime stress in metamphetamine users | Not reported | Mahoney et al., 2015 | |
Working memory | Acute | TSST | Males | Deficit | Luethi et al., 2008 | |
Acute | TSST | Males | Deficit | Schoofs et al., 2008 | ||
Acute | CPT | Males | Deficit | Schoofs et al., 2009 | ||
Acute | Hydrocortisone administration | Males | Deficit | Lupien et al., 1999 | ||
Acute | CPT | Males and females | No effect | Not reported | Porcelli et al., 2008 | |
Acute | TSST | Males | Slight improvement | Weerda et al., 2010 | ||
Acute | TSST | Males and females | Enhanced working memory in males | Y | Cornelisse et al., 2011b | |
Acute | Video of aversive images | Females | No effect | Qin et al., 2009 | ||
Acute | TSST | Males and females | Enhanced working memory in males, impairment in females | Y | Scoriels et al., 2013 |
Abbreviations: CPT, cold pressor test; TSST, Trier Social Stress Test.
Overall, it is vital to understand the relationship between the effects of stress on the brain and how cognitive impairment occurs during exposure to chronic stress. Through better understanding of mechanisms underlying the relationship between stress and cognition, new avenues may appear towards the development of new therapeutic approaches.
2. A brief overview of stress neurobiology
The neuroendocrine stress response is a remarkably well-conserved homeostatic process from amphibians to mammals, a fact that underscores its essential role in species survival and adaptation (Sapolsky 2021). Indeed, the response to stress is fundamentally an adaptive phenomenon directed toward the reallocation of physiological resources in response to an external or internal stimulus that has threatened homeostasis. This process of active adaptation through mobilization of neuroendocrine and immune mechanisms has been called allostasis (Sterling and Eyer, 1988. Allostatic load refers to the cost of this rebalancing process for the organism. In situations of acute or sporadic stress exposure, the cost is low and transient. However, in situations where either the stressor is persistent or the organism is debilitated, the prolonged engagement or overstimulation of allostatic systems causes a physiological burden that may lead to disease (McEwen 1998).
As an example, in socially complex species such as humans and non-human primates, the stress response can be activated and sustained in the absence of an “actual” contingent stressor, in part as a reaction to negative past experiences or in anticipation of negative future events. Such chronic engagement of the stress response system has been associated with an array of health disturbances, including cardiovascular, immunological, and reproductive dysfunction and increased incidence of stress-related psychiatric disorders (Sapolsky 2021). Thus, the current view is that stress-related pathologies develop from the unnecessary, excessive, or over-lasting activation of the stress response system that takes a toll on organism physiology. Fortunately, given the conservation of the stress response across species, it is possible to investigate the mechanisms of adaptive and maladaptive stress response, including effects on cognitive function, in animal models designed for high translational validity.
2.1. Physiological response to stress
For reasons of space, we will only briefly introduce the basic concepts of autonomic and neuroendocrine responses to stress. We refer the interested reader to comprehensive reviews on the subject (Carrier et al., 2021; Godoy et al., 2018; Gray et al., 2017; Hassamal 2023; McEwen and Akil 2020).
Acute stress can be defined as a real or perceived temporary challenge to the organism ability to maintain homeostasis and can be either physiological or psychological in nature. The organism responds to acute stress by rapidly mobilizing the autonomic and neuroendocrine systems, producing physiological changes that facilitate the response to the threat and the return to homeostasis. Autonomic system activation releases epinephrine (E, secreted by the adrenal medulla) and norepinephrine (NE, from the adrenal medulla and sympathetic nerves) that act on peripheral adrenergic receptors. Additionally, catecholamines are released within the brain where they activate central nervous system receptors. Acute catecholamine effects are short-lived, disappearing within 1 h (Tank and Lee Wong 2015), and include cardiovascular actions, metabolic resource allocation and sustained alertness.
The neuroendocrine response is under the control of the hypothalamic-pituitary-adrenal axis (HPA axis). Within this system, glucocorticoids (cortisol in humans and corticosterone in rodents, CORT) are released by the adrenal cortex in response to circulating adrenocorticotropic hormone (ACTH), released by the anterior pituitary. Release of ACTH is in turn elicited by corticotropin-releasing hormone (CRH or CRF) produced by parvocellular neurons of the paraventricular nucleus of the hypothalamus (PVN). In contrast to catecholamine-driven responses, glucocorticoid effects can be both rapid (within minutes after the stimulus) and long-lasting. The long-term effects develop over several hours and include transcriptional effects of activated glucocorticoid receptors (GRs) and epigenetic effects, such as methylation changes in target genes (Gray et al., 2017; Thomassin et al., 2001). Glucocorticoids can also bind to and exert activities through the mineralocorticoid receptors (MRs) (de Kloet 2013). Glucocorticoids participate in the termination of the stress response through a negative feedback process mediated by GRs that reduces the secretion of CRH in the PVN and ACTH in the pituitary to limit the effects of these hormones (Keller-Wood and Dallman 1984). Negative feedback mechanisms include transcriptional effects that are both ligand-dependent (Lachize et al., 2009; McKay and Cidlowski 1998) and ligand-independent (Rainville et al., 2019), as well as signaling effects via membrane-bound GRs (Di et al., 2003). Finally, the HPA axis has a prominent circadian and ultradian rhythms that play an important role in the organism's health (Lightman et al., 2020; Yao and Silver 2022).
With prolonged and/or intense stress exposure (chronic stress), the physiological burden to reinstate allostasis may produce detrimental consequences for the organism. Chronic secretion of glucocorticoids decreases GR expression in the brain which results in reduced negative feedback and dysregulation of the HPA axis (for review see (Herman et al., 2016; Tsigos et al., 2000)).
Because of lowered GR levels, CRH levels increase and the balance between MR and GR expression is changed; these alterations affect the function of other brain areas, notably the prefrontal cortex and hippocampus, and may underlie the emotional and cognitive impairments produced by chronic stress.
2.2. Impact of stress hormones on cognitive function
Indeed, persistent elevation of CORT or CRF has been associated with cognitive impairment especially in hippocampal dependent memory tasks (De Alcubierre et al., 2023; Henckens et al., 2016; Spannenburg and Reed 2023). For example, exposure to elevated glucocorticoids, in healthy humans produces reversible impairments in verbal declarative memory (Newcomer et al., 1999). In patients with Cushing Syndrome, a disorder characterized by hypercortisolemia, the most frequent cognitive symptoms reported are impaired memory (83%) and shortened attention span (66%) (Starkman 2013). Possible mechanisms leading to these impairments were suggested by early animal studies showing that chronic stress or chronic glucocorticoid exposure produced hippocampal and cortical atrophy, shorter dendritic branches and alterations in neurotransmitter levels (Cerqueira et al. 2005a, 2005b; Liston and Gan 2011; Magarinos et al., 1996; Radley et al., 2006; Wellman 2001). Measures of hippocampal volume in humans receiving high doses of glucocorticoids have yielded mixed results with some studies showing decreases in volume (Bermond et al., 2005; Brown et al., 2004; Wilner et al., 2002) and others finding no change (Coluccia et al., 2008; Hajek et al., 2006). In Cushing's patients smaller hippocampal and cortical volumes were found to correlate with longer disease duration (Bauduin et al., 2020; Bourdeau et al., 2002; Starkman et al., 1992). Low glucocorticoids levels also produce impaired cognition, especially in visual memory and attention, as it has been shown in individuals with adrenal insufficiency and with Addison's disease (Klement et al., 2009; Tiemensma et al., 2016). In rodents, adrenalectomy casues dendritic retraction in the mPFC (Cerqueira et al., 2007b). Thus, glucocorticoids effects on brain function follow an inverted-U relationship (Joels 2006), where both low and high levels of glucocorticoids produce deficits, brought about by imbalanced signaling through MR and GR. Indeed, several pharmacological studies with agonists and antagonists of GR and MR have revealed the importance of each receptor in cognitive function (Otte et al., 2007; Piber et al., 2016; Vogel et al., 2016; Wingenfeld and Otte 2019; Yalin et al., 2021; Young et al., 2004). For a recent review see (de Kloet and Joels 2023).
Elevated CRH following chronic stress also has negative effects on cognition that are mainly mediated by CRFR1 receptors (Henckens et al., 2016; Maras and Baram 2012; Uribe-Marino et al., 2016). Similar to glucocorticoids, elevated CRH produces cortical and hippocampal plasticity changes, spine actin depolarization, selective loss of spines, and detrimental effects on learning and memory (Chen et al., 2013; Wang et al., 2011).
2.3. Rodent models of chronic stress
Various rodent models of chronic stress have been developed and shown to produce behavioral and neuronal changes recapitulating elements of core symptoms of mood and anxiety disorders, or other stress-dependent psychophysiological disorders (for comprehensive reviews, see (Buynitsky and Mostofsky 2009; Qiao et al., 2016). As prenatal and early life stress are addressed elsewhere in this issue, here we will focus on adult stress.
Adult life stress is a well-recognized risk factor for psychopathology, and several rodent models have been developed to study this. Here, we will focus on 3 types of adult chronic stress: repeated restraint, chronic intermittent cold stress and chronic unpredictable stress.
Repeated restraint stress has been used as a model of chronic stress since the early 1980s, initially to examine homotypic stress habituation of the adrenergic system, HPA axis, and c-Fos activation in the brain, phenomena that arise within 4–6 days of exposure (Cole et al., 2000; Girotti et al., 2006; Stone and Platt 1982; Watanabe et al., 1994). Studies investigating the duration of stress protocols as a variable revealed that distinct brain regions exhibit differential responses to varying lengths of stress exposure. Daily sessions of restraint stress as short as one week (Brown et al., 2005; Garrett and Wellman 2009) caused dendritic remodeling in the mPFC, whereas a longer duration of the same stress procedure was necessary to observe morphological changes in the hippocampus. For example, Qiao et al., (2014) demonstrated a reduction in apical spine density in hippocampal CA1 and CA3 only when employing daily restraint stress for at least 3 weeks. Similarly, Watanabe and collegues (1992) reported apical dendritic retraction in CA3 after 3 weeks, the only time point they examined. McLaughlin and collegues (2007) found that 6 h of restraint for 21 days caused dendritic retraction in CA3, whereas 2 h of restraint stress for 21 days, 2 h for 10 days, and 6 h for 10 days did not. In conclusion, morphology in the PFC appears to be more sensitive to stress than morphology in the hippocampus.
Additionally, repeated restraint has been shown to decrease sucrose preference (Mao et al., 2022), and increase aggression, allodynia, and inflammatory pain (Bardin et al., 2009; Wood et al., 2003).
Chronic intermittent cold stress (CIC stress) is a homotypic chronic metabolic stressor that reproduces behavioral and functional changes paralleling those found in psychiatric disorders. For example, CIC stress has been shown to sensitize the HPA axis response (Ma and Morilak 2005), sensitize noradrenergic reactivity (Pardon et al., 2003), alter prefrontal and hypothalamic neuronal activity (Acosta et al., 1993; Correll et al., 2005; Moore et al., 2001), change spine and dendritic morphology in the orbitofrontal cortex (Adler et al., 2020), sensitize neuroimmune activity in the prefrontal cortex and hypothalamus (Girotti et al., 2011) and increase microglia-induced neuroinflammation in the hippocampus (Lang et al., 2020).
Another widely used chronic stress paradigm is chronic unpredictable stress. First developed by Katz and colleagues (Katz et al., 1981) as a 3 week variable stress paradigm that included severe stressors such as footshock, cold swimming, and immobilization, this paradigm was subsequently modified by Willner and coworkers (Willner et al., 1987) who substituted the severe stressors with milder ones, to better reproduce the human experience of daily, light but persistent stress exposure. Other variants of the CUS/CMS paradigm have also been developed (Banasr et al., 2007; Li et al., 2011). CUS/CMS induces a spectrum of behavioral abnormalities in rodents that parallel depressive symptoms including changes in reward sensitivity, decreased motivation, reduced grooming, and sleep changes. For a comprehensive review, see (Willner 2005). Along with behavioral effects, chronic unpredictable stress also causes structural changes, such as dendritic morphology and spine changes (Dias-Ferreira et al., 2009; Li et al., 2011).
3. Effects of stress on executive function
3.1. Cognitive flexibility
Cognitive flexibility can be defined as the ability of an individual to adapt their behavior to a changing environment or the ability to suppress old information in order to learn new information. Indeed, a lack of cognitive flexibility often leads to perseverance of behaviors that are no longer beneficial, or the inability to learn a new set of “rules.” Cognitive flexibility is often dysfunctional in patients with stress-related disorders, including depression and PTSD, where biased attention to perceived threats, ruminative focus on negative outcomes and perseverative behaviors are often a main symptom of the disorder (Clancy et al., 2016; Disner et al., 2011). For example, reduced cognitive flexibility shortly after a stressful or traumatic event is predictive of PSTD symptom severity a full year later (Ben-Zion et al., 2018). Two examples of cognitive flexibility that have been studied in relation to stress will be discussed in this review: reversal learning and extradimensional set shifting.
3.1.1. Rodent models of cognitive flexibility
Several rodent models of cognitive flexibility exist, many of which were originally based on human tests, such as the Wisconsin Card Sorting Test (Merriam et al., 1999) or the Cambridge Neuropsychological Test Automated Battery (CANTAB). One example is the attentional set-shifting task (AST) developed by Birrel and Brown (Birrell and Brown 2000) as a rodent analogue of the ID/ED sub-test of the CANTAB cognitive test battery developed by Sahakian and Robbins for human and non-human primates (Robbins et al., 1998). Both reversal learning and set shifting are evaluated in this test. Briefly, in the AST, rats dig for a food reward in small pots defined by cues along two stimulus dimensions: the material with which they are filled (sawdust, beads, etc) and an odor with which they are scented. The rats must learn which of these dimensions is relevant for locating the reward, and then which cue within that dimension is associated with the reward. After mastering a given contingency, the rules are changed, and the rat must learn a new association. For instance, the odor clove signals reward at first and the odor nutmeg does not; after mastering this discrimination, the rule is switched, whereby nutmeg is now associated with the reward, and clove is not (a Reversal). Subjects are then required to learn this new rule until they meet a predefined criterion. During this phase, subjects will produce errors that can be broadly classified as “perseverative,” i.e., persistent responses according to the obsolete rule, or “regressive,” i.e., relapses back to the old contingency after a series of correct responses. After a series of such tests in which the same stimulus dimension is informative regardless of how the rules are changed, the animals form a cognitive set, or a strategy that guides them in adapting to subsequent changes. Then, in the extra-dimensional (ED) cognitive set-shift portion of the test, odor is no longer the relevant dimension, but becomes the distractor, and the digging medium becomes the cue that signals reward. Thus, the animal must abandon the strategy of using odor as a guide and pay attention to tactile information instead. Cognitive flexibility in both reversal and set-shifting is measured by how readily the animal is able to suppress the old information and learn the new rule. More recently, operant-based systems to measure reversal learning and set shifiting have been developend for rats and mice, using operant boxes with retractable levers (Brady and Floresco 2015; Floresco et al., 2008) or touchscreen pads (Turner et al., 2017).
3.1.2. Brain areas and circuits in reversal learning
Human imaging studies show increased activity in medial prefrontal areas (Boehme et al., 2017; Cools et al., 2002), orbitofrontal areas (Ghahremani et al., 2010; Remijnse et al., 2006), parietal cortex, insula and cingulate cortex during reversal tasks (Yaple and Yu 2019). The orbitofrontal cortex is especially important for reversal learning, as evidenced in human studies (Hampshire et al., 2012), and in a large body of animal lesion studies, including monkeys (Dias et al., 1996), mice (Bissonette et al., 2008) and rats (McAlonan and Brown 2003). Other brain regions important for reversal learning are the striatum (Cools et al., 2002; Rogers et al., 2000b), amygdala (Izquierdo et al., 2013; Schoenbaum et al., 2003), and the hippocampus (Mala et al., 2015). Lesions in the ventral mid-thalamus (rhomboid and reuniens nuclei) and mediodorsal thalamus have also been shown to impair reversal learning (Chudasama et al., 2001; Linley et al., 2016). Recently, efforts have been directed at defining specific connectivity between the areas that functionally mediate reversal. Studies in humans and rodents show that major projection pathways involved in reversal include corticostriatal, thalamocortical, and cortico-amygdalar pathways. Thus, in human MRI studies, probabilistic reversal learning was associated with functional connectivity between the lateral orbitofrontal cortex and ventral striatum (Morris et al., 2016b). In rats, optogenetically inducing long-term potentiation (opto-LTP) in a MDT to orbitofrontal projection produces deficits in reversal learning (Adler et al., 2020). In an elegant study using selective viral ablation, Groman and colleagues showed that OFC-amygdala-striatal pathways mediate different aspects of value-reward encoding during a probabilistic reversal test. Rats with ablation of an OFC to nucleus accumbens projection showed deficits in reversal due to the inability to use negative outcomes to guide subsequent behaviors. By contrast, the ablation of an amygdala projection to OFC created deficits by not allowing rats to learn from positive outcomes. Interestingly, ablation of the reciprocal projection (OFC to amygdala) enhanced reversal (Groman et al., 2019). These nuanced outcomes may explain the inconsistencies in early reports of the effects of amygdala lesions on reversal learning (Izquierdo et al., 2013; Schoenbaum et al., 2003; Stalnaker et al., 2007), and indicate the necessity of circuit-level analyses to precisely dissect the anatomical correlates of reversal.
3.1.3. Neurochemical modulation of reversal learning
Glucocorticoids and CRH. Few studies have addressed the effects of elevated stress hormones on reversal learning; however, available evidence suggests that altered stress hormone signaling can negatively impact reversal learning. Thus, mice in which the MR/GR balance is altered by overexpression of MR and underexpression of GR show deficits in reversal tasks and increased perseveration (Harris et al., 2013). In rats, blockade of CRFR1 in the prefrontal cortex prevents stress-induced deficits in temporal order memory and reversal learning (Uribe-Marino et al., 2016).
Serotonin. Ample evidence points to the involvement of serotonergic signaling in the OFC in reversal. Depletion of serotonin either by removal of dietary tryptophan, 5-7-DHT lesions or use of the tryptophan hydroxylase depleting agent PCPA impairs reversal in humans, non-human primates, and rats (Clarke et al., 2007; Lapiz-Bluhm et al., 2009; Rogers et al., 1999). Receptor-specific manipulations with serotonin receptor antagonists or serotonin reuptake inhibitors suggest a complex picture of regulation. Antagonism of 5HT2A receptors impairs performance (Furr et al., 2012; Hervig et al., 2020), whereas antagonism of 5HT2C receptor facilitates reversal (Boulougouris et al., 2008; Nilsson et al., 2012), indicating a specificity in serotonergic modulation of reversal. In general, however, reduced serotonergic signaling in the OFC seems to be associated with increased perseveration (Clarke et al., 2004).
Dopamine. The importance of dopaminergic transmission in reversal behavior has been well documented, especially after extensive training on reversal learning, when the likelihood of reversal is “expected” (Costa et al., 2015; Klanker et al., 2015). There seems to be regional specificity to the effects of dopamine on reversal learning. Unlike serotonin, depleting dopamine in the OFC does not affect reversal learning (Clarke et al., 2007), whereas depletion in the striatum impairs reversal learning (Clarke et al., 2011). In human studies, increased dopaminergic activity in the striatum correlated with optimal reversal learning (Clatworthy et al., 2009). Both D1 and D2 receptor subtypes are involved in modulating reversal learning in humans, non-human primates, and rodents (Boulougouris et al., 2009; Izquierdo et al., 2006; Lee et al., 2007; Marino et al., 2022; Mehta et al., 2001; Smith et al., 1999; van der Schaaf et al., 2014). Recent evidence supports the notion that the differential role of D1 or D2 receptors in reversal may be region-specific (Sala-Bayo et al., 2020; Verharen et al., 2019).
Glutamate. Glutamate transmission in specific areas of the OFC is also implicated in visual reversal learning (Hervig et al., 2020), but little is known regarding the specific receptors involved. Subchronic administration of the NMDAR antagonist phencyclidine (PCP) produces impairments in reversal learning (Abdul-Monim et al., 2007), but subsequent analyses reported no stable effect of PCP on reversal (but reproducible negative effects on set-shifting) (Janhunen et al., 2015). Given the diversity of glutamate receptors and signaling modes, it is likely that more selective targeting of receptors or receptor subunits is required to clarify the involvement of glutamatergic transmission in reversal. In this vein, a role for the metabotropic receptor mGluR5 seems indicated by the fact that mGluR5 knockout mice display impaired visual reversal (Lim et al., 2019). Additionally, multiple studies have reported that antagonism or gene deletion of NMDA receptor subunit 2B (GluN2B) in OFC and dorsal striatum impairs reversal learning (Brigman et al., 2013; Dalton et al., 2011; Thompson et al., 2015a). Further investigation is necessary to better understand the role of glutamate signaling in reversal.
3.1.4. Stress effects on reversal learning
Human studies. Acute stress has been associated with both better performance in reversal learning (Wieland et al., 2023), as well as impairments (Raio et al., 2017) Conversely, early life stress and chronic adult stress impair reversal learning. For example, in healthy young adults, early life stress correlates with increased perseveration on a reversal task and increased propensity for increased alcohol use (Franco and Knowlton 2023). In an adolescent cohort with a history of early life stress, individuals with higher life stress scores had more difficulty in updating contingencies during reversal learning tasks (Harms et al., 2018). In a study that used a modified reversal learing task to assess sensitivity to punishement and reward, adult females with high self-reported chronic stress showed increased perseverative errors in the punishment condition and reduced punishment sensitivity in the reward condition (Monni et al., 2023). Moreover, reversal learning deficits have been documented in several stress-related psychiatric disorders such as major depressive disorder (where it was accompanied by abnormalities in the OFC) obsessive compulsive disorder, and generalized anxiety disorder (Drevets 2007; Szabo et al., 2013), and in individuals repeatedly exposed to traumatic events (Levy-Gigi and Richter-Levin 2014).
Rodent studies. In rats, acute stress is generally associated with better performance in reversal learning (Bryce and Howland 2015; Graybeal et al., 2011; Thai et al., 2013). In contrast, early life stress impaired reversal learning in female mice, and this was associated with increased expression and density of parvalbumin expressing neurons in OFC (Goodwill et al., 2018). Chronic intermittent cold stress in adult rats results in robust deficits in olfactory reversal learning task (Danet et al., 2010; Donegan et al., 2014; Furr et al., 2012; Lapiz-Bluhm et al., 2009; Patton et al., 2017; Wallace et al., 2014). Reversal impairments are also observed after two weeks of chronic unpredictable stress in both olfactory and spatial reversal tasks (Bondi et al. 2007, 2010; Hill et al., 2005; Jett et al., 2015; Jett and Morilak 2013; Quan et al., 2011; Yu et al., 2016). Mechanistically, chronic stress has been shown to decrease the levels of ciliary neurotrophic factor (CNTF) and reduce activation of its downstream effector JAK2 in the OFC. Restoration of CNTF levels within this brain region was sufficient to correct the effects of stress on reversal learning (Girotti et al., 2019). Functionally, chronic stress effects on reversal learning can be recapitulated by optogenetically inducing long term-potentiation (opto-LTP) in the OFC; conversely, chronic stress deficits are ameliorated by optogenetic long term depression (opto-LTD) in the OFC (Adler et al., 2020). Interestingly, JAK signaling is involved in hippocampal LTD (McGregor et al., 2017; Nicolas et al., 2012), and it is possible that this signaling pathway is also implicated in establishing LTD in the OFC. Taken together, these data support the notion that optimal reversal learning is associated with long-term depression in the OFC, in part mediated by CNTF/JAK signaling, that enables flexible behavior. In support of this idea, it is worth noting that depotentiation is associated with other forms of reversal; for example, temporary depotentiation in the hippocampus accompanies spatial reversal learning (Dong et al., 2013; Duffy et al., 2008; Mills et al., 2014; Morice et al., 2007). Conversely, stress may produce reversal deficits by inducing a state of increased excitability and hyperactivity in the OFC that precludes the depotentiation necessary for optimal reversal. Indeed, this possibility is supported by morphological evidence that chronic stress associated with behavioral inflexibility produces hyper-elaboration and increased dendritic length and spine numbers in the OFC (Adler et al., 2020; Liston et al., 2006).
3.1.5. Brain areas and circuits in set-shifting
Human and animal studies have long suggested that the thalamo-fronto-striatal circuit is vital in set-shifting behavior. Prefrontal activity is associated with set-shifting performance in human subjects (Manes et al., 2002; Miller and Cohen 2001; Owen et al., 1991; Rogers et al., 2000a; Stuss et al., 2000). Animal lesion studies indicate that mPFC damage results in deficits specifically in set-shifting while reversal learning remains unaffected (Birrell and Brown 2000; Bissonette et al., 2008). Lesions of mediodorsal thalamus (Ouhaz et al., 2022) or ventral hippocampus (Mala et al., 2015; Placek et al., 2013) also produce deficits in set-shifting. Optogenetically inducing long-term depression specifically within MDT terminals in the mPFC impairs set-shifting (Bulin et al., 2020).
Recent chemogenic manipulation using designer receptors exclusively activated by designer drugs (DREADDs) implicated the LC in modulating the mPFC during set-shifting. Elevated activity in LC neurons projecting specifically to mPFC was improved set-shifting and reduced regressive errors (Cope et al., 2019). Further studies suggest that the LC may be important in “rule switching”, allowing the animals to acquire the new rule easier during set shifting (McBurney-Lin et al., 2022).
3.1.6. Neurochemical modulation of set-shifting
Glucocorticoids and CRH. Few studies have addressed the role of stress hormones in set-shifting. In humans, pharmacological stimulation of GR or MR does not have any effect on flexibility tasks (Deuter et al., 2019; Wingenfeld et al., 2011). However, antagonizing MR with spironolactone diminished set shifting performance in humans, suggesting a potential role for this receptor in cognitive flexibility (Otte et al., 2007). CRH effects on set-shifting may be region-specific and dose-dependent. In one study in rodents, intracerebral administration of CRH impaired set-shifting with an inverted-U dose relationship. Conversely, microinjection into the locus coeruleus improved performance, (Snyder et al., 2012), consistent with evidence that CRH stimulates NE release from the LC (Curtis et al., 1997) and that NE in the mPFC is required for optimal set-shifting (Lapiz and Morilak 2006).
Serotonin. Evidence suggests that, besides reversal, serotonin is also involved in set-shifting. Serotonin transporter (SERT) knockout rats, which have increased extracellular serotonin, perform better than intact controls on set-shifting (Nonkes et al., 2012). However, in the opposite direction, serotonin depletion often has no effect on set-shifting (Clarke et al., 2005; Gallagher et al., 2003; Hughes et al., 2003). Serotonin receptor antagonists often ameliorate stress effects. For example, in rats, the negative effects of chronic restraint stress on set-shifting were ameliorated by the 5-HT7 receptor antagonist amisulpride (Hedlund 2009; Hedlund et al., 2005; Leopoldo et al., 2011; Nikiforuk and Popik 2013). The 5-HT6 receptor antagonists SB 271046 and sertindole were also shown to reduce deficits in set-shifting produced by PCP (Idris et al., 2010; Rodefer et al., 2008).
Dopamine and Norepinephrine. Dopamine activity in the mPFC is required for optimal performance in set-shifting tasks. Thus, in monkeys, depletion of dopamine in the mPFC using 6-OHDA produced deficits in set-shifting (Crofts et al., 2001; Robbins and Roberts 2007) and in humans increasing dopamine activity in the mPFC improved set-shifting (Apud et al., 2007). The role played by different dopamine receptors in this behavior is complex (Floresco 2013). For example, the D1 receptor antagonist SCH23390 infused within the mPFC impaired set-shifting in rats (Haluk and Floresco 2009; Ragozzino 2002), while mPFC infusions of the D4 antagonist L-745,870 improved set-shifting (McQuail et al., 2021). Norepinephrine also contributes to attentional set-shifting. Deafferentiation of noradrenergic input into the mPFC results in set-shifting deficits (McGaughy et al., 2008; Tait et al., 2007). In humans, tonic activity of noradrenergic neurons in the LC (assessed by pupillary dilation) facilitated set-shifting (Pajkossy et al., 2017). In animals, acute activation of noradrenergic signaling in the mPFC by administration of atomoxetine, a serotonin and norepinephrine uptake inhibitor (Newman et al., 2008), or atipamezole, an α2-adrenergic autoreceptor antagonist (Bondi et al., 2010; Lapiz and Morilak 2006), both improved set-shifting.
Glutamate. Glutamate transmission is also implicated in set-shifting. Direct antagonism of AMPA or NMDA receptors, but not mGluR5, in the mPFC produced set-shifting deficits (Jett et al., 2017). By contrast, systemic administration of the partial NMDA receptor agonist D-cycloserine rescued set-shifting impairments induced by scopolamine (Siddik and Fendt 2022). Expression of the GluA1 AMPA receptor subunit decreased within the mPFC of stressed animals that exhibited set-shifting deficits (Sun et al., 2022). Cortical expression of the NMDAR NR1 subunit is decreased in aged rats with set-shifting deficits (McQuail et al., 2021). Electrophysiological experiments have also revealed increased AMPA/NMDA ratios on fast-spiking interneurons in the mPFC of aged rats with set-shifting deficits (McQuail et al., 2021).
3.1.7. Stress effects on set-shifting
Human studies. In humans, not many studies have addressed the effect of stress on set-shifting using the Wisconsin Card test (WCST) or CANTAB tests and the results seem variable. For example, in one study using the WCST, acute stress did not impact cognitive flexibility (Hendrawan et al., 2012), whereas in another study using a virtual TSST and a variant of the WCST, investigators reported improved cognitive flexibility (Delahaye et al., 2015). In work comparing sexes, it was found that acute stress reduced cognitive flexibility measured with the WCST in males but not in females (Kalia et al., 2018; Shields et al., 2016). In other studies using tests of cognitive flexibility other than the WCST, some investigators have reported a negative impact of acute stress on this cognitive domain and no difference between males and females (Alexander et al., 2007; Hillier et al., 2006). In the few studies evaluating chronic stress, the data collected with tests other than the WCST suggest that high perceived stress correlates with set-shifting deficits (Orem et al., 2008). Early life stress stress also correlated with decreased cognitive flexibility in adulthood, especially in individuals with a high degree of self-reported ongoing stress (Kalia and Knauft 2020; Kalia et al., 2021).
Rodent studies. In rodent studies, the effects of acute stress on set-shifting are dependent on the type of stress the subject is exposed to prior to the set-shifting test. For example, a 15-min tail pinch is sufficient to produce deficits in set-shifting, but not reversal learning (Butts et al., 2013), while a 30-min acute restraint stress does not alter set-shifting (Thai et al., 2013). Additionally, a single prolonged stress paradigm (SPS) in which rats are acutely exposed to multiple stressors within a short time (a common model for PTSD), also resulted in set-shifting deficits (George et al., 2015). This suggests there may be a “threshold” above which acute stress produces set-shifting deficits. Subjects exposed to chronic restraint stress or CUS consistently exhibit deficits in set-shifting (Bondi et al., 2008; Fucich et al., 2016; Jett et al., 2013; Liston et al., 2006; Morilak et al., 2005). Interestingly, CIC stress that impaired reversal learning had no effect on set-shifting (Lapiz-Bluhm et al., 2009). Chronic stress can lead to decreased activity in the mPFC in both humans and animal models (Liston et al., 2009), and reduces responsivity of the mPFC to afferent stimulation as measured by local field potentials (Jett et al., 2017). Reduced response to afferent input was unique to the MDT pathway to mPFC, as the response to stimulation from the ventral hippocampus remained intact after CUS (Jett et al., 2017). Using optogenetics to induce long-term depression specifically within the MDT-mPFC pathway mimicked the effects of chronic stress on set-shifting. By contrast, long-term potentiation induced optogenetically within the MDT-mPFC pathway of stressed rats ameliorated the stress-induced deficits in set-shifting (Bulin et al., 2020). Decreased mPFC activity and responsiveness also correlate with stress-induced morphological changes, as subjects exposed to chronic stress exhibit reduced length of the apical dendrite and reduced dendritic branching in glutamatergic pyramidal neurons, and an overall loss of dendritic spines (Anderson et al., 2020; Cerqueira et al. 2005a, 2005b, 2007a, 2007b; Cook and Wellman 2004; Dias-Ferreira et al., 2009; Liston et al., 2006; Radley et al. 2004, 2006; Silva-Gomez et al., 2003).
3.2. Attention and behavioral inhibition
Broadly speaking, attention is the ability to actively process specific information in the environment while ignoring irrelevant information, whereas behavioral inhibition is characterized by the ability to restrain a response. Even within these general definitions, it is apparent that the two constructs are inter-related, in that a degree of inhibitory control is exerted during tasks of attention (when responses to extraneous stimuli need to be restrained) and vice versa, attention to environmental change is a prerequisite for inhibition of a response that has become ineffective (Bari and Robbins 2013). Indeed, as we will discuss later, behavioral tasks measuring deficits in attention also report on behavioral disinhibition.
Impulsivity is a component of normal behavior that manifests as loss of behavioral inhibition, including intolerance for delayed rewards, and actions performed hastily and without due consideration of the consequences. High levels of impulsivity are associated with psychiatric disorders such as attention-deficit disorder, bipolar disorder, substance use disorder, and other addictive disorders (Winstanley et al., 2006). Impulsivity manifests in both the innate tendency of the subject to express behavioral disinhibition (trait impulsivity) and in the response to environmental pressures that may produce disinhibited behavior (state impulsivity). Impulsivity can be conceptualized behaviorally and neurobiologically under two broad categories: motor impulsivity, or the inability to inhibit a prepotent motor response, and choice impulsivity, or the selection of a small, immediate reward in favor of a larger, delayed reward (Bari and Robbins 2013; Dalley and Robbins 2017; Evenden 1999b; Winstanley et al., 2006).
3.2.1. Rodent models for attention and behavioral inhibition
3.2.1.1. Motor impulsivity
The 5-choice serial reaction time test (5-CSRTT) is used to measure both attention and response inhibition in rodents (Bari et al., 2008; Carli et al., 1983). Modified versions of this test, e.g., the 4-choice serial reaction test and the Sussex5CSRTT, are used in humans (Sanchez-Roige et al., 2016; Voon et al., 2014). In the rodent task, subjects are trained to detect brief flashes of light presented randomly in 1 of 5 holes and make a nose-poke response in the correct location to receive a food reward. Responses made before presentation of the stimulus (premature responses) are considered an index of motor impulsivity, whereas responses in one of the unlit holes (incorrect responses) indicate deficits in attention (Eagle and Baunez 2010; Sosa et al., 2021). The Go No-Go test is another motor inhibition test in which separate signals (different tones) are associated with “go” and “no-go” trials; the subject must respond in go trials but inhibit responding in no-go trials. Motor impulsivity can also be evaluated by considering “speed” of inhibitory control, measured with the Stop-Signal-Reaction-Time Test (SSRTT). In this task, rats are presented with two levers. The animal is required to press in quick succession first the left then the right lever to receive a food pellet. On 20% of trials a stop signal (a tone, for example) is played after the left lever press signaling that the subject must withhold responding on the right lever to receive the reward (Eagle and Baunez 2010; Eagle et al., 2008). The critical measure in SSRTT is the time taken to stop the response. Thus, SSRTT specifically measures inhibition of actions that have been started and engage “stopping” processes, whereas Go No-Go and 5-CSRT tests measure the ability to inhibit the initiation of the response and engage “wait” processes during inhibitory responses. Interestingly, several studies have highlighted neuroanatomical and pharmacological differences between “waiting” and “stopping” impulsivity that provide useful frameworks for further mechanistic analysis in rodent models of impulsivity (Dalley et al., 2011; Dalley and Robbins 2017).
3.2.1.2. Choice impulsivity
Impulsive choice is exhibited by subjects who choose an immediately available small reward in preference to a larger but delayed reward. This type of decisional impulsivity is heavily shaped by reward valuation and the loss in perceived reward value when its delivery is delayed. In rodents, impulsive choice is often measured with delay-discounting tests (Evenden 1999a). Briefly, animals choose between two levers, one of which provides a reward of one pellet, the other a reward of 4 pellets. Over the period of the test the delay to the large reward increases from 0 s to 10, 20, 40, and finally 60 s and a delay-discounting curve can be obtained. Animals typically show a strong preference for the larger reward early in the session when the delay is short or absent but shift their preference to the smaller reward as delay to the larger reward increases. Individuals who generate steeper discounting curves, such that each unit of time-delay has a greater negative effect on the valuation of the reward, are described as exhibiting more decisional impulsivity.
3.2.2. Brain regions and circuits involved in attention and behavioral inhibition
3.2.2.1. Motor impulsivity
Many of the neuroanatomical substrates of behavioral inhibition have been identified from rodent brain lesion studies (for review see (Eagle and Baunez 2010)). To summarize, lesions in the OFC, dorsal medial striatum and subthalamic nucleus (STN) affect stopping processes measured on SSRT as well as premature responding on the 5-CSRTT. Additionally, lesions in the infralimbic cortex (Chudasama et al., 2003b), insular cortex (Dambacher et al., 2015), ventral hippocampus (Abela et al., 2013), and cingulate cortex (Muir et al., 1996) all increase premature responding in the 5-CSRTT. The role of the nucleus accumbens may be more nuanced with differential effects mediated by the shell and the core (Besson et al., 2010; Sesia et al., 2008). Recently, using a cued sensory association task, the secondary motor cortex was implicated in coding and controlling premature motor output (Guzulaitis et al., 2022). In humans, motor impulsivity is also associated with corticostriatal and corticosubthalamic connectivity (Aron et al., 2003; Hannah and Aron 2021). Specifically, the STN is well recognized as an inhibitory modulator of cortico-striatal motor outputs (Morris et al., 2016a). Recent evidence from human studies of Parkinson disease (Mosley et al., 2020) confirms previous work that connectivity of premotor areas to the STN signal motor stopping (Aron et al., 2016) but also show a role of STN in facilitating disinhibitory control, possibly via connectivity of STN with prefrontal and orbitofrontal cortices (Dagher 2020; Mosley et al., 2020). In rodents, several lines of evidence, including lesion studies (Baunez and Robbins 1997, Phillips and Brown 2000) and disconnect studies (Chudasama et al., 2003a) have supported the notion of the STN as an “action stop” center. More recently, mapping of the underlying circuitry has confirmed the role of secondary motor cortex to STN connections (Adam et al., 2022) and mPFC to STN connections (Li et al., 2020) in motor inhibition.
3.2.2.2. Choice impulsivity
Lesion studies in rodents and functional imaging studies in humans implicate the nucleus accumbens (NAC) core, basolateral amygdala (BLA), hippocampus, insula, lateral prefrontal cortex (PFC), posterior cingulate cortex, parietal cortex, and medial and lateral orbitofrontal OFC in delay discounting impulsivity (Cardinal et al., 2001; Kable and Glimcher 2007; Tanaka et al., 2004; Winstanley et al., 2004b). With respect to corticosubthalamic circuitry, lesions of the STN actually reduce impulsive choice (Winstanley et al., 2005a). However, there is also evidence for a role of STN to mediate some aspects of cognitive or choice impulsivity, both in rodents and humans, with a putative regulation by associative and limbic cortices (mPFC and OFC) (Heston et al., 2020; Mosley et al., 2020). Indeed, a recent report showed that activity levels in the pre-supplementary motor area were shown to predict impulsive choice in a gambling test in humans (Lohse et al., 2023).
3.2.3. Neurochemical modulation of attention and behavioral inhibition
3.2.3.1. Motor impulsivity
Glucocorticoids and CRH. Studies that have investigated the role of stress hormones in impulsive responding did not report significant effects (Kentrop et al., 2016). Instead, stress hormones seem to affect attention. In particular, MR blockade impairs selective attention in healthy humans (Cornelisse et al., 2011a; Otte et al., 2007) and polymorphisms within the MR gene are associated with increased hyperactivity and decreased attention in ADHD cohorts (Kortmann et al., 2013).
Serotonin. Serotonin neurotransmission plays an important role in both reward evaluation and inhibitory responses (Desrochers et al., 2022). Several lines of evidence, not all in concordance, have shown the importance of serotonin in motor inhibition. Thus, in some studies, increased tonic extracellular serotonin levels in mPFC correlated with impulsive action in the 5-CSRTT (Dalley et al., 2002; Puumala and Sirvio 1998). However, there is also evidence that depleting serotonin in the frontal cortex increases motor impulsivity in rodents (Harrison et al., 1997; Winstanley et al., 2004a) and humans (Worbe et al., 2014) and that increasing serotonin in the cortex improves wait impulsivity. (Fonseca et al., 2015; Miyazaki et al. 2014, 2020). Thus, to fully characterize serotonergic involvement in impulsivity a more nuanced approach that considers receptor-, region- or circuit-specific effects may be necessary. With respect to receptor subtypes, 5-HT2A and 5-HT2C receptors seem to have opposite effects on motor impulsivity; 5-HT2C receptor antagonism (in prefrontal cortex and accumbens) is associated with greater premature responding, and activation of these receptors lowers premature responding in 5-CSRTT. Conversely, 5-HT2A receptor antagonism is associated with decreased premature responding and activation is associated with increased premature responding in 5-CSRTT (Fletcher et al., 2007; Koskinen et al., 2000; Robinson et al., 2008a; Silveira et al., 2020; Winstanley et al., 2004c).
Dopamine and norepinephrine. The dopaminergic and noradrenergic systems are also implicated in the modulation of impulsive responding in the 5-CSRTT, perhaps through cross-signaling. In particular, D2 receptor availability has been associated with high trait impulsivity in humans (Volkow et al. 1993, 2001) and in rodents performing tests of motor impulsivity (Caprioli et al., 2013; Dalley et al., 2007; Pattij et al., 2007). Increasing norepinephrine (and dopamine) levels with NE reuptake inhibitors such as atomoxetine and desipramine decreases impulsive responding in the 5-CSRTT (Navarra et al., 2008; Paine et al., 2007; Paterson et al., 2011; Robinson et al., 2008b). Finally, interactions between the noradrenergic and dopaminergic systems were reported in a study where the beneficial effects of atomoxetine and duloxetine (a serotonin and norepinephrine reuptake inhibitor) on decreasing motor impulsivity were blocked by a D1-like receptor antagonist (Sasamori et al., 2019).
Glutamate and GABA. Other neurotransmitter systems that have a role in impulsive action include the ionotropic and metabotropic glutamate receptors (Higgins et al., 2016; Isherwood et al., 2017). In particular, metabotropic glutamate receptor 5 (mGluR5) is differentially involved in impulsive action and impulsive choice, as positive allosteric modulation of this receptor reduces impulsive action but does not affect impulsive choice (Isherwood et al., 2015). GABAergic transmission also plays a role in motor impulsivity. Thus, high trait impulsivity in rats is associated with reduced GABA levels in the ventral striatum and decreasing glutamate decarboxylase (GAD65/67) expression in the nucleus accumbens increased impulsivity in low impulsivity rats (Caprioli et al., 2014; Sawiak et al., 2016).
3.2.3.2. Choice impulsivity
Serotonin. Serotonin's role in behavioral inhibition is not restricted to motor impulsivity but extends to choice impulsivity, although in this instance again, pharmacological effects are complex and outcomes have varied across studies. Some studies indicated that depleting central serotonin in rodents does not produce effects on a delay discounting task (Winstanley et al., 2004a), in concordance with human studies. (Crean et al., 2002; Dougherty et al., 2015; Worbe et al., 2014). In other work, however, central 5-HT depletion did increase impulsive choice in rodents (Mobini et al., 2000) and serotonin efflux in the dorsal raphe nucleus is higher while waiting for delayed rewards (Miyazaki et al., 2011). Moreover, in rodents, selective serotonin reuptake inhibitors (SSRIs) increase selection of larger, delayed rewards (Bizot et al., 1999). Thus, serotonin effects appear to be complex and may depend on interplay with other neurotransmitter systems (Winstanley et al., 2005b).
Dopamine and norepinephrine. Depletion of dopamine in the striatum increases discounting rates in delay discounting tests (Tedford et al., 2015), and rats with higher discounting rates have a blunted cue dopamine response (Moschak and Carelli 2017). Accordingly, optogenetic stimulation that induces dopamine release within the nucleus accumbens produces a shift toward waiting for delayed larger rewards in rats (Saddoris et al., 2015). Expression of D2 receptors is important for choice impulsivity as knock-down of these receptors in the ventral tegmental area shifted preference for the smaller, immediate reward in rats (Bernosky-Smith et al., 2018). As with other neurotransmitters, however, the effects of dopamine on delay discounting seems to be region-dependent. Thus, discounting rates are increased by infusions of D2, but not D1, receptor antagonists in nucleus accumbens (Yates and Bardo 2017). Conversely, discounting rates are increased when D1, but not D2, antagonists are infused in the prefrontal cortex (Loos et al., 2010). These functional differences may be linked to the heterogeneity of dopamine receptor expression in cortical compared to subcortical regions.
A large volume of work with norepinephrine reuptake inhibitors suggests an important role for norepinephrine not only in impulsive action but also in impulsive choice. Pharmacological increases in extracellular norepinephrine levels by norepinephrine transporter (NET) blockers, such as atomoxetine, consistently increase preference for larger delayed rewards in delay discounting tasks (Robinson et al., 2008b; Sun et al., 2012). These effects appear to be mediated by the α2 adrenergic receptor, as administration of α2 agonists decrease impulsive choice (Nishitomi et al., 2018) and α2 antagonists increase impulsive choice (Schippers et al., 2016). Despite the clear role of norepinephrine in impulsivity it is important to note the possibility that some of the behavioral effects described above may be in part due to changes in dopaminergic tone, as atomoxetine also increases cortical dopamine levels (Bymaster et al., 2002) and α2 agonists have been found to decrease dopamine release (Ihalainen and Tanila 2002).
Glutamate and GABA. Like motor impulsivity, choice impulsivity is also regulated by excitatory and inhibitory neurotransmitters. Thus, levels of cortical glutamate-glutamine and GABA predict impulsive decision making and gambling severity in humans (Weidacker et al., 2020). In rodents, ionotropic glutamate receptors have been shown to be implicated in delay discounting (Yates and Bardo 2017; Yates et al., 2015).
3.2.4. Stress effects on behavioral inhibition
Human studies. In humans there is evidence that both acute and chronic stress affect behavioral inhibition. A meta-analysis of interactions of acute stress and risky behaviors revealed that overall, stress exposure led to more disadvantageous and risky decisions than nonstress conditions (Starcke and Brand 2016). Interestingly, stress does not uniformly increase impulsive responding, rather it interacts with trait impulsivity as a factor to reveal preexisting biases in goal-directed behavior. Thus, in healthy adults acute stress impaired decision making and speed of choice in both non-gambling and gambling settings, and these effects were more pronounced in individuals with higher impulsivity (Canale et al., 2017; Raio et al., 2020). However, in humans other complicating factors, such as Individual perception of stress, may also affect choice behavior. In one study, researchers found that in the presence of an acute stressor individuals who are more likely to perceive stressful situations as such show a preference for larger, delayed rewards, while those with low perceived stress prefer short, immediate rewards (Lempert et al., 2012). The authors suggest that individuals with high perceived stress may interpret the acute stress as more threatening and this reduces their reward response in the short term. Indeed, a correlation between acute stress, decreased reward sensitivity and increased choice for later reward have been documented in previous work (Bogdan and Pizzagalli 2006). More prolonged stress has been linked to increased impulsivity and increased risk of developing specific addictive behaviors. In one study, greater lifetime stress exposure was significantly correlated with a greater tendency to make impulsive decisions under negative emotionality and was predictive of developing food addiction but not alcohol addiction (McMullin et al., 2021). In another study, stress significantly correlated with higher impulsivity in methamphetamine users but not in cocaine users (Mahoney et al., 2015).
Rodent studies. Fewer studies have addressed the role of stress on impulsivity in rodents. Perinatal stress has been shown to increase impulsive action but reduce impulsive choice in adulthood (reviewed in (Sanchez and Bangasser 2022)). Moreover, sex difference may be at play, as the type of perinatal stress applied elicits different effects on impulsivity (especially impulsive choice) in males versus females (Paine et al., 2021; Sanchez and Bangasser 2022). Conversely, administration of corticosterone to adolescent rats decreased premature responding at longer intertrial intervals on the 5-CSRTT but increased delay discounting (Torregrossa et al., 2012). Thus, with the caveat that corticosterone administration does not fully recapitulate the effects of natural stress, it appears that stress experienced before adulthood may have differential effects on impulsive action and impulsive choice. Stress applied in adulthood is also capable of altering impulse control in rodents. Accordingly, the response of rats in a motor impulsivity test analogous to the 5-CSRTT varies in a manner that is dependent on the type of stress used: while an acute stressor did not change performance, CIC stress lowered premature responses while CUS increased premature responses at longer intertrial interval times (Girotti et al., 2022). CUS also increased impulsive choice in a genetic mouse model of schizophrenia (Buhusi et al., 2017). Taken together, a large body of evidence suggests that stress has a nuanced effect on impulsive responding that depends on individual baseline setpoints of goal-directed behavior.
3.3. Working memory
Working memory is defined as the temporary storage and manipulation of content used to perform cognitive tasks such as language comprehension, learning, and reasoning (Baddeley 1992). This also requires attentional processes, in particular focused or selective attention (Diamond 2013). In humans working memory can directly be assessed with simple ordering tasks (presenting subjects with random numbers and asking them to reorder them in ascending or descending order). The N-back task, in which the subject is presented with a series of stimuli and is asked to indicate when a current stimulus matches one from n steps earlier, is also a widely used task to assess working memory, although selective and sustained attention are also heavily employed in this task (Owen et al., 2005; Pelegrina et al., 2015).
3.3.1. Rodent models for working memory
In rodents, T-maze alternation tasks, the 8-arm radial maze, and object recognition tasks with variable lengths of delay are some of the most used tests of spatial and non-spatial working memory (Kinnavane et al., 2015; Lalonde 2002; Olton 1987; Warburton and Brown 2015). These tasks rely on the intrinsic propensity of the animal to explore novel locations or objects when given a choice between a previously experienced situation and a novel one. Deficits in working memory will diminish preference for the novel location/object, as the animal does not retain information about previously encountered situations.
3.3.2. Brains areas and circuits of working memory
Human and non-human primate studies have revealed a central role of the frontal cortex in the regulation of working memory. Early studies in monkeys identified neurons in the PFC that maintain robust activity during cue presentation and during the delay period while the subjects keep stimulus information in working memory. This sustained activity is correlated with the ability to remember the information and is susceptible to distractions (Fuster & Alexander 1971, 1973). Human neuroimaging studies also found sustained activation in the lateral PFC while the subjects kept visual items in working memory (Courtney et al., 1998; Sakai et al., 2002). Moreover, specific involvement of parietal, sensory and premotor cortices has been shown in both human and primate studies of working memory (Khan and Muly 2011; Klingberg et al., 1997).
Rodent lesion studies have shown the importance of perirhinal, entorhinal cortex, hippocampus, prefrontal cortex and insular cortex for recognition memory (Albasser et al., 2012; Barker and Warburton 2011; Bermudez-Rattoni 2014; Ennaceur and Aggleton 1997; Tuscher et al., 2018; Warburton and Brown 2015). Subcortical areas involved in recognition memory include thalamus, hypothalamus, striatum, septum, amygdala and cerebellum (for a comprehensive review see (Chao et al., 2022)). At the circuit level, electrophysiology studies implicated connectivity between PFC-thalamus (reuniens nucleus) and hippocampus as mediating behavior in a T-maze test (Hallock et al., 2016; Ito et al., 2015). Moreover, chemogenic inhibition showed that reciprocal PFC-mediodorsal thalamus (MDT) projections mediate different phases of working memory processing, and that MDT input is necessary in sustaining prefrontal activity during working memory maintenance (Bolkan et al., 2017).
3.3.3. Neurochemical modulation of working memory
Glucocorticoids and CRH. Several reports indicate that alteration of stress hormone signaling negatively impacts working memory. MR seems to be particularly important for working memory. For example, individuals with Addison's disease performed better in working memory tasks when both receptors were activated compared to when only GRs were activated (Tytherleigh et al., 2004). In another study, mifepristone, a GR antagonist that shifts the MR/GR balance towards MR, improved spatial working memory and recognition memory in individuals with bipolar disorder (Young et al., 2004). CRHR1 signaling is also implicated in working memory. In humans CRHR1 polymorphisms interact with early life stress to produce deficits in working memory (Fuge et al., 2014). Administration of a selective CRFR1 antagonist prevented cognitive impairments in recognition memory in stressed mice (Philbert et al., 2013).
Dopamine and Norepinephrine. The importance of dopamine in working memory was established from observations that monkeys with dopamine depletion in the PFC showed profound deficits in working memory (Brozoski et al., 1979). Early studies aimed at defining receptor subtypes unveiled a critical role for D1 receptors in the effect of dopamine on working memory. Interestingly, optimal outcomes follow an inverted-U-shaped dose-response curve where both insufficient and excessive D1 activation result in impairments (Williams and Castner 2006; Williams and Goldman-Rakic 1995). Since D1 receptors are more abundant than D2 receptors in the prefrontal cortex they have been the focus of much early research; however, more recent studies have also highlighted a potential role for D2 receptor signaling in working memory. For example, bromocriptine, a D2 receptor agonist, improved working memory in a variety of rodent maze tests (Onaolapo and Onaolapo 2013; Phelps et al., 2015; Tarantino et al., 2011), and treatment with a D2 receptor antagonist (haloperidol) produced deficits in spatial working memory and object recognition (Terry et al. 2007a, 2007b).
Noradrenergic receptor signaling is also important for working memory, in particular α2 adrenoreceptors. Prefrontal cortex infusion of yohimbine, a noradrenergic α2 receptor antagonist, impairs performance on working memory tasks (Li et al., 1999), whereas, administration of the α2-adrenoceptors agonist guanfacine improves working memory functions in humans (Jakala et al., 1999; Swartz et al., 2008), monkeys (Arnsten et al., 1988) and rats (Hains et al., 2015).
Other neurotransmitter systems. In addition to catecholamines, there is evidence for an involvement of cholinergic receptors, with differential roles of nicotinic and muscarinic receptors in different aspects of the working memory processing (Granon et al., 1995). The glutamate system is also important for working memory function. For example, in rodents, infusion of NMDA within the septum improves consolidation in a working memory task (Puma et al., 1998). Administration of NMDA receptor antagonists either systemically or locally within the dorsomedial PFC impairs spatial working memory in rats (Aura and Riekkinen 1999; Gutnikov and Rawlins 1996). Interestingly, the NMDA antagonist AP5 infused within the septum, showed dose-dependent effects, with improved performance in a recognition task at low doses (Puma and Bizot 1998) and impairment at high doses (Puma et al., 1998), underscoring the potential subtleties of pharmacological manipulation on this behavioral measure. Beside excitatory transmission, evidence shows that inhibitory transmission also modulates working memory. For example, levels of GABA in the prefrontal cortex change during working memory tasks in humans (Michels et al., 2012; Yoon et al., 2016) and blockade of GABAA receptors in the prefrontal cortex of monkeys and rats reduces working memory capacity (Auger and Floresco 2014; Rao et al., 2000).
Finally, the role of serotonin in working memory is not well defined. Work in non-human primates suggests that 5-HT2A receptor stimulation facilitates spatial working memory (Williams et al., 2002), and studies in rodents indicate that prefrontal serotonin depletion may exert negative effects on select aspects of working memory (Gonzalez-Burgos et al., 2012). However, many outstanding questions remain, in part because serotonergic influence on working memory may, in fact, not be direct but derive from interplay with other neurotransmitter systems (such as the cholinergic system) (Ohno and Watanabe 1997).
3.3.4. Stress effects on working memory
Human studies. Stress has been reported to have mixed effects on working memory in healthy humans. Thus, some studies reported impairments in working memory after acute stress (Luethi et al., 2008; Schoofs et al. 2008, 2009) or after acute administration of corticosteroids (Lupien et al., 1999). But other studies found no effect of stress (Porcelli et al., 2008) or even enhancement of working memory (Cornelisse et al., 2011b; Weerda et al., 2010). Some studies showed enhancement of working memory in males but no effect or impairments in females (Cornelisse et al., 2011b; Qin et al., 2009; Schoofs et al., 2013). One issue may be the use of widely different tasks to measure working memory, some relying on visual imagery, others on letter recognition or digit-based n-back tasks that may differentially recruit other executive functions such as sustained attention.
Another important factor influencing stress outcomes on working memory may be related to the timing of behavioral testing with respect to stress exposure. In a recent metanalysis of existing literature by (Geissler et al., 2023) the authors found that negative effects of stress on working memory are clustered within two timeframes of 0–9 min and 25–50 min post-stressor. The authors suggest these timeframes represent effects mediated by sympathetic nervous system activation (early phase) and cortisol effects (late phase).
Rodent studies. In rodents, stress has also been shown to produce different effects depending on the timing and stressor type (for a metanalysis see (Moreira et al., 2016) Acute stress was shown to improve working memory with a mechanism attributed to glucocorticoid receptor-dependent regulation of glutamate receptors in the prefrontal cortex in male rats (Yuen et al. 2009, 2011). Another group reported improved performance in male rats in the T-maze soon after application of acute footshock, but worse performance at later times of testing (Musazzi et al., 2019). In a study comparing male and female rats it was shown that 60 min restraint produced impairments in proestrus female rats but not male rats, whereas 2h restraint produced deficits in both sexes, leading the authors to conclude that high estrogen levels may render females more sensitive to the effects of stress on working memory (Shansky et al., 2006). With respect to chronic stress, some models have been shown to improve working memory, particularly in spatial working memory tests (see (Conrad 2010). However, chronic stress is also reported to impair working memory in male rats (Cerqueira et al., 2007a; Mika et al., 2012; Wright and Conrad 2008). In one study, one week of repeated restraint impaired working memory in male rats but not female rats (Wei et al., 2014). Early life stress has generally produced deficits in working memory in the male offspring (Banqueri et al., 2021; Brenhouse and Andersen 2011), and can exacerbate outcomes after additional insults (Sanchez et al., 2021; Viola et al., 2019). Some studies, however, did not observe the negative impacts of early stress on working memory in males but did observe slight deficits in females, again highlighting that stress outcomes are greatly nuanced and variable depending on protocols, sex, and age of testing (Sun et al., 2020).
4. Treatment strategies for cognitive impairment in stress-related psychiatric illness
This section will delve into current clinical interventions and novel therapeutic targets identified either preclinically or clinically for treatment to mitigate stress-induced cognitive dysfunctions. When appropriate, we discuss key preclinical mechanistic studies with translational potential. In other instances, such as the case concerning device-based neuromodulation approaches (section 4.2), we focus primarily on clinical studies.
4.1. Pharmacological treatments
4.1.1. Monoamine neurotransmission targeting drugs
As discussed in the previous sections, monoaminergic neurotransmission involving serotonin, norepinephrine and dopamine exert a critical role in brain circuits associated with cognitive functions.
Pehrson and colleagues (Pehrson et al., 2015) conducted an in-depth review of preclinical studies in rodents on monoamine neurotransmission-targeting drugs and their impact on cognition. Monoamine-targeting drugs, often referred to as conventional antidepressants, are the leading prescribed drugs to treat stress-related psychiatric disorders. While conventional antidepressants have shown varying degrees of clinical efficacy regarding mood improvement, their impact on cognitive symptoms remains negligible (Rosenblat et al., 2015; Shilyansky et al., 2016), except for vortioxetine (Bennabi et al., 2019), a multi-modal drug that is the only FDA-approved antidepressant for which the prescribing information mentions enhancements in cognitive domains.
4.1.1.1. Serotonergic system targeting drugs
As previously noted, the serotonergic system has crucial regulatory effects on different domains of cognitive function. Not surprisingly then, several preclinical studies have supported the use of serotonergic modulators to improve cognitive deficits. For example, CIC-induced deficits in reversal learning in male rats were rescued by acute and chronic treatment with the selective serotonin reuptake inhibitor (SSRI), citalopram (Danet et al., 2010). This effect was specific to the serotonin system because desipramine treatment (a norepinephrine reuptake blocker) did not rescue the CIC stress-induced reversal learning deficit (Danet et al., 2010). SSRIs such as citalopram, escitalopram and fluoxetine have also shown to be effective in ameliorating CUS-induced set-shifting deficits in male rats (Bondi et al., 2008; Minchew et al., 2021; Nikiforuk and Popik 2011).
Vortioxetine, an N-arylpiperazine derivative, is a potent selective inhibitor of the SERT, similar in this respect to other SSRIs. However, it also has potent actions at multiple 5-HT receptors (partial 5-HT1B agonism, 5-HT1A agonism, and 5-HT1D, 5-HT3, 5-HT7 antagonism), a property not shared by other SSRIs. In addition, vortioxetine can modulate the dopaminergic, noradrenergic, histaminergic, cholinergic, GABAergic, and glutamatergic systems (Pehrson et al., 2016). An in-depth review of preclinical and clinical findings implicating the antidepressant effects of vortioxetine was published by Sanchez and colleagues (Sanchez et al., 2015). Preclinical studies carried out in male rats have also shown that vortioxetine efficacy in rescuing 5-HT depletion-induced deficits in reversal learning is likely due to its action at postsynaptic 5-HT receptors, as PCPA-induced blockade of serotonin reuptake did not prevent the beneficial effects of vortioxetine (Wallace et al., 2014). In the same study, the authors also showed that chronic vortioxetine treatment rescued CIC-induced deficits in reversal learning. Vortioxetine has recently been shown to recover set-shifting deficits in male rats. (Vaiana et al., 2023). In a preclinical study relevant to the study of depression and anxiety-related disorders, vortioxetine mitigated the excessive display of conditioned fear and rescued adaptive coping behaviors compromised by chronic stress in male rats (Hatherall et al., 2017).
It is worth noting that, more recently, a promising target that has garnered attention in the realm of therapeutics for stress-related psychiatric disorders is the 5-HT4 receptors. As such, an extensive review of the role of 5-HT4 receptor pharmacology, its role in cognition, and its potential as therapeutics for stress-related psychiatric disorders has been published (Murphy et al., 2021).
4.1.1.2. Noradrenergic system targeting drugs
Lapiz and colleagues demonstrated that naïve male rats treated subchronically with the NE-reuptake blocker desipramine showed improvement in set-shifting correlated with upregulation of mPFC NE (Lapiz et al., 2007). Chronic treatment with milnacipran, a serotonin-norepinephrine uptake inhibitor, can also restore set-shifting compromised by stress (Craine et al., 2023; Naegeli et al., 2013). In male rats subject to CUS, chronic treatments with either the norepinephrine reuptake blocker desipramine or the SSRI escitalopram prevented the deficit in set-shifting induced by stress (Bondi et al. 2008, 2010). In male rats. acute postsynaptic α1 adrenergic receptor antagonism with benoxathian administration into the mPFC before testing prevented the beneficial effects of desipramine (Bondi et al., 2010), whereas infusions of β1 or β2 adrenergic receptor antagonists into the mPFC do not have a similar blocking effect (Lapiz and Morilak 2006). Furthermore, the same authors demonstrated that the systemic administration of atipamezole, an α2A-adrenergic receptor antagonist, enhances set-shifting in male rats. However, while the faciliatory effects of norepinephrine persisted during chronic stress, they became detrimental to reversal learning in male rats (Jett et al., 2013).
These data show that while antagonism of α2A-adrenergic receptor is associated with cognitive improvement (Bondi et al., 2007), it is essential to recognize that treatments targeting the modulatory role of α2A-adrenergic receptors in rescuing stress-induced cognitive deficits will vary as a function of the inverted-U shaped noradrenergic-mediated response to stress (Arnsten 2011, 2015; Berridge and Arnsten 2013).
4.1.1.2.1. The α2A-adrenergic receptor agonist guanfacine
Guanfacine is a selective and potent agonist of postsynaptic α2A-adrenergic receptors, capable of strengthening PFC network connections (Arnsten and Jin 2012; Arnsten 2020). Moreover, guanfacine treatment exhibits pro-cognitive effects across species (Arnsten 2020). In 2009, the FDA approved a long-acting formulation of the drug (Intuniv™) for the treatment of ADHD in children.
Mice with a single copy deletion of the neurofibromatosis 1 (NF1) gene, namely, NF1 (±) mice, are useful for the study of ADHD and associated deficits in inhibitory control, as NF1 mutations in humans are associated with ADHD in clinical populations (Hyman et al., 2005; Hyman et al., 2005) Using the delay discounting test as a proxy for choice impulsivity, NF1 (±) male mice displayed greater impulsivity when compared to the wild-type male mice. Treatment with guanfacine (0.3 mg/kg, i.p.) was effective in rescuing NF1 (±) mice deficits in choice impulsivity (Lukkes et al., 2020).
The repurposing of guanfacine for various indications linked to neurological and psychiatric disorders has increased significantly (Woolley 2023), bearing profound implications for the amelioration of cognitive deficits associated with cognitive dysfunction in a clinical population characterized as biotype 6 described in section 4.4.
4.1.2. Multiple neurotransmitter system targeting drug: modafinil success in the clinic
Modafinil is a prescription medication used to promote wakefulness and enhance cognitive function with a complex pharmacological profile, including the blockade of monoamines (noradrenaline, dopamine, and serotonin) as well as modulation of the glutamatergic and GABAergic system (Mereu et al., 2013). Modafinil is FDA-approved for use in individuals with multiple sleep-associated disorders, effectively aiding them in maintaining alertness during waking hours. In preclinical studies, acute but not chronic modafinil treatment promotes neurogenesis by increasing precursor cell proliferation and survival (Brandt et al., 2014). Modafinil treatment reversed stress-induced spatial memory deficits in rats (Alam and Choudhary 2023).In healthy individuals, short-term modafinil treatment improves cognition and attentional set-shifting (Turner et al., 2004). Schizophrenia-related cognitive impairments were also improved with acute modafinil treatment (Scoriels et al., 2013; Turner et al., 2004) but not with chronic administration (Sinkeviciute et al., 2018).
In remitted depressed patients with persistent cognitive symptoms, modafinil treatment significantly improved episodic and working memory, but no beneficial effects were detected on attention and planning strategies (Kaser et al., 2017). Furthermore, although systematic reviews and meta-analyses of the clinical use of augmentation drugs for depression show favorable results for modafinil (Goss et al., 2013; Nunez et al., 2022), clinical trials evaluating its effects on various cognitive domains are lacking.
4.1.3. Ketamine as a breakthrough leading to novel therapeutic modalities: glutamatergic and GABAergic system-targeting drugs
Ketamine, an NMDA receptor channel blocker, is a rapidly acting antidepressant with its (S)-isomer, (S)-ketamine (intranasal Spravato), approved by the FDA in 2019 for Treatment Resistant Depression (TRD). The fact that ketamine is mechanistically distinct from conventional monoamine-based antidepressants has stimulated research for drugs that would reproduce its rapid therapeutic effects associated with the improvement of depressive symptomatology without the confounds of its psychotomimetic and abuse-related properties.
The disinhibition hypothesis posits that ketamine initially blocks NMDA receptors on fast-spiking GABAergic neurons, thus promoting local inhibition of interneuron (Zanos and Gould 2018). Consequently, transient disinhibition of glutamatergic pyramidal cells occurs and rapidly induces AMPA receptor-mediated synaptic plasticity in relevant corticolimbic circuits. Furthermore, the disinhibition-induced plasticity of excitatory synapses weakened by chronic stress has become a therapeutic principle for achieving rapid (24 h after treatment) restoration of stress-induced deficits in reward sensitivity related to mood disorders (Thompson et al., 2015b).
In preclinical studies, ketamine has shown promising effects in correcting the effects of stress on executive functions. Ketamine administered 24 h before testing rescued deficits in set-shifting and reversal learning in stressed male rats (Jett et al., 2015). When ketamine was administered daily before stress, it prevented stress-induced deficits in set-shifting (Nikiforuk and Popik 2014). The prophylactic effects of ketamine have been reviewed recently (Evers et al., 2022). Moreover, a subanesthetic dose of ketamine in combination with an AMPAkine reduces premature responses in a motor impulsivity test in non-stressed rats (Davis-Reyes et al., 2021). Ketamine administration to TRD patients has been linked to improvements in executive function, visual memory, and working memory (Gill et al., 2021; Shiroma et al. 2014, 2022; Stippl et al., 2021). In addition, ketamine is associated with a reduction in suicidal ideation, with indications associating this effect to enhanced inhibitory control (Lee et al., 2016; Zhang and Ho 2016). It is worth noting that acute ketamine has been reported to also induce cognitive impairments (likely due to its psychotomimetic-inducing properties) in healthy but not in TRD patients. Specifically, in healthy volunteers, but not in TRD patients, ketamine administration was correlated with transient decreases in working memory, attention, and long-term memory measured up to 1 h post treatment (Harborne et al., 1996; Hetem et al., 2000; Morgan et al., 2004). Measures of functional connectivity in these cohorts revealed increases and decreases in frontostriatal functional connectivity in TRD and healthy individuals, respectively (Mkrtchian et al., 2021), reflecting the contrasting impacts of ketamine on executive function.
Given the side effects associated with the use of ketamine (Short et al., 2018), an alternative approach to promoting disinhibition of pyramidal neurons is to use subtype-selective modulation of benzodiazepine (BZ) site–containing GABAA receptors (GABAARs). Although α5-containing GABAARs (α5-GABAARs) comprise only 10% of total brain GABAARs, the expression of α5-GABAARs is enriched in the hippocampus and the prefrontal cortex. Selective a5-GABAAR negative allosteric modulators (a5-GABAAR-NAMs) such as L-655,708, aIA, and MRK-016 are being developed as potential cognitive enhancers with the idea of selectively targeting hippocampus and cortex (Atack 2011). Although selective a5-GABAAR-NAMs have been shown preclinically to improve memory in naïve rodents (Chambers et al., 2002; Street et al., 2004), they have not been extensively tested preclinically in stress-induced cognitive dysfunctions. Moreover, poor pharmacokinetics, toxicity issues, and low tolerance in older adults prevented further testing in humans (Atack 2010, 2011; Atack et al., 2009).
A more recently screened selective α5-GABAAR-NAM, Basmisanil, mitigated diazepam-induced hippocampal-dependent spatial learning impairment in male rats (assessed using the Morris water maze), and improved prefrontal cortex-mediated executive function in male non-human primates (evaluated using the object retrieval task in cynomolgus macaques) (Hipp et al., 2021). In addition, Basmisanil did not demonstrate anxiogenic or proconvulsant effects in male rats and Phase I clinical studies (Hipp et al., 2021). Therefore, Basmisanil possesses a favorable profile for exploring the potential therapeutic advantages of α5-GABAAR modulation.
In contrast to selective NAMs acting through the a5-GABAAR benzodiazepine binding site, S44819 is a recently identified GABA competitive antagonist at a5-GABAAR. It shares pro-cognitive effects with NAMs in preclinical studies, including inhibiting a5-GABAAR tonic current, enhancing LTP, reversing scopolamine-induced spatial working memory impairment, and improving object recognition memory (Etherington et al., 2017; Ling et al., 2015). Additional research is needed to explore its potential impact on stress-induced executive function deficits.
In addition to the beneficial effects of a5-GABAAR-NAMs discussed thus far, there is a body of work showing that selective positive allosteric modulators of the a5-GABAAR (a5-GABAAR-PAMs) have potential for the treatment of mood disorders as well as cognitive dysfunctions (Luscher et al., 2023; Prevot et al., 2019). Chronic treatment with the selective a5-GABAAR-PAM GL-II-73 rescued stress-induced and age-related working memory deficits in mice (Prevot et al., 2019). A putative reconciliation and working hypothesis explaining how a5-GABAAR-NAMs and a5-GABAAR-PAMs might exert similar outcomes has been proposed by Prevot and Sibille (Prevot and Sibille 2021) and is that both treatments increase signal-to-noise ratio of hippocampal and cortical transmission, although via distinct mechanism.
Many other compounds targeting the GABAergic system, collectively known as GABAkines, are currently being tested for the treatment of neurological and stress-induced psychiatric disorders (Cerne et al., 2022; Witkin et al., 2022; Zou et al., 2023).
4.2. Non-pharmacological treatments
4.2.1. Device-based neuromodulation
In contrast to pharmacological interventions, which predominantly target neurochemical communication between cells, device-based neuromodulation offers a distinct approach by selectively targeting specific components of neural cell activity, namely the membrane potentials in axons and dendrites. Device-based neuromodulation holds the potential to serve a dual role: first, as a valuable research tool, enabling the selective and non-invasive alteration of neural excitability to explore its effects on brain circuits and behavior; second, as an effective non-invasive therapeutic approach for stress-induced cognitive dysfunctions (Begemann et al., 2020). In this section, we describe clinical studies.
4.2.1.1. Transcranial magnetic stimulation (TMS)
During TMS, an alternating magnetic field produces weak electrical currents in the brain. The precise control of the magnetic field allows for non-invasive targeted stimulation, making it a valuable tool in neuroscience research and clinical practice. In 2008, TMS became FDA-approved for TRD. In 2017, deep TMS was approved as an adjunct treatment for OCD, and in 2020, for smoking cessation. The disadvantage of TMS is that it must be applied in a specialized clinical facility, as the equipment is large and expensive.
To test the hypothesis that impulsivity could be associated with poor weight management in obese patients, one group received deep TMS with active stimulation in the PFC and insula for 5 weeks, while the other received sham stimulation. The results showed a reduction of impulsivity in patients receiving deep TMS accompanied by significant improvement in body mass index, illustrating the effectiveness of deep TMS therapy in restoring the top-down inhibitory control of the PFC (Luzi et al., 2021).
A recent review evaluating the effectiveness of repetitive TMS (rTMS) to the PFC for TRD concludes that rTMS is reliable, well-tolerated, and efficacious (Adu et al., 2022). Nonetheless, although emerging evidence substantiates its short-term effectiveness in improving depressive symptomatology, there is a scarcity of literature supporting its long-term effects in cognitive symptom domains in TRD patients. Furthermore, to establish a standardized approach to rTMS implementation, further research is essential to explore variables such as frequency, intensity, pulse quantities, site of application, and inclusion of cognitive tests.
Regarding treatment for trauma-related disorders, a meta-analysis performed by Cirillo and colleagues (Cirillo et al., 2019) evaluating the effects of rTMS (ranging from 1 to 20 Hz) with traditional figure-of-eight coils to DLPFC indicates that rTMS is effective for PTSD and generalized anxiety disorders.
4.2.1.2. Transcranial direct current stimulation (tDCS)
tDCS is a safe way to non-invasively manipulate brain activity using weak electrical currents between two electrodes, one acting as a positive pole (anode) and the other as a negative pole (cathode) over the targeted cortical regions. The effects of tDCS will vary based on the polarity configuration, such that anodal stimulation promotes cell membrane depolarization, consequently increasing brain excitability. Cathodal stimulation has the opposite effect. The advantages of tDCS are portability and relatively low cost.
Evidence-based reviews of tDCS effectiveness in neurological and psychiatric disorders have been published (Fregni et al., 2021). Additionally, although the tDCS-induced current density is restricted primarily to the brain region surrounding the electrodes, the effects can be extended to distant neural networks, promoting broader connectivity-related effects (Ardolino et al., 2005; Cambiaghi et al., 2020). Researchers have increasingly used tDCS to investigate how cognitive and behavioral functions are linked to specific brain circuits. For example, applying anodal tDCS to the dorsolateral prefrontal cortex (DLPFC) in healthy individuals has been shown to improve planning (Dockery et al., 2009), working memory (Zaehle et al., 2011), attention (Stone and Tesche 2009), and reduced risk-taking behavior (Fecteau et al., 2007; Pripfl et al., 2013). Moreover, in patients with gambling disorder, anodal tDCS over the DLPFC improved decision-making and impulse control (Salatino et al., 2022). Another critical brain region with an established role in decision-making and impulse control is the OFC, and a small study in healthy individuals whereby anodal tDCS targeted the OFC showed improvement in decision-making and impulse control (Ouellet et al., 2015). A recent pilot study has established the feasibility of the use of tDCS over the OFC as a neuromodulatory non-invasive treatment of OCD (Fineberg et al., 2023).
In summary, non-invasive brain neuromodulation holds great promise regarding treatment options for cognitive dysfunctions. Recent research, with its emphasis on customizing brain stimulation to individual requirements, is pivotal in enhancing the effectiveness and safety of these techniques for all individuals. This personalized approach seeks to extend the advantages of brain stimulation to a broader spectrum of patients. A limited preliminary study suggested that anodal tDCS may improve extinction consolidation in patients with PTSD (Van't Wout et al., 2017).
4.2.2. Cognitive behavior therapy (CBT)
CBT encompasses a variety of techniques and strategies, including exposure therapy, aimed at identifying and modifying negative thought patterns and behaviors. Negatively biased cognitive inflexibility is present in both PTSD and depression, and it manifests as the automatic generation of thoughts, reactions, and interpretations consistently reinforced by avoidance behaviors.
Evidence-based exposure therapy treatments for PTSD revolve around reducing avoidance behaviors and actively engaging with traumatic memories. The cognitive element in the case of exposure therapy, where extinction plays a crucial role, is new learning-induced circuit-level plasticity, enabling cognitive flexibility and transitioning towards more adaptive and suitable behavioral responses. In a preclinical model of prolonged exposure therapy, one session of cue-conditioned fear extinction learning 24 h before testing rescued CUS-induced set-shifting impairments. Inhibition of protein synthesis within the IL mPFC during extinction prevented these behavioral effects, and the activity of IL mPFC glutamatergic neurons was necessary and sufficient for the therapeutic effects of extinction (Fucich et al. 2016, 2018; Paredes and Morilak 2019). At the circuit level, extinction reversed the CUS-induced attenuation of medial dorsal thalamus glutamatergic afferent-evoked electrical responses in IL mPFC (Fucich et al., 2018). While the ventral hippocampal afferent-evoked electrical responses in the IL mPFC were not affected by CUS, BDNF signaling within this pathway was necessary for the therapeutic effects of extinction (Paredes and Morilak 2023).
Although exposure therapy successfully improves PTSD-related cognitive dysfunctions (Nijdam and Vermetten 2018), a significant shortcoming is the high drop-out rate. Therefore, novel therapeutic approaches to enhance the effectiveness of exposure therapy are needed, as discussed in 4.2.3.
4.2.3. Therapy enhancement by adjunct treatment
Research evaluating novel therapeutic agents given in combination with well-established standard treatments, especially when patients have only partially responded, has gained attention in clinical and preclinical settings. The goal is to identify synergistic pairings that can enhance treatment effectiveness while minimizing the time it takes to achieve a response, and to minimize potential adverse effects or issues related to tolerability, to improve treatment results.
Krystal and Neumeister (Krystal and Neumeister 2009) proposed that combined exposure therapy and pharmacotherapy for PTSD act to enhance neuroplasticity, likely by activating neurotrophic factor signaling. In preclinical studies carried out in both male and female rats, combining a sub-therapeutic dose of ketamine with an abbreviated, sub-threshold extinction learning protocol completely reversed CUS-induced set-shifting deficits (Paredes et al., 2022). These outcomes may arise from molecular mechanisms shared by extinction and ketamine, such as enhanced neuronal plasticity in relevant circuitry (e.g., hippocampus-mPFC) or increased BDNF signaling (Paredes and Morilak 2023). Other recent research combining the entactogen phenethylamine 3,4-methylenedioxymethamphetamine (MDMA) with psychotherapy has shown improvement in PTSD symptoms Such studies suggest novel drugs that may be useful in combination with behavioral interventions to shorten treatment duration and potentially enhance efficacy (Bahji et al., 2020; Mitchell et al., 2023; Sessa et al., 2019).
4.3. Future directions and conclusions
One promising approach to achieve better treatment outcomes is associated with the characterization of biotypes of stress-related psychiatric disorders, made possible by recent advances in brain imaging and computation. Six biotypes of depression have been characterized, and each one of those biotypes are clustered based on distinct dysfunctions in large-scale brain connectivity amongst patients with varying degrees of symptomatology (Drysdale et al., 2017; Williams 2016). These clusters can provide valuable insights into clinical outcomes following treatment interventions. Of relevance to this review, biotype 6 of depression, also known as the cognitive dysfunction biotype, is characterized by disruption of cognitive control circuitry connectivity (heavily linked to the DLPFC/ACC/Precentral Gyrus/Dorsal Parietal Cortex) (Hack et al., 2023; Williams 2016), and no response to SSRIs (Tozzi et al., 2020). A clinical trial-guided study is currently in Phase 4, testing the drug guanfacine, which mechanism of action was discussed in 4.1.1.2, as a novel therapy for cognitive impairment in biotype 6 of depression, targeting the DLPFC (NCT04181736).
Therefore, assessment of individual brain connectivity maps may inform predictions regarding treatment response, facilitating selection of more tailored and effective treatments for patients with varying degrees of executive function impairment.
In summary, the field is confronted with several crucial challenges.
-
1.
Mechanistic Insights into Subtypes: There is a need to develop a deeper mechanistic understanding of various subtypes of stress-related psychiatric disorders to pave the way to develop precise individualized therapeutic strategies. Preclinically, such insights will serve as a testing ground for tailored interventions.
-
2.
Emphasis on Prediction and Prevention: Shifting the focus toward then predicting and preventing stress-related cognitive dysfunction is a crucial challenge. Identifying subtypes and risk factors, and strategies to mitigate them, the field can lead to significant strides in reducing the overall burden of these conditions.
These challenges underscore the need for multidisciplinary complementary research combining reductionist science, personalized medicine, and validated animal models to address stress-related cognitive dysfunctions and develop strategies to manage them before they progress to clinical disorder.
Funding
This work was supported by the National Institute of Mental Health (research grant R01MH053851), by the US Department of Veterans Affairs Biomedical Laboratory Research and Development Program (merit award I01BX000559, and grant I01BX003512) and by a UTH Center for Biomedical Neuroscience Pilot Project Grant. The contents of this paper do not represent the views of the Department of Veterans Affairs or the US Government.
CRediT authorship contribution statement
Milena Girotti: Writing – review & editing, Writing – original draft, Conceptualization. Sarah E. Bulin: Writing – review & editing, Writing – original draft, Conceptualization. Flavia R. Carreno: Writing – review & editing, Writing – original draft, Conceptualization.
Declaration of competing interest
The authors have nothing to declare.
Handling Editor: Rita Valentino
References
- Abdul-Monim Z., Neill J.C., Reynolds G.P. Sub-chronic psychotomimetic phencyclidine induces deficits in reversal learning and alterations in parvalbumin-immunoreactive expression in the rat. J. Psychopharmacol. 2007;21:198–205. doi: 10.1177/0269881107067097. [DOI] [PubMed] [Google Scholar]
- Abela A.R., Dougherty S.D., Fagen E.D., Hill C.J., Chudasama Y. Inhibitory control deficits in rats with ventral hippocampal lesions. Cereb Cortex. 2013;23:1396–1409. doi: 10.1093/cercor/bhs121. [DOI] [PubMed] [Google Scholar]
- Acosta G.B., Otero Losada M.E., Rubio M.C. Area-dependent changes in GABAergic function after acute and chronic cold stress. Neurosci. Lett. 1993;154:175–178. doi: 10.1016/0304-3940(93)90200-5. [DOI] [PubMed] [Google Scholar]
- Adam E.M., Johns T., Sur M. Dynamic control of visually guided locomotion through corticosubthalamic projections. Cell Rep. 2022;40 doi: 10.1016/j.celrep.2022.111139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adams T.G., Kelmendi B., Brake C.A., Gruner P., Badour C.L., Pittenger C. The role of stress in the pathogenesis and maintenance of obsessive-compulsive disorder. Chronic Stress (Thousand Oaks) 2018;2 doi: 10.1177/2470547018758043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adler S.M., Girotti M., Morilak D.A. Optogenetically-induced long term depression in the rat orbitofrontal cortex ameliorates stress-induced reversal learning impairment. Neurobiol Stress. 2020;13 doi: 10.1016/j.ynstr.2020.100258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adu M.K., Shalaby R., Chue P., Agyapong V.I.O. Repetitive transcranial magnetic stimulation for the treatment of resistant depression: a scoping review. Behav. Sci. 2022;12 doi: 10.3390/bs12060195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alam N., Choudhary K. Neurochemical effects of methylphenidate and modafinil in ameliorating stress-induced cognitive deficits. ACS Pharmacol. Transl. Sci. 2023;6:1357–1372. doi: 10.1021/acsptsci.3c00077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Albasser M.M., Amin E., Lin T.C., Iordanova M.D., Aggleton J.P. Evidence that the rat hippocampus has contrasting roles in object recognition memory and object recency memory. Behav. Neurosci. 2012;126:659–669. doi: 10.1037/a0029754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander J.K., Hillier A., Smith R.M., Tivarus M.E., Beversdorf D.Q. Beta-adrenergic modulation of cognitive flexibility during stress. J Cogn Neurosci. 2007;19:468–478. doi: 10.1162/jocn.2007.19.3.468. [DOI] [PubMed] [Google Scholar]
- Anderson R.M., Johnson S.B., Lingg R.T., Hinz D.C., Romig-Martin S.A., Radley J.J. Evidence for similar prefrontal structural and functional alterations in male and female rats following chronic stress or glucocorticoid exposure. Cereb Cortex. 2020;30:353–370. doi: 10.1093/cercor/bhz092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Apud J.A., Mattay V., Chen J., Kolachana B.S., Callicott J.H., et al. Tolcapone improves cognition and cortical information processing in normal human subjects. Neuropsychopharmacology. 2007;32:1011–1020. doi: 10.1038/sj.npp.1301227. [DOI] [PubMed] [Google Scholar]
- Ardolino G., Bossi B., Barbieri S., Priori A. Non-synaptic mechanisms underlie the after-effects of cathodal transcutaneous direct current stimulation of the human brain. J Physiol. 2005;568:653–663. doi: 10.1113/jphysiol.2005.088310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnsten A.F. Stress weakens prefrontal networks: molecular insults to higher cognition. Nat. Neurosci. 2015;18:1376–1385. doi: 10.1038/nn.4087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnsten A.F., Cai J.X., Goldman-Rakic P.S. The alpha-2 adrenergic agonist guanfacine improves memory in aged monkeys without sedative or hypotensive side effects: evidence for alpha-2 receptor subtypes. J. Neurosci. 1988;8:4287–4298. doi: 10.1523/JNEUROSCI.08-11-04287.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnsten A.F., Jin L.E. Guanfacine for the treatment of cognitive disorders: a century of discoveries at Yale. Yale J. Biol. Med. 2012;85:45–58. [PMC free article] [PubMed] [Google Scholar]
- Arnsten A.F.T. Catecholamine influences on dorsolateral prefrontal cortical networks. Biol Psychiatry. 2011;69:e89–e99. doi: 10.1016/j.biopsych.2011.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnsten A.F.T. Guanfacine's mechanism of action in treating prefrontal cortical disorders: Successful translation across species. Neurobiol. Learn. Mem. 2020;176 doi: 10.1016/j.nlm.2020.107327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aron A.R., Fletcher P.C., Bullmore E.T., Sahakian B.J., Robbins T.W. Stop-signal inhibition disrupted by damage to right inferior frontal gyrus in humans. Nat. Neurosci. 2003;6:115–116. doi: 10.1038/nn1003. [DOI] [PubMed] [Google Scholar]
- Aron A.R., Herz D.M., Brown P., Forstmann B.U., Zaghloul K. Frontosubthalamic circuits for control of action and cognition. J. Neurosci. 2016;36:11489–11495. doi: 10.1523/JNEUROSCI.2348-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atack J.R. Preclinical and clinical pharmacology of the GABAA receptor alpha5 subtype-selective inverse agonist alpha5IA. Pharmacol. Ther. 2010;125:11–26. doi: 10.1016/j.pharmthera.2009.09.001. [DOI] [PubMed] [Google Scholar]
- Atack J.R. GABAA receptor subtype-selective modulators. II. alpha5-selective inverse agonists for cognition enhancement. Curr. Top. Med. Chem. 2011;11:1203–1214. doi: 10.2174/156802611795371314. [DOI] [PubMed] [Google Scholar]
- Atack J.R., Maubach K.A., Wafford K.A., O'Connor D., Rodrigues A.D., et al. In vitro and in vivo properties of 3-tert-butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA receptor alpha5 subtype-selective inverse agonist. J Pharmacol Exp Ther. 2009;331:470–484. doi: 10.1124/jpet.109.157636. [DOI] [PubMed] [Google Scholar]
- Auger M.L., Floresco S.B. Prefrontal cortical GABA modulation of spatial reference and working memory. Int. J. Neuropsychopharmacol. 2014;18 doi: 10.1093/ijnp/pyu013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aura J., Riekkinen P., Jr. Blockade of NMDA receptors located at the dorsomedial prefrontal cortex impairs spatial working memory in rats. Neuroreport. 1999;10:243–248. doi: 10.1097/00001756-199902050-00008. [DOI] [PubMed] [Google Scholar]
- Baddeley A. Working memory. Science. 1992;255:556–559. doi: 10.1126/science.1736359. [DOI] [PubMed] [Google Scholar]
- Bahji A., Forsyth A., Groll D., Hawken E.R. Efficacy of 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for posttraumatic stress disorder: a systematic review and meta-analysis. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2020;96 doi: 10.1016/j.pnpbp.2019.109735. [DOI] [PubMed] [Google Scholar]
- Banasr M., Valentine G.W., Li X.-Y., Gourley S.L., Taylor J.R., Duman R.S. Chronic unpredictable stress decreases cell proliferation in the cerebral cortex of the adult rat. Biol Psychiatry. 2007;62:496–504. doi: 10.1016/j.biopsych.2007.02.006. [DOI] [PubMed] [Google Scholar]
- Bangasser D.A., Kawasumi Y. Cognitive disruptions in stress-related psychiatric disorders: a role for corticotropin releasing factor (CRF) Horm. Behav. 2015;76:125–135. doi: 10.1016/j.yhbeh.2015.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banqueri M., Gutierrez-Menendez A., Mendez M., Conejo N.M., Arias J.L. Early life stress due to repeated maternal separation alters the working memory acquisition brain functional network. Stress. 2021;24:87–95. doi: 10.1080/10253890.2020.1777974. [DOI] [PubMed] [Google Scholar]
- Bardin L., Malfetes N., Newman-Tancredi A., Depoortere R. Chronic restraint stress induces mechanical and cold allodynia, and enhances inflammatory pain in rat: relevance to human stress-associated painful pathologies. Behav. Brain Res. 2009;205:360–366. doi: 10.1016/j.bbr.2009.07.005. [DOI] [PubMed] [Google Scholar]
- Bari A., Dalley J.W., Robbins T.W. The application of the 5-choice serial reaction time task for the assessment of visual attentional processes and impulse control in rats. Nat. Protoc. 2008;3:759–767. doi: 10.1038/nprot.2008.41. [DOI] [PubMed] [Google Scholar]
- Bari A., Robbins T.W. Inhibition and impulsivity: behavioral and neural basis of response control. Prog Neurobiol. 2013;108:44–79. doi: 10.1016/j.pneurobio.2013.06.005. [DOI] [PubMed] [Google Scholar]
- Barker G.R., Warburton E.C. When is the hippocampus involved in recognition memory? J. Neurosci. 2011;31:10721–10731. doi: 10.1523/JNEUROSCI.6413-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bauduin S., van der Pal Z., Pereira A.M., Meijer O.C., Giltay E.J., et al. Cortical thickness abnormalities in long-term remitted Cushing's disease. Transl. Psychiatry. 2020;10:293. doi: 10.1038/s41398-020-00980-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baunez C., Robbins T.W. Bilateral lesions of the subthalamic nucleus induce multiple deficits in an attentional task in rats. Eur. J. Neurosci. 1997;9:2086–2099. doi: 10.1111/j.1460-9568.1997.tb01376.x. [DOI] [PubMed] [Google Scholar]
- Begemann M.J., Brand B.A., Curcic-Blake B., Aleman A., Sommer I.E. Efficacy of non-invasive brain stimulation on cognitive functioning in brain disorders: a meta-analysis. Psychol. Med. 2020;50:2465–2486. doi: 10.1017/S0033291720003670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ben-Zion Z., Fine N.B., Keynan N.J., Admon R., Green N., et al. Cognitive flexibility predicts PTSD symptoms: observational and interventional studies. Front Psychiatry. 2018;9:477. doi: 10.3389/fpsyt.2018.00477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennabi D., Haffen E., Van Waes V. Vortioxetine for cognitive enhancement in major depression: from animal models to clinical research. Front Psychiatry. 2019;10:771. doi: 10.3389/fpsyt.2019.00771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bermond B., Surachno S., Lok A., ten Berge I.J., Plasmans B., et al. Memory functions in prednisone-treated kidney transplant patients. Clin. Transplant. 2005;19:512–517. doi: 10.1111/j.1399-0012.2005.00376.x. [DOI] [PubMed] [Google Scholar]
- Bermudez-Rattoni F. The forgotten insular cortex: its role on recognition memory formation. Neurobiol. Learn. Mem. 2014;109:207–216. doi: 10.1016/j.nlm.2014.01.001. [DOI] [PubMed] [Google Scholar]
- Bernosky-Smith K.A., Qiu Y.Y., Feja M., Lee Y.B., Loughlin B., et al. Ventral tegmental area D2 receptor knockdown enhances choice impulsivity in a delay-discounting task in rats. Behav. Brain Res. 2018;341:129–134. doi: 10.1016/j.bbr.2017.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berridge C.W., Arnsten A.F. Psychostimulants and motivated behavior: arousal and cognition. Neurosci. Biobehav. Rev. 2013;37:1976–1984. doi: 10.1016/j.neubiorev.2012.11.005. [DOI] [PubMed] [Google Scholar]
- Besson M., Belin D., McNamara R., Theobald D.E., Castel A., et al. Dissociable control of impulsivity in rats by dopamine d2/3 receptors in the core and shell subregions of the nucleus accumbens. Neuropsychopharmacology. 2010;35:560–569. doi: 10.1038/npp.2009.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birrell J.M., Brown V.J. Medial frontal cortex mediates perceptual attentional set shifting in the rat. J. Neurosci. 2000;20:4320–4324. doi: 10.1523/JNEUROSCI.20-11-04320.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bissonette G.B., Martins G.J., Franz T.M., Harper E.S., Schoenbaum G., Powell E.M. Double dissociation of the effects of medial and orbital prefrontal cortical lesions on attentional and affective shifts in mice. J. Neurosci. 2008;28:11124–11130. doi: 10.1523/JNEUROSCI.2820-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bizot J., Le Bihan C., Puech A.J., Hamon M., Thiebot M. Serotonin and tolerance to delay of reward in rats. Psychopharmacology (Berl) 1999;146:400–412. doi: 10.1007/pl00005485. [DOI] [PubMed] [Google Scholar]
- Boehme R., Lorenz R.C., Gleich T., Romund L., Pelz P., et al. Reversal learning strategy in adolescence is associated with prefrontal cortex activation. Eur. J. Neurosci. 2017;45:129–137. doi: 10.1111/ejn.13401. [DOI] [PubMed] [Google Scholar]
- Bogdan R., Pizzagalli D.A. Acute stress reduces reward responsiveness: implications for depression. Biol Psychiatry. 2006;60:1147–1154. doi: 10.1016/j.biopsych.2006.03.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolkan S.S., Stujenske J.M., Parnaudeau S., Spellman T.J., Rauffenbart C., et al. Thalamic projections sustain prefrontal activity during working memory maintenance. Nat. Neurosci. 2017;20:987–996. doi: 10.1038/nn.4568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bondi C.O., Barrera G., Lapiz M.D.S., Bédard T., Mahan A., Morilak D.A. Noradrenergic facilitation of shock-probe defensive burying in lateral septum of rats, and modulation by chronic treatment with desipramine. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2007;31:482–495. doi: 10.1016/j.pnpbp.2006.11.015. [DOI] [PubMed] [Google Scholar]
- Bondi C.O., Jett J.D., Morilak D.A. Beneficial effects of desipramine on cognitive function of chronically stressed rats are mediated by alpha1-adrenergic receptors in medial prefrontal cortex. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2010;34:913–923. doi: 10.1016/j.pnpbp.2010.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bondi C.O., Rodriguez G., Gould G.G., Frazer A., Morilak D.A. Chronic unpredictable stress induces a cognitive deficit and anxiety-like behavior in rats that is prevented by chronic antidepressant drug treatment. Neuropsychopharmacology. 2008;33:320–331. doi: 10.1038/sj.npp.1301410. [DOI] [PubMed] [Google Scholar]
- Boulougouris V., Castane A., Robbins T.W. Dopamine D2/D3 receptor agonist quinpirole impairs spatial reversal learning in rats: investigation of D3 receptor involvement in persistent behavior. Psychopharmacology (Berl) 2009;202:611–620. doi: 10.1007/s00213-008-1341-2. [DOI] [PubMed] [Google Scholar]
- Boulougouris V., Glennon J.C., Robbins T.W. Dissociable effects of selective 5-HT2A and 5-HT2C receptor antagonists on serial spatial reversal learning in rats. Neuropsychopharmacology. 2008;33:2007–2019. doi: 10.1038/sj.npp.1301584. [DOI] [PubMed] [Google Scholar]
- Bourdeau I., Bard C., Noel B., Leclerc I., Cordeau M.P., et al. Loss of brain volume in endogenous Cushing's syndrome and its reversibility after correction of hypercortisolism. J. Clin. Endocrinol. Metab. 2002;87:1949–1954. doi: 10.1210/jcem.87.5.8493. [DOI] [PubMed] [Google Scholar]
- Brady A.M., Floresco S.B. Operant procedures for assessing behavioral flexibility in rats. J. Vis. Exp. 2015 doi: 10.3791/52387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brandt M.D., Ellwardt E., Storch A. Short- and long-term treatment with modafinil differentially affects adult hippocampal neurogenesis. Neuroscience. 2014;278:267–275. doi: 10.1016/j.neuroscience.2014.08.014. [DOI] [PubMed] [Google Scholar]
- Brenhouse H.C., Andersen S.L. Nonsteroidal anti-inflammatory treatment prevents delayed effects of early life stress in rats. Biol Psychiatry. 2011;70:434–440. doi: 10.1016/j.biopsych.2011.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brigman J.L., Daut R.A., Wright T., Gunduz-Cinar O., Graybeal C., et al. GluN2B in corticostriatal circuits governs choice learning and choice shifting. Nat. Neurosci. 2013;16:1101–1110. doi: 10.1038/nn.3457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown E.S., Jw D., Frol A., Bobadilla L., Khan D.A., et al. Hippocampal volume, spectroscopy, cognition, and mood in patients receiving corticosteroid therapy. Biol Psychiatry. 2004;55:538–545. doi: 10.1016/j.biopsych.2003.09.010. [DOI] [PubMed] [Google Scholar]
- Brown S.M., Henning S., Wellman C.L. Mild, short-term stress alters dendritic morphology in rat medial prefrontal cortex. Cereb Cortex. 2005;15:1714–1722. doi: 10.1093/cercor/bhi048. [DOI] [PubMed] [Google Scholar]
- Brozoski T.J., Brown R.M., Rosvold H.E., Goldman P.S. Cognitive deficit caused by regional depletion of dopamine in prefrontal cortex of rhesus monkey. Science. 1979;205:929–932. doi: 10.1126/science.112679. [DOI] [PubMed] [Google Scholar]
- Bryce C.A., Howland J.G. Stress facilitates late reversal learning using a touchscreen-based visual discrimination procedure in male Long Evans rats. Behav. Brain Res. 2015;278:21–28. doi: 10.1016/j.bbr.2014.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buhusi M., Olsen K., Buhusi C.V. Increased temporal discounting after chronic stress in CHL1-deficient mice is reversed by 5-HT2C agonist Ro 60-0175. Neuroscience. 2017;357:110–118. doi: 10.1016/j.neuroscience.2017.05.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bulin S.E., Hohl K.M., Paredes D., Silva J.D., Morilak D.A. Bidirectional optogenetically-induced plasticity of evoked responses in the rat medial prefrontal cortex can impair or enhance cognitive set-shifting. eNeuro. 2020;7 doi: 10.1523/ENEURO.0363-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butts K.A., Floresco S.B., Phillips A.G. Acute stress impairs set-shifting but not reversal learning. Behav. Brain Res. 2013;252:222–229. doi: 10.1016/j.bbr.2013.06.007. [DOI] [PubMed] [Google Scholar]
- Buynitsky T., Mostofsky D.I. Restraint stress in biobehavioral research: recent developments. Neurosci. Biobehav. Rev. 2009;33:1089–1098. doi: 10.1016/j.neubiorev.2009.05.004. [DOI] [PubMed] [Google Scholar]
- Bymaster F.P., Katner J.S., Nelson D.L., Hemrick-Luecke S.K., Threlkeld P.G., et al. Atomoxetine increases extracellular levels of norepinephrine and dopamine in prefrontal cortex of rat: a potential mechanism for efficacy in attention deficit/hyperactivity disorder. Neuropsychopharmacology. 2002;27:699–711. doi: 10.1016/S0893-133X(02)00346-9. [DOI] [PubMed] [Google Scholar]
- Cambiaghi M., Buffelli M., Masin L., Valtorta F., Comai S. Transcranial direct current stimulation of the mouse prefrontal cortex modulates serotonergic neural activity of the dorsal raphe nucleus. Brain Stimul. 2020;13:548–550. doi: 10.1016/j.brs.2020.01.012. [DOI] [PubMed] [Google Scholar]
- Canale N., Rubaltelli E., Vieno A., Pittarello A., Billieux J. Impulsivity influences betting under stress in laboratory gambling. Sci. Rep. 2017;7 doi: 10.1038/s41598-017-10745-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caprioli D., Hong Y.T., Sawiak S.J., Ferrari V., Williamson D.J., et al. Baseline-dependent effects of cocaine pre-exposure on impulsivity and D2/3 receptor availability in the rat striatum: possible relevance to the attention-deficit hyperactivity syndrome. Neuropsychopharmacology. 2013;38:1460–1471. doi: 10.1038/npp.2013.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caprioli D., Sawiak S.J., Merlo E., Theobald D.E., Spoelder M., et al. Gamma aminobutyric acidergic and neuronal structural markers in the nucleus accumbens core underlie trait-like impulsive behavior. Biol Psychiatry. 2014;75:115–123. doi: 10.1016/j.biopsych.2013.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardinal R.N., Pennicott D.R., Sugathapala C.L., Robbins T.W., Everitt B.J. Impulsive choice induced in rats by lesions of the nucleus accumbens core. Science. 2001;292:2499–2501. doi: 10.1126/science.1060818. [DOI] [PubMed] [Google Scholar]
- Carli M., Robbins T.W., Evenden J.L., Everitt B.J. Effects of lesions to ascending noradrenergic neurones on performance of a 5-choice serial reaction task in rats; implications for theories of dorsal noradrenergic bundle function based on selective attention and arousal. Behav. Brain Res. 1983;9:361–380. doi: 10.1016/0166-4328(83)90138-9. [DOI] [PubMed] [Google Scholar]
- Carrier M., Simoncicova E., St-Pierre M.K., McKee C., Tremblay M.E. Psychological stress as a risk factor for accelerated cellular aging and cognitive decline: the involvement of microglia-neuron crosstalk. Front. Mol. Neurosci. 2021;14 doi: 10.3389/fnmol.2021.749737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerne R., Lippa A., Poe M.M., Smith J.L., Jin X., et al. GABAkines - advances in the discovery, development, and commercialization of positive allosteric modulators of GABA(A) receptors. Pharmacol. Ther. 2022;234 doi: 10.1016/j.pharmthera.2021.108035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerqueira J.J., Catania C., Sotiropoulos I., Schubert M., Kalisch R., et al. Corticosteroid status influences the volume of the rat cingulate cortex - a magnetic resonance imaging study. J. Psychiatr. Res. 2005;39:451–460. doi: 10.1016/j.jpsychires.2005.01.003. [DOI] [PubMed] [Google Scholar]
- Cerqueira J.J., Mailliet F., Almeida O.F., Jay T.M., Sousa N. The prefrontal cortex as a key target of the maladaptive response to stress. J. Neurosci. 2007;27:2781–2787. doi: 10.1523/JNEUROSCI.4372-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerqueira J.J., Pego J.M., Taipa R., Bessa J.M., Almeida O.F., Sousa N. Morphological correlates of corticosteroid-induced changes in prefrontal cortex-dependent behaviors. J. Neurosci. 2005;25:7792–7800. doi: 10.1523/JNEUROSCI.1598-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerqueira J.J., Taipa R., Uylings H.B., Almeida O.F., Sousa N. Specific configuration of dendritic degeneration in pyramidal neurons of the medial prefrontal cortex induced by differing corticosteroid regimens. Cereb Cortex. 2007;17:1998–2006. doi: 10.1093/cercor/bhl108. [DOI] [PubMed] [Google Scholar]
- Chambers M.S., Atack J.R., Bromidge F.A., Broughton H.B., Cook S., et al. 6,7-Dihydro-2-benzothiophen-4(5H)-ones: a novel class of GABA-A alpha5 receptor inverse agonists. J. Med. Chem. 2002;45:1176–1179. doi: 10.1021/jm010471b. [DOI] [PubMed] [Google Scholar]
- Chao O.Y., Nikolaus S., Yang Y.M., Huston J.P. Neuronal circuitry for recognition memory of object and place in rodent models. Neurosci. Biobehav. Rev. 2022;141 doi: 10.1016/j.neubiorev.2022.104855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y., Kramar E.A., Chen L.Y., Babayan A.H., Andres A.L., et al. Impairment of synaptic plasticity by the stress mediator CRH involves selective destruction of thin dendritic spines via RhoA signaling. Mol Psychiatry. 2013;18:485–496. doi: 10.1038/mp.2012.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chudasama Y., Baunez C., Robbins T.W. Functional disconnection of the medial prefrontal cortex and subthalamic nucleus in attentional performance: evidence for corticosubthalamic interaction. J. Neurosci. 2003;23:5477–5485. doi: 10.1523/JNEUROSCI.23-13-05477.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chudasama Y., Bussey T.J., Muir J.L. Effects of selective thalamic and prelimbic cortex lesions on two types of visual discrimination and reversal learning. Eur. J. Neurosci. 2001;14:1009–1020. doi: 10.1046/j.0953-816x.2001.01607.x. [DOI] [PubMed] [Google Scholar]
- Chudasama Y., Passetti F., Rhodes S.E., Lopian D., Desai A., Robbins T.W. Dissociable aspects of performance on the 5-choice serial reaction time task following lesions of the dorsal anterior cingulate, infralimbic and orbitofrontal cortex in the rat: differential effects on selectivity, impulsivity and compulsivity. Behav. Brain Res. 2003;146:105–119. doi: 10.1016/j.bbr.2003.09.020. [DOI] [PubMed] [Google Scholar]
- Cirillo P., Gold A.K., Nardi A.E., Ornelas A.C., Nierenberg A.A., et al. Transcranial magnetic stimulation in anxiety and trauma-related disorders: a systematic review and meta-analysis. Brain Behav. 2019;9 doi: 10.1002/brb3.1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clancy F., Prestwich A., Caperon L., O'Connor D.B. Perseverative cognition and health behaviors: a systematic review and meta-analysis. Front. Hum. Neurosci. 2016;10:534. doi: 10.3389/fnhum.2016.00534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke H.F., Dalley J.W., Crofts H.S., Robbins T.W., Roberts A.C. Cognitive inflexibility after prefrontal serotonin depletion. Science. 2004;304:878–880. doi: 10.1126/science.1094987. [DOI] [PubMed] [Google Scholar]
- Clarke H.F., Hill G.J., Robbins T.W., Roberts A.C. Dopamine, but not serotonin, regulates reversal learning in the marmoset caudate nucleus. J. Neurosci. 2011;31:4290–4297. doi: 10.1523/JNEUROSCI.5066-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke H.F., Walker S.C., Crofts H.S., Dalley J.W., Robbins T.W., Roberts A.C. Prefrontal serotonin depletion affects reversal learning but not attentional set shifting. J. Neurosci. 2005;25:532–538. doi: 10.1523/JNEUROSCI.3690-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke H.F., Walker S.C., Dalley J.W., Robbins T.W., Roberts A.C. Cognitive inflexibility after prefrontal serotonin depletion is behaviorally and neurochemically specific. Cerebr. Cortex. 2007;17:18–27. doi: 10.1093/cercor/bhj120. [DOI] [PubMed] [Google Scholar]
- Clatworthy P.L., Lewis S.J., Brichard L., Hong Y.T., Izquierdo D., et al. Dopamine release in dissociable striatal subregions predicts the different effects of oral methylphenidate on reversal learning and spatial working memory. J. Neurosci. 2009;29:4690–4696. doi: 10.1523/JNEUROSCI.3266-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cole M.A., Kalman B.A., Pace T.W.W., Topczewski F., Lowrey M.J., Spencer R.L. Selective blockade of the mineralocorticoid receptor impairs hypothalamic-pituitary-adrenal axis expression of habituation. J. Neuroendocrinol. 2000;12:1034–1042. doi: 10.1046/j.1365-2826.2000.00555.x. [DOI] [PubMed] [Google Scholar]
- Coluccia D., Wolf O.T., Kollias S., Roozendaal B., Forster A., de Quervain D.J. Glucocorticoid therapy-induced memory deficits: acute versus chronic effects. J. Neurosci. 2008;28:3474–3478. doi: 10.1523/JNEUROSCI.4893-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conrad C.D. A critical review of chronic stress effects on spatial learning and memory. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2010;34:742–755. doi: 10.1016/j.pnpbp.2009.11.003. [DOI] [PubMed] [Google Scholar]
- Cook S.C., Wellman C.L. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. J. Neurobiol. 2004;60:236–248. doi: 10.1002/neu.20025. [DOI] [PubMed] [Google Scholar]
- Cools R., Clark L., Owen A.M., Robbins T.W. Defining the neural mechanisms of probabilistic reversal learning using event-related functional magnetic resonance imaging. J. Neurosci. 2002;22:4563–4567. doi: 10.1523/JNEUROSCI.22-11-04563.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cope Z.A., Vazey E.M., Floresco S.B., Aston Jones G.S. DREADD-mediated modulation of locus coeruleus inputs to mPFC improves strategy set-shifting. Neurobiol. Learn. Mem. 2019;161:1–11. doi: 10.1016/j.nlm.2019.02.009. [DOI] [PubMed] [Google Scholar]
- Cornelisse S., Joels M., Smeets T. A randomized trial on mineralocorticoid receptor blockade in men: effects on stress responses, selective attention, and memory. Neuropsychopharmacology. 2011;36:2720–2728. doi: 10.1038/npp.2011.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cornelisse S., van Stegeren A.H., Joels M. Implications of psychosocial stress on memory formation in a typical male versus female student sample. Psychoneuroendocrinology. 2011;36:569–578. doi: 10.1016/j.psyneuen.2010.09.002. [DOI] [PubMed] [Google Scholar]
- Correll C.M., Rosenkranz J.A., Grace A.A. Chronic cold stress alters prefrontal cortical modulation of amygdala neuronal activity in rats. Biol Psychiatry. 2005;58:382–391. doi: 10.1016/j.biopsych.2005.04.009. [DOI] [PubMed] [Google Scholar]
- Costa V.D., Tran V.L., Turchi J., Averbeck B.B. Reversal learning and dopamine: a bayesian perspective. J. Neurosci. 2015;35:2407–2416. doi: 10.1523/JNEUROSCI.1989-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Courtney S.M., Petit L., Maisog J.M., Ungerleider L.G., Haxby J.V. An area specialized for spatial working memory in human frontal cortex. Science. 1998;279:1347–1351. doi: 10.1126/science.279.5355.1347. [DOI] [PubMed] [Google Scholar]
- Craine T.J., Race N.S., Kutash L.A., Iouchmanov A.L., Moschonas E.H., et al. Milnacipran ameliorates executive function impairments following frontal lobe traumatic brain injury in male rats: a multimodal behavioral assessment. J. Neurotrauma. 2023;40:112–124. doi: 10.1089/neu.2022.0289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crean J., Richards J.B., de Wit H. Effect of tryptophan depletion on impulsive behavior in men with or without a family history of alcoholism. Behav. Brain Res. 2002;136:349–357. doi: 10.1016/s0166-4328(02)00132-8. [DOI] [PubMed] [Google Scholar]
- Crofts H.S., Dalley J.W., Collins P., Van Denderen J.C., Everitt B.J., et al. Differential effects of 6-OHDA lesions of the frontal cortex and caudate nucleus on the ability to acquire an attentional set. Cerebr. Cortex. 2001;11:1015–1026. doi: 10.1093/cercor/11.11.1015. [DOI] [PubMed] [Google Scholar]
- Curtis A.L., Lechner S.M., Pavcovich L.A., Valentino R.J. Activation of the locus coeruleus noradrenergic system by intracoerulear microinfusion of corticotropin-releasing factor: effects on discharge rate, cortical norepinephrine levels and cortical electroencephalographic activity. J Pharmacol Exp Ther. 1997;281:163–172. [PubMed] [Google Scholar]
- Dagher A. Mapping the hyper-direct circuitry of impulsivity. Brain. 2020;143:1973–1974. doi: 10.1093/brain/awaa187. [DOI] [PubMed] [Google Scholar]
- Dalley J.W., Everitt B.J., Robbins T.W. Impulsivity, compulsivity, and top-down cognitive control. Neuron. 2011;69:680–694. doi: 10.1016/j.neuron.2011.01.020. [DOI] [PubMed] [Google Scholar]
- Dalley J.W., Fryer T.D., Brichard L., Robinson E.S., Theobald D.E., et al. Nucleus accumbens D2/3 receptors predict trait impulsivity and cocaine reinforcement. Science. 2007;315:1267–1270. doi: 10.1126/science.1137073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dalley J.W., Robbins T.W. Fractionating impulsivity: neuropsychiatric implications. Nat. Rev. Neurosci. 2017;18:158–171. doi: 10.1038/nrn.2017.8. [DOI] [PubMed] [Google Scholar]
- Dalley J.W., Theobald D.E., Eagle D.M., Passetti F., Robbins T.W. Deficits in impulse control associated with tonically-elevated serotonergic function in rat prefrontal cortex. Neuropsychopharmacology. 2002;26:716–728. doi: 10.1016/S0893-133X(01)00412-2. [DOI] [PubMed] [Google Scholar]
- Dalton G.L., Ma L.M., Phillips A.G., Floresco S.B. Blockade of NMDA GluN2B receptors selectively impairs behavioral flexibility but not initial discrimination learning. Psychopharmacology. 2011;216:525–535. doi: 10.1007/s00213-011-2246-z. [DOI] [PubMed] [Google Scholar]
- Dambacher F., Sack A.T., Lobbestael J., Arntz A., Brugman S., Schuhmann T. Out of control: evidence for anterior insula involvement in motor impulsivity and reactive aggression. Soc Cogn Affect Neurosci. 2015;10:508–516. doi: 10.1093/scan/nsu077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Danet M., Lapiz-Bluhm S., Morilak D.A. A cognitive deficit induced in rats by chronic intermittent cold stress is reversed by chronic antidepressant treatment. Int. J. Neuropsychopharmacol. 2010;13:997–1009. doi: 10.1017/S1461145710000039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis-Reyes B.D., Smith A.E., Xu J., Cunningham K.A., Zhou J., Anastasio N.C. Subanesthetic ketamine with an AMPAkine attenuates motor impulsivity in rats. Behav. Pharmacol. 2021;32:335–344. doi: 10.1097/FBP.0000000000000623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Alcubierre D., Ferrari D., Mauro G., Isidori A.M., Tomlinson J.W., Pofi R. Glucocorticoids and cognitive function: a walkthrough in endogenous and exogenous alterations. J. Endocrinol. Invest. 2023;46:1961–1982. doi: 10.1007/s40618-023-02091-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Kloet E.R. Functional profile of the binary brain corticosteroid receptor system: mediating, multitasking, coordinating, integrating. Eur. J. Pharmacol. 2013;719:53–62. doi: 10.1016/j.ejphar.2013.04.053. [DOI] [PubMed] [Google Scholar]
- de Kloet E.R., Joels M. The cortisol switch between vulnerability and resilience. Mol Psychiatry. 2023 doi: 10.1038/s41380-022-01934-8. [DOI] [PubMed] [Google Scholar]
- Delahaye M., Lemoine P., Cartwright S., Deuring G., Beck J., et al. Learning aptitude, spatial orientation and cognitive flexibility tested in a virtual labyrinth after virtual stress induction. BMC Psychol. 2015;3:22. doi: 10.1186/s40359-015-0080-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desrochers S.S., Spring M.G., Nautiyal K.M. A role for serotonin in modulating opposing drive and brake circuits of impulsivity. Front. Behav. Neurosci. 2022;16 doi: 10.3389/fnbeh.2022.791749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deuter C.E., Wingenfeld K., Schultebraucks K., Otte C., Kuehl L.K. Influence of glucocorticoid and mineralocorticoid receptor stimulation on task switching. Horm. Behav. 2019;109:18–24. doi: 10.1016/j.yhbeh.2019.01.007. [DOI] [PubMed] [Google Scholar]
- Di S., Malcher-Lopes R., Halmos K.C., Tasker J.G. Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback mechanism. J. Neurosci. 2003;23:4850–4857. doi: 10.1523/JNEUROSCI.23-12-04850.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diamond A. Executive functions. Annu. Rev. Psychol. 2013;64:135–168. doi: 10.1146/annurev-psych-113011-143750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dias R., Robbins T.W., Roberts A.C. Primate analogue of the Wisconsin Card Sorting Test: effects of excitotoxic lesions of the prefrontal cortex of the marmoset. Behav. Neurosci. 1996;110:872–886. doi: 10.1037//0735-7044.110.5.872. [DOI] [PubMed] [Google Scholar]
- Dias-Ferreira E., Sousa J.C., Melo I., Morgado P., Mesquita A.R., et al. Chronic stress causes frontostriatal reorganization and affects decision-making. Science. 2009;325:621–625. doi: 10.1126/science.1171203. [DOI] [PubMed] [Google Scholar]
- Disner S.G., Beevers C.G., Haigh E.A.P., Beck A.T. Neural mechanisms of the cognitive model of depression. Nat. Rev. Neurosci. 2011;12:467–477. doi: 10.1038/nrn3027. [DOI] [PubMed] [Google Scholar]
- Dockery C.A., Hueckel-Weng R., Birbaumer N., Plewnia C. Enhancement of planning ability by transcranial direct current stimulation. J. Neurosci. 2009;29:7271–7277. doi: 10.1523/JNEUROSCI.0065-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donegan J.J., Girotti M., Weinberg M.S., Morilak D.A. A novel role for brain interleukin-6: facilitation of cognitive flexibility in rat orbitofrontal cortex. J. Neurosci. 2014;34:953–962. doi: 10.1523/JNEUROSCI.3968-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong Z., Bai Y., Wu X., Li H., Gong B., et al. Hippocampal long-term depression mediates spatial reversal learning in the Morris water maze. Neuropharmacology. 2013;64:65–73. doi: 10.1016/j.neuropharm.2012.06.027. [DOI] [PubMed] [Google Scholar]
- Dossi G., Delvecchio G., Prunas C., Soares J.C., Brambilla P. Neural bases of cognitive impairments in post-traumatic stress disorders: a mini-review of functional magnetic resonance imaging findings. Front Psychiatry. 2020;11:176. doi: 10.3389/fpsyt.2020.00176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dougherty D.M., Mullen J., Hill-Kapturczak N., Liang Y., Karns T.E., et al. Effects of tryptophan depletion and a simulated alcohol binge on impulsivity. Exp. Clin. Psychopharmacol. 2015;23:109–121. doi: 10.1037/a0038943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drevets W.C. Orbitofrontal cortex function and structure in depression. Ann. N. Y. Acad. Sci. 2007;1121:499–527. doi: 10.1196/annals.1401.029. [DOI] [PubMed] [Google Scholar]
- Drysdale A.T., Grosenick L., Downar J., Dunlop K., Mansouri F., et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017;23:28–38. doi: 10.1038/nm.4246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duffy S., Labrie V., Roder J.C. D-serine augments NMDA-NR2B receptor-dependent hippocampal long-term depression and spatial reversal learning. Neuropsychopharmacology. 2008;33:1004–1018. doi: 10.1038/sj.npp.1301486. [DOI] [PubMed] [Google Scholar]
- Eagle D.M., Baunez C. Is there an inhibitory-response-control system in the rat? Evidence from anatomical and pharmacological studies of behavioral inhibition. Neurosci. Biobehav. Rev. 2010;34:50–72. doi: 10.1016/j.neubiorev.2009.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eagle D.M., Baunez C., Hutcheson D.M., Lehmann O., Shah A.P., Robbins T.W. Stop-signal reaction-time task performance: role of prefrontal cortex and subthalamic nucleus. Cereb Cortex. 2008;18:178–188. doi: 10.1093/cercor/bhm044. [DOI] [PubMed] [Google Scholar]
- Ennaceur A., Aggleton J.P. The effects of neurotoxic lesions of the perirhinal cortex combined to fornix transection on object recognition memory in the rat. Behav. Brain Res. 1997;88:181–193. doi: 10.1016/s0166-4328(97)02297-3. [DOI] [PubMed] [Google Scholar]
- Etherington L.A., Mihalik B., Palvolgyi A., Ling I., Pallagi K., et al. Selective inhibition of extra-synaptic alpha5-GABA(A) receptors by S44819, a new therapeutic agent. Neuropharmacology. 2017;125:353–364. doi: 10.1016/j.neuropharm.2017.08.012. [DOI] [PubMed] [Google Scholar]
- Ettman C.K., Abdalla S.M., Cohen G.H., Sampson L., Vivier P.M., Galea S. Prevalence of depression symptoms in US adults before and during the COVID-19 pandemic. JAMA Netw. Open. 2020;3 doi: 10.1001/jamanetworkopen.2020.19686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evenden J. The pharmacology of impulsive behaviour in rats V: the effects of drugs on responding under a discrimination task using unreliable visual stimuli. Psychopharmacology (Berl) 1999;143:111–122. doi: 10.1007/s002130050926. [DOI] [PubMed] [Google Scholar]
- Evenden J.L. Varieties of impulsivity. Psychopharmacology (Berl) 1999;146:348–361. doi: 10.1007/pl00005481. [DOI] [PubMed] [Google Scholar]
- Evers A.G., Murrough J.W., Charney D.S., Costi S. Ketamine as a prophylactic resilience-enhancing agent. Front Psychiatry. 2022;13 doi: 10.3389/fpsyt.2022.833259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fecteau S., Pascual-Leone A., Zald D.H., Liguori P., Theoret H., et al. Activation of prefrontal cortex by transcranial direct current stimulation reduces appetite for risk during ambiguous decision making. J. Neurosci. 2007;27:6212–6218. doi: 10.1523/JNEUROSCI.0314-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fineberg N.A., Cinosi E., Smith M.V.A., Busby A.D., Wellsted D., et al. Feasibility, acceptability and practicality of transcranial stimulation in obsessive compulsive symptoms (FEATSOCS): a randomised controlled crossover trial. Compr Psychiatry. 2023;122 doi: 10.1016/j.comppsych.2023.152371. [DOI] [PubMed] [Google Scholar]
- Fletcher P.J., Tampakeras M., Sinyard J., Higgins G.A. Opposing effects of 5-HT(2A) and 5-HT(2C) receptor antagonists in the rat and mouse on premature responding in the five-choice serial reaction time test. Psychopharmacology (Berl) 2007;195:223–234. doi: 10.1007/s00213-007-0891-z. [DOI] [PubMed] [Google Scholar]
- Floresco S.B. Prefrontal dopamine and behavioral flexibility: shifting from an "inverted-U" toward a family of functions. Front. Neurosci. 2013;7:62. doi: 10.3389/fnins.2013.00062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Floresco S.B., Block A.E., Tse M.T. Inactivation of the medial prefrontal cortex of the rat impairs strategy set-shifting, but not reversal learning, using a novel, automated procedure. Behav. Brain Res. 2008;190:85–96. doi: 10.1016/j.bbr.2008.02.008. [DOI] [PubMed] [Google Scholar]
- Fonseca M.S., Murakami M., Mainen Z.F. Activation of dorsal raphe serotonergic neurons promotes waiting but is not reinforcing. Curr. Biol. 2015;25:306–315. doi: 10.1016/j.cub.2014.12.002. [DOI] [PubMed] [Google Scholar]
- Franco C.Y., Knowlton B.J. Effects of early-life stress on probabilistic reversal learning and response perseverance in young adults. Neurobiol. Learn. Mem. 2023;205 doi: 10.1016/j.nlm.2023.107839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fregni F., El-Hagrassy M.M., Pacheco-Barrios K., Carvalho S., Leite J., et al. Evidence-based guidelines and secondary meta-analysis for the use of transcranial direct current stimulation in neurological and psychiatric disorders. Int. J. Neuropsychopharmacol. 2021;24:256–313. doi: 10.1093/ijnp/pyaa051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fucich E.A., Paredes D., Morilak D.A. Therapeutic effects of extinction learning as a model of exposure therapy in rats. Neuropsychopharmacology. 2016;41:3092–3102. doi: 10.1038/npp.2016.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fucich E.A., Paredes D., Saunders M., Morilak D.A. Activity in the ventral medial prefrontal cortex is necessary for the therapeutic effects of extinction in rats. J. Neurosci. 2018 doi: 10.1523/JNEUROSCI.0635-17.2017. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuge P., Aust S., Fan Y., Weigand A., Gartner M., et al. Interaction of early life stress and corticotropin-releasing hormone receptor gene: effects on working memory. Biol Psychiatry. 2014;76:888–894. doi: 10.1016/j.biopsych.2014.04.016. [DOI] [PubMed] [Google Scholar]
- Furr A., Lapiz-Bluhm D.S., Morilak D.A. 5-HT2A-receptors in the orbitofrontal cortex facilitate reversal learning and contribute to the beneficial cognitive effects of chronic citalopram treatment in rats. Int. J. Neuropsychopharmacol. 2012;15:1295–1305. doi: 10.1017/S1461145711001441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuster J.M., Alexander G.E. Neuron activity related to short-term memory. Science. 1971;173:652–654. doi: 10.1126/science.173.3997.652. [DOI] [PubMed] [Google Scholar]
- Fuster J.M., Alexander G.E. Firing changes in cells of the nucleus medialis dorsalis associated with delayed response behavior. Brain Res. 1973;61:79–91. doi: 10.1016/0006-8993(73)90517-9. [DOI] [PubMed] [Google Scholar]
- Gallagher P., Massey A.E., Young A.H., McAllister-Williams R.H. Effects of acute tryptophan depletion on executive function in healthy male volunteers. BMC Psychiatr. 2003;3:10. doi: 10.1186/1471-244X-3-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garrett J.E., Wellman C.L. Chronic stress effects on dendritic morphology in medial prefrontal cortex: sex differences and estrogen dependence. Neuroscience. 2009;162:195–207. doi: 10.1016/j.neuroscience.2009.04.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geissler C.F., Friehs M.A., Frings C., Domes G. Time-dependent effects of acute stress on working memory performance: a systematic review and hypothesis. Psychoneuroendocrinology. 2023;148 doi: 10.1016/j.psyneuen.2022.105998. [DOI] [PubMed] [Google Scholar]
- George S.A., Rodriguez-Santiago M., Riley J., Abelson J.L., Floresco S.B., Liberzon I. Alterations in cognitive flexibility in a rat model of post-traumatic stress disorder. Behav. Brain Res. 2015;286:256–264. doi: 10.1016/j.bbr.2015.02.051. [DOI] [PubMed] [Google Scholar]
- Ghahremani D.G., Monterosso J., Jentsch J.D., Bilder R.M., Poldrack R.A. Neural components underlying behavioral flexibility in human reversal learning. Cereb Cortex. 2010;20:1843–1852. doi: 10.1093/cercor/bhp247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gill H., Gill B., Rodrigues N.B., Lipsitz O., Rosenblat J.D., et al. The effects of ketamine on cognition in treatment-resistant depression: a systematic review and priority avenues for future research. Neurosci. Biobehav. Rev. 2021;120:78–85. doi: 10.1016/j.neubiorev.2020.11.020. [DOI] [PubMed] [Google Scholar]
- Girotti M., Carreno F.R., Morilak D.A. Role of orbitofrontal cortex and differential effects of acute and chronic stress on motor impulsivity measured with 1-choice serial reaction time test in male rats. Int. J. Neuropsychopharmacol. 2022;25:1026–1036. doi: 10.1093/ijnp/pyac062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Girotti M., Donegan J.J., Morilak D.A. Chronic intermittent cold stress sensitizes neuro-immune reactivity in the rat brain. Psychoneuroendocrinology. 2011;36:1164–1174. doi: 10.1016/j.psyneuen.2011.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Girotti M., Pace T.W., Gaylord R.I., Rubin B.A., Herman J.P., Spencer R.L. Habituation to repeated restraint stress is associated with lack of stress-induced c-fos expression in primary sensory processing areas of the rat brain. Neuroscience. 2006;138:1067–1081. doi: 10.1016/j.neuroscience.2005.12.002. [DOI] [PubMed] [Google Scholar]
- Girotti M., Silva J.D., George C.M., Morilak D.A. Ciliary neurotrophic factor signaling in the rat orbitofrontal cortex ameliorates stress-induced deficits in reversal learning. Neuropharmacology. 2019;160 doi: 10.1016/j.neuropharm.2019.107791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Godoy L.D., Rossignoli M.T., Delfino-Pereira P., Garcia-Cairasco N., de Lima Umeoka E.H. A comprehensive overview on stress neurobiology: basic concepts and clinical implications. Front. Behav. Neurosci. 2018;12:127. doi: 10.3389/fnbeh.2018.00127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez-Burgos I., Fletes-Vargas G., Gonzalez-Tapia D., Gonzalez-Ramirez M.M., Rivera-Cervantes M.C., Martinez-Degollado M. Prefrontal serotonin depletion impairs egocentric, but not allocentric working memory in rats. Neurosci. Res. 2012;73:321–327. doi: 10.1016/j.neures.2012.05.003. [DOI] [PubMed] [Google Scholar]
- Goodwill H.L., Manzano-Nieves G., LaChance P., Teramoto S., Lin S., et al. Early life stress drives sex-selective impairment in reversal learning by affecting parvalbumin interneurons in orbitofrontal cortex of mice. Cell Rep. 2018;25:2299–22307 e4. doi: 10.1016/j.celrep.2018.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goss A.J., Kaser M., Costafreda S.G., Sahakian B.J., Fu C.H. Modafinil augmentation therapy in unipolar and bipolar depression: a systematic review and meta-analysis of randomized controlled trials. J. Clin. Psychiatry. 2013;74:1101–1107. doi: 10.4088/JCP.13r08560. [DOI] [PubMed] [Google Scholar]
- Granon S., Poucet B., Thinus-Blanc C., Changeux J.P., Vidal C. Nicotinic and muscarinic receptors in the rat prefrontal cortex: differential roles in working memory, response selection and effortful processing. Psychopharmacology (Berl) 1995;119:139–144. doi: 10.1007/BF02246154. [DOI] [PubMed] [Google Scholar]
- Gray J.D., Kogan J.F., Marrocco J., McEwen B.S. Genomic and epigenomic mechanisms of glucocorticoids in the brain. Nat. Rev. Endocrinol. 2017;13:661–673. doi: 10.1038/nrendo.2017.97. [DOI] [PubMed] [Google Scholar]
- Graybeal C., Feyder M., Schulman E., Saksida L.M., Bussey T.J., et al. Paradoxical reversal learning enhancement by stress or prefrontal cortical damage: rescue with BDNF. Nat. Neurosci. 2011;14:1507–1509. doi: 10.1038/nn.2954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groman S.M., Keistler C., Keip A.J., Hammarlund E., DiLeone R.J., et al. Orbitofrontal circuits control multiple reinforcement-learning processes. Neuron. 2019;103:734–746 e3. doi: 10.1016/j.neuron.2019.05.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gutnikov S.A., Rawlins J.N. Systemic NMDA antagonist CGP-37849 produces non-specific impairment in a working memory task: the effect does not resemble those of AP5 and of lesions of the hippocampus or fornix. Neuropsychologia. 1996;34:311–314. doi: 10.1016/0028-3932(95)00113-1. [DOI] [PubMed] [Google Scholar]
- Guzulaitis R., Godenzini L., Palmer L.M. Neural basis of anticipation and premature impulsive action in the frontal cortex. Nat. Neurosci. 2022;25:1683–1692. doi: 10.1038/s41593-022-01198-z. [DOI] [PubMed] [Google Scholar]
- Hack L.M., Tozzi L., Zenteno S., Olmsted A.M., Hilton R., et al. A cognitive biotype of depression and symptoms, behavior measures, neural circuits, and differential treatment outcomes: a prespecified secondary analysis of a randomized clinical trial. JAMA Netw. Open. 2023;6 doi: 10.1001/jamanetworkopen.2023.18411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hains A.B., Yabe Y., Arnsten A.F. Chronic stimulation of alpha-2A-adrenoceptors with guanfacine protects rodent prefrontal cortex dendritic spines and cognition from the effects of chronic stress. Neurobiol Stress. 2015;2:1–9. doi: 10.1016/j.ynstr.2015.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hajek T., Kopecek M., Preiss M., Alda M., Hoschl C. Prospective study of hippocampal volume and function in human subjects treated with corticosteroids. Eur Psychiatry. 2006;21:123–128. doi: 10.1016/j.eurpsy.2005.01.005. [DOI] [PubMed] [Google Scholar]
- Hallock H.L., Wang A., Griffin A.L. Ventral midline thalamus is critical for hippocampal-prefrontal synchrony and spatial working memory. J. Neurosci. 2016;36:8372–8389. doi: 10.1523/JNEUROSCI.0991-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haluk D.M., Floresco S.B. Ventral striatal dopamine modulation of different forms of behavioral flexibility. Neuropsychopharmacology. 2009;34:2041–2052. doi: 10.1038/npp.2009.21. [DOI] [PubMed] [Google Scholar]
- Hampshire A., Chaudhry A.M., Owen A.M., Roberts A.C. Dissociable roles for lateral orbitofrontal cortex and lateral prefrontal cortex during preference driven reversal learning. Neuroimage. 2012;59:4102–4112. doi: 10.1016/j.neuroimage.2011.10.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hannah R., Aron A.R. Towards real-world generalizability of a circuit for action-stopping. Nat. Rev. Neurosci. 2021;22:538–552. doi: 10.1038/s41583-021-00485-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harborne G.C., Watson F.L., Healy D.T., Groves L. The effects of sub-anaesthetic doses of ketamine on memory, cognitive performance and subjective experience in healthy volunteers. J. Psychopharmacol. 1996;10:134–140. doi: 10.1177/026988119601000208. [DOI] [PubMed] [Google Scholar]
- Harms M.B., Shannon Bowen K.E., Hanson J.L., Pollak S.D. Instrumental learning and cognitive flexibility processes are impaired in children exposed to early life stress. Dev. Sci. 2018;21 doi: 10.1111/desc.12596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harris A.P., Holmes M.C., de Kloet E.R., Chapman K.E., Seckl J.R. Mineralocorticoid and glucocorticoid receptor balance in control of HPA axis and behaviour. Psychoneuroendocrinology. 2013;38:648–658. doi: 10.1016/j.psyneuen.2012.08.007. [DOI] [PubMed] [Google Scholar]
- Harrison A.A., Everitt B.J., Robbins T.W. Central 5-HT depletion enhances impulsive responding without affecting the accuracy of attentional performance: interactions with dopaminergic mechanisms. Psychopharmacology (Berl) 1997;133:329–342. doi: 10.1007/s002130050410. [DOI] [PubMed] [Google Scholar]
- Hassamal S. Chronic stress, neuroinflammation, and depression: an overview of pathophysiological mechanisms and emerging anti-inflammatories. Front Psychiatry. 2023;14 doi: 10.3389/fpsyt.2023.1130989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hatherall L., Sanchez C., Morilak D.A. Chronic vortioxetine treatment reduces exaggerated expression of conditioned fear memory and restores active coping behavior in chronically stressed rats. Int. J. Neuropsychopharmacol. 2017;20:316–323. doi: 10.1093/ijnp/pyw105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hedlund P.B. The 5-HT7 receptor and disorders of the nervous system: an overview. Psychopharmacology (Berl) 2009;206:345–354. doi: 10.1007/s00213-009-1626-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hedlund P.B., Huitron-Resendiz S., Henriksen S.J., Sutcliffe J.G. 5-HT7 receptor inhibition and inactivation induce antidepressantlike behavior and sleep pattern. Biol Psychiatry. 2005;58:831–837. doi: 10.1016/j.biopsych.2005.05.012. [DOI] [PubMed] [Google Scholar]
- Henckens M.J., Deussing J.M., Chen A. Region-specific roles of the corticotropin-releasing factor-urocortin system in stress. Nat. Rev. Neurosci. 2016;17:636–651. doi: 10.1038/nrn.2016.94. [DOI] [PubMed] [Google Scholar]
- Hendrawan D., Yamakawa K., Kimura M., Murakami H., Ohira H. Executive functioning performance predicts subjective and physiological acute stress reactivity: preliminary results. Int. J. Psychophysiol. 2012;84:277–283. doi: 10.1016/j.ijpsycho.2012.03.006. [DOI] [PubMed] [Google Scholar]
- Herman J.P., McKlveen J.M., Ghosal S., Kopp B., Wulsin A., et al. Regulation of the hypothalamic-pituitary-adrenocortical stress response. Compr. Physiol. 2016;6:603–621. doi: 10.1002/cphy.c150015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hervig M.E., Piilgaard L., Bozic T., Alsio J., Robbins T.W. Glutamatergic and serotonergic modulation of rat medial and lateral orbitofrontal cortex in visual serial reversal learning. Psychol Neurosci. 2020;13:438–458. doi: 10.1037/pne0000221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heston J., Friedman A., Baqai M., Bavafa N., Aron A.R., Hnasko T.S. Activation of subthalamic nucleus stop circuit disrupts cognitive performance. eNeuro. 2020;7 doi: 10.1523/ENEURO.0159-20.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hetem L.A., Danion J.M., Diemunsch P., Brandt C. Effect of a subanesthetic dose of ketamine on memory and conscious awareness in healthy volunteers. Psychopharmacology (Berl) 2000;152:283–288. doi: 10.1007/s002130000511. [DOI] [PubMed] [Google Scholar]
- Higgins G.A., Silenieks L.B., MacMillan C., Sevo J., Zeeb F.D., Thevarkunnel S. Enhanced attention and impulsive action following NMDA receptor GluN2B-selective antagonist pretreatment. Behav. Brain Res. 2016;311:1–14. doi: 10.1016/j.bbr.2016.05.025. [DOI] [PubMed] [Google Scholar]
- Hill M.N., Patel S., Carrier E.J., Rademacher D.J., Ormerod B.K., et al. Downregulation of endocannabinoid signaling in the hippocampus following chronic unpredictable stress. Neuropsychopharmacology. 2005;30:508–515. doi: 10.1038/sj.npp.1300601. [DOI] [PubMed] [Google Scholar]
- Hillier A., Alexander J.K., Beversdorf D.Q. The effect of auditory stressors on cognitive flexibility. Neurocase. 2006;12:228–231. doi: 10.1080/13554790600878887. [DOI] [PubMed] [Google Scholar]
- Hipp J.F., Knoflach F., Comley R., Ballard T.M., Honer M., et al. Basmisanil, a highly selective GABA(A)-alpha5 negative allosteric modulator: preclinical pharmacology and demonstration of functional target engagement in man. Sci. Rep. 2021;11:7700. doi: 10.1038/s41598-021-87307-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hughes J.H., Gallagher P., Stewart M.E., Matthews D., Kelly T.P., Young A.H. The effects of acute tryptophan depletion on neuropsychological function. J. Psychopharmacol. 2003;17:300–309. doi: 10.1177/02698811030173012. [DOI] [PubMed] [Google Scholar]
- Hyman S.L., Shores A., North K.N. The nature and frequency of cognitive deficits in children with neurofibromatosis type 1. Neurology. 2005;65:1037–1044. doi: 10.1212/01.wnl.0000179303.72345.ce. [DOI] [PubMed] [Google Scholar]
- Idris N., Neill J., Grayson B., Bang-Andersen B., Witten L.M., et al. Sertindole improves sub-chronic PCP-induced reversal learning and episodic memory deficits in rodents: involvement of 5-HT(6) and 5-HT (2A) receptor mechanisms. Psychopharmacology (Berl) 2010;208:23–36. doi: 10.1007/s00213-009-1702-5. [DOI] [PubMed] [Google Scholar]
- Ihalainen J.A., Tanila H. In vivo regulation of dopamine and noradrenaline release by alpha2A-adrenoceptors in the mouse prefrontal cortex. Eur. J. Neurosci. 2002;15:1789–1794. doi: 10.1046/j.1460-9568.2002.02014.x. [DOI] [PubMed] [Google Scholar]
- Isherwood S.N., Pekcec A., Nicholson J.R., Robbins T.W., Dalley J.W. Dissociable effects of mGluR5 allosteric modulation on distinct forms of impulsivity in rats: interaction with NMDA receptor antagonism. Psychopharmacology (Berl) 2015;232:3327–3344. doi: 10.1007/s00213-015-3984-0. [DOI] [PubMed] [Google Scholar]
- Isherwood S.N., Robbins T.W., Nicholson J.R., Dalley J.W., Pekcec A. Selective and interactive effects of D(2) receptor antagonism and positive allosteric mGluR4 modulation on waiting impulsivity. Neuropharmacology. 2017;123:249–260. doi: 10.1016/j.neuropharm.2017.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ito H.T., Zhang S.J., Witter M.P., Moser E.I., Moser M.B. A prefrontal-thalamo-hippocampal circuit for goal-directed spatial navigation. Nature. 2015;522:50–55. doi: 10.1038/nature14396. [DOI] [PubMed] [Google Scholar]
- Izquierdo A., Darling C., Manos N., Pozos H., Kim C., et al. Basolateral amygdala lesions facilitate reward choices after negative feedback in rats. J. Neurosci. 2013;33:4105–4109. doi: 10.1523/JNEUROSCI.4942-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Izquierdo A., Wiedholz L.M., Millstein R.A., Yang R.J., Bussey T.J., et al. Genetic and dopaminergic modulation of reversal learning in a touchscreen-based operant procedure for mice. Behav. Brain Res. 2006;171:181–188. doi: 10.1016/j.bbr.2006.03.029. [DOI] [PubMed] [Google Scholar]
- Jakala P., Riekkinen M., Sirvio J., Koivisto E., Kejonen K., et al. Guanfacine, but not clonidine, improves planning and working memory performance in humans. Neuropsychopharmacology. 1999;20:460–470. doi: 10.1016/S0893-133X(98)00127-4. [DOI] [PubMed] [Google Scholar]
- Janhunen S.K., Svard H., Talpos J., Kumar G., Steckler T., et al. The subchronic phencyclidine rat model: relevance for the assessment of novel therapeutics for cognitive impairment associated with schizophrenia. Psychopharmacology (Berl) 2015;232:4059–4083. doi: 10.1007/s00213-015-3954-6. [DOI] [PubMed] [Google Scholar]
- Jett J.D., Boley A.M., Girotti M., Shah A., Lodge D.J., Morilak D.A. Antidepressant-like cognitive and behavioral effects of acute ketamine administration associated with plasticity in the ventral hippocampus to medial prefrontal cortex pathway. Psychopharmacology. 2015;232:3123–3133. doi: 10.1007/s00213-015-3957-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jett J.D., Bulin S.E., Hatherall L.C., McCartney C.M., Morilak D.A. Deficits in cognitive flexibility induced by chronic unpredictable stress are associated with impaired glutamate neurotransmission in the rat medial prefrontal cortex. Neuroscience. 2017;346:284–297. doi: 10.1016/j.neuroscience.2017.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jett J.D., Evans L., Girotti M., Lodge D., Morilak D.A. Effects of acute ketamine administration on chronic stress-induced cognitive deficits in rats. Soc. Neurosci. Abstr. 2013;39 Online Program no.: 730.15. [Google Scholar]
- Jett J.D., Morilak D.A. Too much of a good thing: blocking noradrenergic facilitation in medial prefrontal cortex prevents the detrimental effects of chronic stress on cognition. Neuropsychopharmacology. 2013;38:585–595. doi: 10.1038/npp.2012.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joels M. Corticosteroid effects in the brain: U-shape it. Trends Pharmacol. Sci. 2006;27:244–250. doi: 10.1016/j.tips.2006.03.007. [DOI] [PubMed] [Google Scholar]
- Kable J.W., Glimcher P.W. The neural correlates of subjective value during intertemporal choice. Nat. Neurosci. 2007;10:1625–1633. doi: 10.1038/nn2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalia V., Knauft K. Emotion regulation strategies modulate the effect of adverse childhood experiences on perceived chronic stress with implications for cognitive flexibility. PLoS One. 2020;15 doi: 10.1371/journal.pone.0235412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalia V., Knauft K., Hayatbini N. Adverse childhood experiences (ACEs) associated with reduced cognitive flexibility in both college and community samples. PLoS One. 2021;16 doi: 10.1371/journal.pone.0260822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalia V., Vishwanath K., Knauft K., Vellen B.V., Luebbe A., Williams A. Acute stress attenuates cognitive flexibility in males only: an fNIRS examination. Front. Psychol. 2018;9:2084. doi: 10.3389/fpsyg.2018.02084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaser M., Deakin J.B., Michael A., Zapata C., Bansal R., et al. Modafinil improves episodic memory and working memory cognition in patients with remitted depression: a double-blind, randomized, placebo-controlled study. Biol Psychiatry Cogn Neurosci Neuroimaging. 2017;2:115–122. doi: 10.1016/j.bpsc.2016.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katz R.J., Roth K.A., Carroll B.J. Acute and chronic stress effects on open field activity in the rat: implications for a model of depression. Neurosci Biobehav Revs. 1981;5:247–251. doi: 10.1016/0149-7634(81)90005-1. [DOI] [PubMed] [Google Scholar]
- Keller-Wood M.E., Dallman M.F. Corticosteroid inhibition of ACTH secretion. Endocr. Rev. 1984;5:1–24. doi: 10.1210/edrv-5-1-1. [DOI] [PubMed] [Google Scholar]
- Kentrop J., van der Tas L., Loi M., van I.M.H., Bakermans-Kranenburg M.J., et al. Mifepristone treatment during early adolescence fails to restore maternal deprivation-induced deficits in behavioral inhibition of adult male rats. Front. Behav. Neurosci. 2016;10:122. doi: 10.3389/fnbeh.2016.00122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khan Z.U., Muly E.C. Molecular mechanisms of working memory. Behav. Brain Res. 2011;219:329–341. doi: 10.1016/j.bbr.2010.12.039. [DOI] [PubMed] [Google Scholar]
- Kinnavane L., Albasser M.M., Aggleton J.P. Advances in the behavioural testing and network imaging of rodent recognition memory. Behav. Brain Res. 2015;285:67–78. doi: 10.1016/j.bbr.2014.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klanker M., Sandberg T., Joosten R., Willuhn I., Feenstra M., Denys D. Phasic dopamine release induced by positive feedback predicts individual differences in reversal learning. Neurobiol. Learn. Mem. 2015;125:135–145. doi: 10.1016/j.nlm.2015.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klement J., Hubold C., Hallschmid M., Loeck C., Oltmanns K.M., et al. Effects of glucose infusion on neuroendocrine and cognitive parameters in Addison disease. Metabolism. 2009;58:1825–1831. doi: 10.1016/j.metabol.2009.06.015. [DOI] [PubMed] [Google Scholar]
- Klingberg T., O'Sullivan B.T., Roland P.E. Bilateral activation of fronto-parietal networks by incrementing demand in a working memory task. Cereb Cortex. 1997;7:465–471. doi: 10.1093/cercor/7.5.465. [DOI] [PubMed] [Google Scholar]
- Kortmann G.L., Contini V., Bertuzzi G.P., Mota N.R., Rovaris D.L., et al. The role of a mineralocorticoid receptor gene functional polymorphism in the symptom dimensions of persistent ADHD. Eur. Arch. Psychiatr. Clin. Neurosci. 2013;263:181–188. doi: 10.1007/s00406-012-0321-z. [DOI] [PubMed] [Google Scholar]
- Koskinen T., Ruotsalainen S., Sirvio J. The 5-HT(2) receptor activation enhances impulsive responding without increasing motor activity in rats. Pharmacol. Biochem. Behav. 2000;66:729–738. doi: 10.1016/s0091-3057(00)00241-0. [DOI] [PubMed] [Google Scholar]
- Krystal J.H., Neumeister A. Noradrenergic and serotonergic mechanisms in the neurobiology of posttraumatic stress disorder and resilience. Brain Res. 2009;1293:13–23. doi: 10.1016/j.brainres.2009.03.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lachize S., Apostolakis E.M., van der Laan S., Tijssen A.M., Xu J., et al. Steroid receptor coactivator-1 is necessary for regulation of corticotropin-releasing hormone by chronic stress and glucocorticoids. Proc Natl Acad Sci U S A. 2009;106:8038–8042. doi: 10.1073/pnas.0812062106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lalonde R. The neurobiological basis of spontaneous alternation. Neurosci. Biobehav. Rev. 2002;26:91–104. doi: 10.1016/s0149-7634(01)00041-0. [DOI] [PubMed] [Google Scholar]
- Lang L., Xu B., Yuan J., Li S., Lian S., et al. GABA-mediated activated microglia induce neuroinflammation in the hippocampus of mice following cold exposure through the NLRP3 inflammasome and NF-kappaB signaling pathways. Int Immunopharmacol. 2020;89 doi: 10.1016/j.intimp.2020.106908. [DOI] [PubMed] [Google Scholar]
- Lapiz M.D., Morilak D.A. Noradrenergic modulation of cognitive function in rat medial prefrontal cortex as measured by attentional set shifting capability. Neuroscience. 2006;137:1039–1049. doi: 10.1016/j.neuroscience.2005.09.031. [DOI] [PubMed] [Google Scholar]
- Lapiz M.D.S., Bondi C.O., Morilak D.A. Chronic treatment with desipramine improves cognitive performance of rats in an attentional set shifting test. Neuropsychopharmacology. 2007;32:1000–1010. doi: 10.1038/sj.npp.1301235. [DOI] [PubMed] [Google Scholar]
- Lapiz-Bluhm M.D., Soto-Pina A.E., Hensler J.G., Morilak D.A. Chronic intermittent cold stress and serotonin depletion induce deficits of reversal learning in an attentional set-shifting test in rats. Psychopharmacology (Berl) 2009;202:329–341. doi: 10.1007/s00213-008-1224-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee B., Groman S., London E.D., Jentsch J.D. Dopamine D2/D3 receptors play a specific role in the reversal of a learned visual discrimination in monkeys. Neuropsychopharmacology. 2007;32:2125–2134. doi: 10.1038/sj.npp.1301337. [DOI] [PubMed] [Google Scholar]
- Lee Y., Syeda K., Maruschak N.A., Cha D.S., Mansur R.B., et al. A new perspective on the anti-suicide effects with ketamine treatment: a procognitive effect. J. Clin. Psychopharmacol. 2016;36:50–56. doi: 10.1097/JCP.0000000000000441. [DOI] [PubMed] [Google Scholar]
- Lempert K.M., Porcelli A.J., Delgado M.R., Tricomi E. Individual differences in delay discounting under acute stress: the role of trait perceived stress. Front. Psychol. 2012;3:251. doi: 10.3389/fpsyg.2012.00251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leopoldo M., Lacivita E., Berardi F., Perrone R., Hedlund P.B. Serotonin 5-HT7 receptor agents: structure-activity relationships and potential therapeutic applications in central nervous system disorders. Pharmacol. Ther. 2011;129:120–148. doi: 10.1016/j.pharmthera.2010.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levy-Gigi E., Richter-Levin G. The hidden price of repeated traumatic exposure. Stress. 2014;17:343–351. doi: 10.3109/10253890.2014.923397. [DOI] [PubMed] [Google Scholar]
- Li B., Nguyen T.P., Ma C., Dan Y. Inhibition of impulsive action by projection-defined prefrontal pyramidal neurons. Proc Natl Acad Sci U S A. 2020;117:17278–17287. doi: 10.1073/pnas.2000523117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B.M., Mao Z.M., Wang M., Mei Z.T. Alpha-2 adrenergic modulation of prefrontal cortical neuronal activity related to spatial working memory in monkeys. Neuropsychopharmacology. 1999;21:601–610. doi: 10.1016/S0893-133X(99)00070-6. [DOI] [PubMed] [Google Scholar]
- Li N., Liu R.-J., Dwyer J.M., Banasr M., Lee B., et al. Glutamate N-methyl-D-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry. 2011;69:754–761. doi: 10.1016/j.biopsych.2010.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lightman S.L., Birnie M.T., Conway-Campbell B.L. Dynamics of ACTH and cortisol secretion and implications for disease. Endocr. Rev. 2020;41 doi: 10.1210/endrev/bnaa002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim J., Kim E., Noh H.J., Kang S., Phillips B.U., et al. Assessment of mGluR5 KO mice under conditions of low stress using a rodent touchscreen apparatus reveals impaired behavioural flexibility driven by perseverative responses. Mol. Brain. 2019;12:37. doi: 10.1186/s13041-019-0441-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ling I., Mihalik B., Etherington L.A., Kapus G., Palvolgyi A., et al. A novel GABA(A) alpha 5 receptor inhibitor with therapeutic potential. Eur. J. Pharmacol. 2015;764:497–507. doi: 10.1016/j.ejphar.2015.07.005. [DOI] [PubMed] [Google Scholar]
- Linley S.B., Gallo M.M., Vertes R.P. Lesions of the ventral midline thalamus produce deficits in reversal learning and attention on an odor texture set shifting task. Brain Res. 2016;1649:110–122. doi: 10.1016/j.brainres.2016.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liston C., Gan W.B. Glucocorticoids are critical regulators of dendritic spine development and plasticity in vivo. Proc Natl Acad Sci U S A. 2011;108:16074–16079. doi: 10.1073/pnas.1110444108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liston C., McEwen B.S., Casey B.J. Psychosocial stress reversibly disrupts prefrontal processing and attentional control. Proc Natl Acad Sci U S A. 2009;106:912–917. doi: 10.1073/pnas.0807041106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liston C., Miller M.M., Goldwater D.S., Radley J.J., Rocher A.B., et al. Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting. J. Neurosci. 2006;26:7870–7874. doi: 10.1523/JNEUROSCI.1184-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lohse A., Lokkegaard A., Siebner H.R., Meder D. Linking impulsivity to activity levels in pre-supplementary motor area during sequential gambling. J. Neurosci. 2023;43:1414–1421. doi: 10.1523/JNEUROSCI.1287-22.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loos M., Pattij T., Janssen M.C., Counotte D.S., Schoffelmeer A.N., et al. Dopamine receptor D1/D5 gene expression in the medial prefrontal cortex predicts impulsive choice in rats. Cereb Cortex. 2010;20:1064–1070. doi: 10.1093/cercor/bhp167. [DOI] [PubMed] [Google Scholar]
- Luethi M., Meier B., Sandi C. Stress effects on working memory, explicit memory, and implicit memory for neutral and emotional stimuli in healthy men. Front. Behav. Neurosci. 2008;2:5. doi: 10.3389/neuro.08.005.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lukkes J.L., Drozd H.P., Fitz S.D., Molosh A.I., Clapp D.W., Shekhar A. Guanfacine treatment improves ADHD phenotypes of impulsivity and hyperactivity in a neurofibromatosis type 1 mouse model. J. Neurodev. Disord. 2020;12(2) doi: 10.1186/s11689-019-9304-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lupien S.J., Gillin C.J., Hauger R.L. Working memory is more sensitive than declarative memory to the acute effects of corticosteroids: a dose-response study in humans. Behav. Neurosci. 1999;113:420–430. doi: 10.1037//0735-7044.113.3.420. [DOI] [PubMed] [Google Scholar]
- Lupien S.J., McEwen B.S., Gunnar M.R., Heim C. Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nat. Rev. Neurosci. 2009;10:434–445. doi: 10.1038/nrn2639. [DOI] [PubMed] [Google Scholar]
- Luscher B., Maguire J.L., Rudolph U., Sibille E. GABA(A) receptors as targets for treating affective and cognitive symptoms of depression. Trends Pharmacol. Sci. 2023;44:586–600. doi: 10.1016/j.tips.2023.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luzi L., Gandini S., Massarini S., Bellerba F., Terruzzi I., et al. Reduction of impulsivity in patients receiving deep transcranial magnetic stimulation treatment for obesity. Endocrine. 2021;74:559–570. doi: 10.1007/s12020-021-02802-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma S., Morilak D.A. Chronic intermittent cold stress sensitizes the HPA response to a novel acute stress by enhancing noradrenergic influence in the rat paraventricular nucleus. J. Neuroendocrinol. 2005;17:761–769. doi: 10.1111/j.1365-2826.2005.01372.x. [DOI] [PubMed] [Google Scholar]
- Maeng L.Y., Milad M.R. Post-traumatic stress disorder: the relationship between the fear response and chronic stress. Chronic Stress. 2017;1 doi: 10.1177/2470547017713297. Thousand Oaks. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magarinos A.M., McEwen B.S., Flugge G., Fuchs E. Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews. J. Neurosci. 1996;16:3534–3540. doi: 10.1523/JNEUROSCI.16-10-03534.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahoney J.J., 3rd, Thompson-Lake D.G., Cooper K., Verrico C.D., Newton T.F., De La Garza R., 2nd A comparison of impulsivity, depressive symptoms, lifetime stress and sensation seeking in healthy controls versus participants with cocaine or methamphetamine use disorders. J. Psychopharmacol. 2015;29:50–56. doi: 10.1177/0269881114560182. [DOI] [PubMed] [Google Scholar]
- Mala H., Andersen L.G., Christensen R.F., Felbinger A., Hagstrom J., et al. Prefrontal cortex and hippocampus in behavioural flexibility and posttraumatic functional recovery: reversal learning and set-shifting in rats. Brain Res. Bull. 2015;116:34–44. doi: 10.1016/j.brainresbull.2015.05.006. [DOI] [PubMed] [Google Scholar]
- Manes F., Sahakian B., Clark L., Rogers R., Antoun N., et al. Decision-making processes following damage to the prefrontal cortex. Brain. 2002;125:624–639. doi: 10.1093/brain/awf049. [DOI] [PubMed] [Google Scholar]
- Mao Y., Xu Y., Yuan X. Validity of chronic restraint stress for modeling anhedonic-like behavior in rodents: a systematic review and meta-analysis. J. Int. Med. Res. 2022;50 doi: 10.1177/03000605221075816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maras P.M., Baram T.Z. Sculpting the hippocampus from within: stress, spines, and CRH. Trends in Neuroscience. 2012;35:315–324. doi: 10.1016/j.tins.2012.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marino R.A., Gaprielian P., Levy R. Systemic D1-R and D2-R antagonists in non-human primates differentially impact learning and memory while impairing motivation and motor performance. Eur. J. Neurosci. 2022;56:4121–4140. doi: 10.1111/ejn.15743. [DOI] [PubMed] [Google Scholar]
- McAlonan K., Brown V.J. Orbital prefrontal cortex mediates reversal learning and not attentional set shifting in the rat. Behav. Brain Res. 2003;146:97–103. doi: 10.1016/j.bbr.2003.09.019. [DOI] [PubMed] [Google Scholar]
- McBurney-Lin J., Vargova G., Garad M., Zagha E., Yang H. The locus coeruleus mediates behavioral flexibility. Cell Rep. 2022;41 doi: 10.1016/j.celrep.2022.111534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McEwen B.S. Stress, adaptation, and disease. Allostasis and allostatic load. Ann. N. Y. Acad. Sci. 1998;840:33–44. doi: 10.1111/j.1749-6632.1998.tb09546.x. [DOI] [PubMed] [Google Scholar]
- McEwen B.S., Akil H. Revisiting the stress concept: implications for affective disorders. J. Neurosci. 2020;40:12–21. doi: 10.1523/JNEUROSCI.0733-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGaughy J., Ross R.S., Eichenbaum H. Noradrenergic, but not cholinergic, deafferentation of prefrontal cortex impairs attentional set-shifting. Neuroscience. 2008;153:63–71. doi: 10.1016/j.neuroscience.2008.01.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGregor G., Irving A.J., Harvey J. Canonical JAK-STAT signaling is pivotal for long-term depression at adult hippocampal temporoammonic-CA1 synapses. FASEB (Fed. Am. Soc. Exp. Biol.) J. 2017;31:3449–3466. doi: 10.1096/fj.201601293RR. [DOI] [PubMed] [Google Scholar]
- McKay L.I., Cidlowski J.A. Cross-talk between nuclear factor-kappa B and the steroid hormone receptors: mechanisms of mutual antagonism. Mol. Endocrinol. 1998;12:45–56. doi: 10.1210/mend.12.1.0044. [DOI] [PubMed] [Google Scholar]
- McMullin S.D., Shields G.S., Slavich G.M., Buchanan T.W. Cumulative lifetime stress exposure predicts greater impulsivity and addictive behaviors. J. Health Psychol. 2021;26:2921–2936. doi: 10.1177/1359105320937055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McQuail J.A., Beas B.S., Kelly K.B., Hernandez C.M., 3rd, Bizon J.L., Frazier C.J. Attenuated NMDAR signaling on fast-spiking interneurons in prefrontal cortex contributes to age-related decline of cognitive flexibility. Neuropharmacology. 2021;197 doi: 10.1016/j.neuropharm.2021.108720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mehta M.A., Swainson R., Ogilvie A.D., Sahakian J., Robbins T.W. Improved short-term spatial memory but impaired reversal learning following the dopamine D(2) agonist bromocriptine in human volunteers. Psychopharmacology (Berl) 2001;159:10–20. doi: 10.1007/s002130100851. [DOI] [PubMed] [Google Scholar]
- Mereu M., Bonci A., Newman A.H., Tanda G. The neurobiology of modafinil as an enhancer of cognitive performance and a potential treatment for substance use disorders. Psychopharmacology (Berl) 2013;229:415–434. doi: 10.1007/s00213-013-3232-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merriam E.P., Thase M.E., Haas G.L., Keshavan M.S., Sweeney J.A. Prefrontal cortical dysfunction in depression determined by Wisconsin Card Sorting Test performance. Am J Psychiatry. 1999;156:780–782. doi: 10.1176/ajp.156.5.780. [DOI] [PubMed] [Google Scholar]
- Michels L., Martin E., Klaver P., Edden R., Zelaya F., et al. Frontal GABA levels change during working memory. PLoS One. 2012;7 doi: 10.1371/journal.pone.0031933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mika A., Mazur G.J., Hoffman A.N., Talboom J.S., Bimonte-Nelson H.A., et al. Chronic stress impairs prefrontal cortex-dependent response inhibition and spatial working memory. Behav. Neurosci. 2012;126:605–619. doi: 10.1037/a0029642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Millan M.J., Agid Y., Brune M., Bullmore E.T., Carter C.S., et al. Cognitive dysfunction in psychiatric disorders: characteristics, causes and the quest for improved therapy. Nat. Rev. Drug Discov. 2012;11:141–168. doi: 10.1038/nrd3628. [DOI] [PubMed] [Google Scholar]
- Miller E.K., Cohen J.D. An integrative theory of prefrontal cortex function. Annu. Rev. Neurosci. 2001;24:167–202. doi: 10.1146/annurev.neuro.24.1.167. [DOI] [PubMed] [Google Scholar]
- Mills F., Bartlett T.E., Dissing-Olesen L., Wisniewska M.B., Kuznicki J., et al. Cognitive flexibility and long-term depression (LTD) are impaired following beta-catenin stabilization in vivo. Proc Natl Acad Sci U S A. 2014;111:8631–8636. doi: 10.1073/pnas.1404670111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minchew H.M., Radabaugh H.L., LaPorte M.L., Free K.E., Cheng J.P., Bondi C.O. A combined therapeutic regimen of citalopram and environmental enrichment ameliorates attentional set-shifting performance after brain trauma. Eur. J. Pharmacol. 2021;904 doi: 10.1016/j.ejphar.2021.174174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitchell J.M., Ot'alora G.M., van der Kolk B., Shannon S., Bogenschutz M., et al. MDMA-assisted therapy for moderate to severe PTSD: a randomized, placebo-controlled phase 3 trial. Nat Med. 2023;29:2473–2480. doi: 10.1038/s41591-023-02565-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyazaki K., Miyazaki K.W., Sivori G., Yamanaka A., Tanaka K.F., Doya K. Serotonergic projections to the orbitofrontal and medial prefrontal cortices differentially modulate waiting for future rewards. Sci. Adv. 2020;6 doi: 10.1126/sciadv.abc7246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyazaki K.W., Miyazaki K., Doya K. Activation of the central serotonergic system in response to delayed but not omitted rewards. Eur. J. Neurosci. 2011;33:153–160. doi: 10.1111/j.1460-9568.2010.07480.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyazaki K.W., Miyazaki K., Tanaka K.F., Yamanaka A., Takahashi A., et al. Optogenetic activation of dorsal raphe serotonin neurons enhances patience for future rewards. Curr. Biol. 2014;24:2033–2040. doi: 10.1016/j.cub.2014.07.041. [DOI] [PubMed] [Google Scholar]
- Mkrtchian A., Evans J.W., Kraus C., Yuan P., Kadriu B., et al. Ketamine modulates fronto-striatal circuitry in depressed and healthy individuals. Mol Psychiatry. 2021;26:3292–3301. doi: 10.1038/s41380-020-00878-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mobini S., Chiang T.J., Ho M.Y., Bradshaw C.M., Szabadi E. Effects of central 5-hydroxytryptamine depletion on sensitivity to delayed and probabilistic reinforcement. Psychopharmacology (Berl) 2000;152:390–397. doi: 10.1007/s002130000542. [DOI] [PubMed] [Google Scholar]
- Monni A., Scandola M., Helie S., Scalas L.F. Cognitive flexibility assessment with a new Reversal learning task paradigm compared with the Wisconsin card sorting test exploring the moderating effect of gender and stress. Psychol. Res. 2023;87:1439–1453. doi: 10.1007/s00426-022-01763-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore H., Rose H.J., Grace A.A. Chronic cold stress reduces the spontaneous activity of ventral tegmental dopamine neurons. Neuropsychopharmacology. 2001;24:410–419. doi: 10.1016/S0893-133X(00)00188-3. [DOI] [PubMed] [Google Scholar]
- Moreira P.S., Almeida P.R., Leite-Almeida H., Sousa N., Costa P. Impact of chronic stress protocols in learning and memory in rodents: systematic review and meta-analysis. PLoS One. 2016;11 doi: 10.1371/journal.pone.0163245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan C.J., Mofeez A., Brandner B., Bromley L., Curran H.V. Acute effects of ketamine on memory systems and psychotic symptoms in healthy volunteers. Neuropsychopharmacology. 2004;29:208–218. doi: 10.1038/sj.npp.1300342. [DOI] [PubMed] [Google Scholar]
- Morice E., Billard J.M., Denis C., Mathieu F., Betancur C., et al. Parallel loss of hippocampal LTD and cognitive flexibility in a genetic model of hyperdopaminergia. Neuropsychopharmacology. 2007;32:2108–2116. doi: 10.1038/sj.npp.1301354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morilak D.A., Ma S., Fleming T., Ji L.L., Mifflin S.W., Cunningham J.T. Chronic cold stress and chronic intermittent hypoxia sensitize acute stress-induced ACTH secretion and Fos staining in LC and forebrain of rats. Soc. Neurosci. Abstr. 2005;31 Online: Program no. 526.7. [Google Scholar]
- Morris L.S., Kundu P., Baek K., Irvine M.A., Mechelmans D.J., et al. Jumping the gun: mapping neural correlates of waiting impulsivity and relevance across alcohol misuse. Biol Psychiatry. 2016;79:499–507. doi: 10.1016/j.biopsych.2015.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris L.S., Kundu P., Dowell N., Mechelmans D.J., Favre P., et al. Fronto-striatal organization: defining functional and microstructural substrates of behavioural flexibility. Cortex. 2016;74:118–133. doi: 10.1016/j.cortex.2015.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moschak T.M., Carelli R.M. Impulsive rats exhibit blunted dopamine release dynamics during a delay discounting task independent of cocaine history. eNeuro. 2017;4 doi: 10.1523/ENEURO.0119-17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mosley P.E., Paliwal S., Robinson K., Coyne T., Silburn P., et al. The structural connectivity of subthalamic deep brain stimulation correlates with impulsivity in Parkinson's disease. Brain. 2020;143:2235–2254. doi: 10.1093/brain/awaa148. [DOI] [PubMed] [Google Scholar]
- Muir J.L., Everitt B.J., Robbins T.W. The cerebral cortex of the rat and visual attentional function: dissociable effects of mediofrontal, cingulate, anterior dorsolateral, and parietal cortex lesions on a five-choice serial reaction time task. Cereb Cortex. 1996;6:470–481. doi: 10.1093/cercor/6.3.470. [DOI] [PubMed] [Google Scholar]
- Murphy S.E., de Cates A.N., Gillespie A.L., Godlewska B.R., Scaife J.C., et al. Translating the promise of 5HT(4) receptor agonists for the treatment of depression. Psychol. Med. 2021;51:1111–1120. doi: 10.1017/S0033291720000604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Musazzi L., Sala N., Tornese P., Gallivanone F., Belloli S., et al. Acute inescapable stress rapidly increases synaptic energy metabolism in prefrontal cortex and alters working memory performance. Cereb Cortex. 2019;29:4948–4957. doi: 10.1093/cercor/bhz034. [DOI] [PubMed] [Google Scholar]
- Naegeli K.J., O'Connor J.A., Banerjee P., Morilak D.A. Effects of milnacipran on cognitive flexibility following chronic stress in rats. Eur. J. Pharmacol. 2013;703:62–66. doi: 10.1016/j.ejphar.2013.02.006. [DOI] [PubMed] [Google Scholar]
- Navarra R., Graf R., Huang Y., Logue S., Comery T., et al. Effects of atomoxetine and methylphenidate on attention and impulsivity in the 5-choice serial reaction time test. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2008;32:34–41. doi: 10.1016/j.pnpbp.2007.06.017. [DOI] [PubMed] [Google Scholar]
- Newcomer J.W., Selke G., Melson A.K., Hershey T., Craft S., et al. Decreased memory performance in healthy humans induced by stress-level cortisol treatment. Arch Gen Psychiatry. 1999;56:527–533. doi: 10.1001/archpsyc.56.6.527. [DOI] [PubMed] [Google Scholar]
- Newman L.A., Darling J., McGaughy J. Atomoxetine reverses attentional deficits produced by noradrenergic deafferentation of medial prefrontal cortex. Psychopharmacology (Berl) 2008;200:39–50. doi: 10.1007/s00213-008-1097-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicolas C.S., Peineau S., Amici M., Csaba Z., Fafouri A., et al. The JAK/STAT pathway is involved in synaptic plasticity. Neuron. 2012;73:374–390. doi: 10.1016/j.neuron.2011.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nijdam M.J., Vermetten E. Moving forward in treatment of posttraumatic stress disorder: innovations to exposure-based therapy. Eur. J. Psychotraumatol. 2018;9 doi: 10.1080/20008198.2018.1458568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nikiforuk A., Popik P. Long-lasting cognitive deficit induced by stress is alleviated by acute administration of antidepressants. Psychoneuroendocrinology. 2011;36:28–39. doi: 10.1016/j.psyneuen.2010.06.001. [DOI] [PubMed] [Google Scholar]
- Nikiforuk A., Popik P. Amisulpride promotes cognitive flexibility in rats: the role of 5-HT7 receptors. Behav. Brain Res. 2013;248:136–140. doi: 10.1016/j.bbr.2013.04.008. [DOI] [PubMed] [Google Scholar]
- Nikiforuk A., Popik P. Ketamine prevents stress-induced cognitive inflexibility in rats. Psychoneuroendocrinology. 2014;40:119–122. doi: 10.1016/j.psyneuen.2013.11.009. [DOI] [PubMed] [Google Scholar]
- Nilsson S.R., Ripley T.L., Somerville E.M., Clifton P.G. Reduced activity at the 5-HT(2C) receptor enhances reversal learning by decreasing the influence of previously non-rewarded associations. Psychopharmacology (Berl) 2012;224:241–254. doi: 10.1007/s00213-012-2746-5. [DOI] [PubMed] [Google Scholar]
- Nishitomi K., Yano K., Kobayashi M., Jino K., Kano T., et al. Systemic administration of guanfacine improves food-motivated impulsive choice behavior primarily via direct stimulation of postsynaptic alpha(2A)-adrenergic receptors in rats. Behav. Brain Res. 2018;345:21–29. doi: 10.1016/j.bbr.2018.02.022. [DOI] [PubMed] [Google Scholar]
- Nonkes L.J., vande V., II, de Leeuw M.J., Wijlaars L.P., Maes J.H., Homberg J.R. Serotonin transporter knockout rats show improved strategy set-shifting and reduced latent inhibition. Learn. Mem. 2012;19:190–193. doi: 10.1101/lm.025908.112. [DOI] [PubMed] [Google Scholar]
- Nunez N.A., Joseph B., Pahwa M., Kumar R., Resendez M.G., et al. Augmentation strategies for treatment resistant major depression: a systematic review and network meta-analysis. J. Affect. Disord. 2022;302:385–400. doi: 10.1016/j.jad.2021.12.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohno M., Watanabe S. Differential effects of 5-HT3 receptor antagonism on working memory failure due to deficiency of hippocampal cholinergic and glutamatergic transmission in rats. Brain Res. 1997;762:211–215. doi: 10.1016/s0006-8993(97)00448-4. [DOI] [PubMed] [Google Scholar]
- Olton D.S. The radial arm maze as a tool in behavioral pharmacology. Physiol. Behav. 1987;40:793–797. doi: 10.1016/0031-9384(87)90286-1. [DOI] [PubMed] [Google Scholar]
- Onaolapo O.J., Onaolapo A.Y. Subchronic oral bromocriptine methanesulfonate enhances open field novelty-induced behavior and spatial memory in male Swiss albino mice. Neurosci J. 2013;2013 doi: 10.1155/2013/948241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orem D.M., Petrac D.C., Bedwell J.S. Chronic self-perceived stress and set-shifting performance in undergraduate students. Stress. 2008;11:73–78. doi: 10.1080/10253890701535103. [DOI] [PubMed] [Google Scholar]
- Orsini C.A., Truckenbrod L.M., Wheeler A.R. Regulation of sex differences in risk-based decision making by gonadal hormones: insights from rodent models. Behav Processes. 2022;200 doi: 10.1016/j.beproc.2022.104663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otte C., Moritz S., Yassouridis A., Koop M., Madrischewski A.M., et al. Blockade of the mineralocorticoid receptor in healthy men: effects on experimentally induced panic symptoms, stress hormones, and cognition. Neuropsychopharmacology. 2007;32:232–238. doi: 10.1038/sj.npp.1301217. [DOI] [PubMed] [Google Scholar]
- Ouellet J., McGirr A., Van den Eynde F., Jollant F., Lepage M., Berlim M.T. Enhancing decision-making and cognitive impulse control with transcranial direct current stimulation (tDCS) applied over the orbitofrontal cortex (OFC): a randomized and sham-controlled exploratory study. J. Psychiatr. Res. 2015;69:27–34. doi: 10.1016/j.jpsychires.2015.07.018. [DOI] [PubMed] [Google Scholar]
- Ouhaz Z., Perry B.A.L., Nakamura K., Mitchell A.S. Mediodorsal thalamus is critical for updating during extradimensional shifts but not reversals in the attentional set-shifting task. eNeuro. 2022;9 doi: 10.1523/ENEURO.0162-21.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Owen A.M., McMillan K.M., Laird A.R., Bullmore E. N-back working memory paradigm: a meta-analysis of normative functional neuroimaging studies. Hum. Brain Mapp. 2005;25:46–59. doi: 10.1002/hbm.20131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Owen A.M., Roberts A.C., Polkey C.E., Sahakian B.J., Robbins T.W. Extra-dimensional versus intra-dimensional set shifting performance following frontal lobe excisions, temporal lobe excisions or amygdalo-hippocampectomy in man. Neuropsychologia. 1991;29:993–1006. doi: 10.1016/0028-3932(91)90063-e. [DOI] [PubMed] [Google Scholar]
- Paine T.A., Brainard S., Keppler E., Poyle R., Sai-Hardebeck E., et al. Juvenile stress increases cocaine-induced impulsivity in female rats. Behav. Brain Res. 2021;414 doi: 10.1016/j.bbr.2021.113488. [DOI] [PubMed] [Google Scholar]
- Paine T.A., Tomasiewicz H.C., Zhang K., Carlezon W.A., Jr. Sensitivity of the five-choice serial reaction time task to the effects of various psychotropic drugs in Sprague-Dawley rats. Biol Psychiatry. 2007;62:687–693. doi: 10.1016/j.biopsych.2006.11.017. [DOI] [PubMed] [Google Scholar]
- Pajkossy P., Szollosi A., Demeter G., Racsmany M. Tonic noradrenergic activity modulates explorative behavior and attentional set shifting: evidence from pupillometry and gaze pattern analysis. Psychophysiology. 2017;54:1839–1854. doi: 10.1111/psyp.12964. [DOI] [PubMed] [Google Scholar]
- Pardon M.-C., Ma S., Morilak D.A. Chronic cold stress sensitizes brain noradrenergic reactivity and noradrenergic facilitation of the HPA stress response in Wistar Kyoto rats. Brain Res. 2003;971:55–65. doi: 10.1016/s0006-8993(03)02355-2. [DOI] [PubMed] [Google Scholar]
- Paredes D., Knippenberg A.R., Bulin S.E., Keppler L.J., Morilak D.A. Adjunct treatment with ketamine enhances the therapeutic effects of extinction learning after chronic unpredictable stress. Neurobiol Stress. 2022;19 doi: 10.1016/j.ynstr.2022.100468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paredes D., Morilak D.A. A rodent model of exposure therapy: the use of fear extinction as a therapeutic intervention for PTSD. Front. Behav. Neurosci. 2019;13:46. doi: 10.3389/fnbeh.2019.00046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paredes D., Morilak D.A. Ventral hippocampal input to infralimbic cortex is necessary for the therapeutic-like effects of extinction in stressed rats. Int. J. Neuropsychopharmacol. 2023;26:529–536. doi: 10.1093/ijnp/pyad043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paterson N.E., Ricciardi J., Wetzler C., Hanania T. Sub-optimal performance in the 5-choice serial reaction time task in rats was sensitive to methylphenidate, atomoxetine and d-amphetamine, but unaffected by the COMT inhibitor tolcapone. Neurosci. Res. 2011;69:41–50. doi: 10.1016/j.neures.2010.10.001. [DOI] [PubMed] [Google Scholar]
- Patriquin M.A., Mathew S.J. The neurobiological mechanisms of generalized anxiety disorder and chronic stress. Chronic Stress. 2017:1. doi: 10.1177/2470547017703993. Thousand Oaks. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pattij T., Janssen M.C., Vanderschuren L.J., Schoffelmeer A.N., van Gaalen M.M. Involvement of dopamine D1 and D2 receptors in the nucleus accumbens core and shell in inhibitory response control. Psychopharmacology (Berl) 2007;191:587–598. doi: 10.1007/s00213-006-0533-x. [DOI] [PubMed] [Google Scholar]
- Patton M.S., Lodge D.J., Morilak D.A., Girotti M. Ketamine corrects stress-induced cognitive dysfunction through JAK2/STAT3 signaling in the orbitofrontal cortex. Neuropsychopharmacology. 2017;42:1220–1230. doi: 10.1038/npp.2016.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pehrson A.L., Jeyarajah T., Sanchez C. Regional distribution of serotonergic receptors: a systems neuroscience perspective on the downstream effects of the multimodal-acting antidepressant vortioxetine on excitatory and inhibitory neurotransmission. CNS Spectr. 2016;21:162–183. doi: 10.1017/S1092852915000486. [DOI] [PubMed] [Google Scholar]
- Pehrson A.L., Leiser S.C., Gulinello M., Dale E., Li Y., et al. Treatment of cognitive dysfunction in major depressive disorder--a review of the preclinical evidence for efficacy of selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors and the multimodal-acting antidepressant vortioxetine. Eur. J. Pharmacol. 2015;753:19–31. doi: 10.1016/j.ejphar.2014.07.044. [DOI] [PubMed] [Google Scholar]
- Pelegrina S., Lechuga M.T., Garcia-Madruga J.A., Elosua M.R., Macizo P., et al. Normative data on the n-back task for children and young adolescents. Front. Psychol. 2015;6:1544. doi: 10.3389/fpsyg.2015.01544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phelps T.I., Bondi C.O., Ahmed R.H., Olugbade Y.T., Kline A.E. Divergent long-term consequences of chronic treatment with haloperidol, risperidone, and bromocriptine on traumatic brain injury-induced cognitive deficits. J. Neurotrauma. 2015;32:590–597. doi: 10.1089/neu.2014.3711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philbert J., Belzung C., Griebel G. The CRF(1) receptor antagonist SSR125543 prevents stress-induced cognitive deficit associated with hippocampal dysfunction: comparison with paroxetine and D-cycloserine. Psychopharmacology (Berl) 2013;228:97–107. doi: 10.1007/s00213-013-3020-1. [DOI] [PubMed] [Google Scholar]
- Piber D., Schultebraucks K., Mueller S.C., Deuter C.E., Wingenfeld K., Otte C. Mineralocorticoid receptor stimulation effects on spatial memory in healthy young adults: a study using the virtual Morris Water Maze task. Neurobiol. Learn. Mem. 2016;136:139–146. doi: 10.1016/j.nlm.2016.10.006. [DOI] [PubMed] [Google Scholar]
- Placek K., Dippel W.C., Jones S., Brady A.M. Impairments in set-shifting but not reversal learning in the neonatal ventral hippocampal lesion model of schizophrenia: further evidence for medial prefrontal deficits. Behav. Brain Res. 2013;256:405–413. doi: 10.1016/j.bbr.2013.08.034. [DOI] [PubMed] [Google Scholar]
- Porcelli A.J., Cruz D., Wenberg K., Patterson M.D., Biswal B.B., Rypma B. The effects of acute stress on human prefrontal working memory systems. Physiol. Behav. 2008;95:282–289. doi: 10.1016/j.physbeh.2008.04.027. [DOI] [PubMed] [Google Scholar]
- Prevot T., Sibille E. Altered GABA-mediated information processing and cognitive dysfunctions in depression and other brain disorders. Mol Psychiatry. 2021;26:151–167. doi: 10.1038/s41380-020-0727-3. [DOI] [PubMed] [Google Scholar]
- Prevot T.D., Li G., Vidojevic A., Misquitta K.A., Fee C., et al. Novel benzodiazepine-like ligands with various anxiolytic, antidepressant, or pro-cognitive profiles. Mol. Neuropsychiatry. 2019;5:84–97. doi: 10.1159/000496086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pripfl J., Neumann R., Kohler U., Lamm C. Effects of transcranial direct current stimulation on risky decision making are mediated by 'hot' and 'cold' decisions, personality, and hemisphere. Eur. J. Neurosci. 2013;38:3778–3785. doi: 10.1111/ejn.12375. [DOI] [PubMed] [Google Scholar]
- Puma C., Baudoin C., Bizot J.C. Effects of intraseptal infusions of N-methyl-D-aspartate receptor ligands on memory in an object recognition task in rats. Neurosci. Lett. 1998;244:97–100. doi: 10.1016/s0304-3940(98)00137-2. [DOI] [PubMed] [Google Scholar]
- Puma C., Bizot J.C. Intraseptal infusions of a low dose of AP5, a NMDA receptor antagonist, improves memory in an object recognition task in rats. Neurosci. Lett. 1998;248:183–186. doi: 10.1016/s0304-3940(98)00358-9. [DOI] [PubMed] [Google Scholar]
- Puumala T., Sirvio J. Changes in activities of dopamine and serotonin systems in the frontal cortex underlie poor choice accuracy and impulsivity of rats in an attention task. Neuroscience. 1998;83:489–499. doi: 10.1016/s0306-4522(97)00392-8. [DOI] [PubMed] [Google Scholar]
- Qiao H., An S.C., Ren W., Ma X.M. Progressive alterations of hippocampal CA3-CA1 synapses in an animal model of depression. Behav. Brain Res. 2014;275:191–200. doi: 10.1016/j.bbr.2014.08.040. [DOI] [PubMed] [Google Scholar]
- Qiao H., Li M.X., Xu C., Chen H.B., An S.C., Ma X.M. Dendritic spines in depression: what we learned from animal models. Neural Plast. 2016;2016 doi: 10.1155/2016/8056370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin S., Hermans E.J., van Marle H.J., Luo J., Fernandez G. Acute psychological stress reduces working memory-related activity in the dorsolateral prefrontal cortex. Biol Psychiatry. 2009;66:25–32. doi: 10.1016/j.biopsych.2009.03.006. [DOI] [PubMed] [Google Scholar]
- Quan M., Zheng C., Zhang N., Han D., Tian Y., et al. Impairments of behavior, information flow between thalamus and cortex, and prefrontal cortical synaptic plasticity in an animal model of depression. Brain Res. Bull. 2011;85:109–116. doi: 10.1016/j.brainresbull.2011.03.002. [DOI] [PubMed] [Google Scholar]
- Radley J.J., Rocher A.B., Miller M., Janssen W.G., Liston C., et al. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cereb Cortex. 2006;16:313–320. doi: 10.1093/cercor/bhi104. [DOI] [PubMed] [Google Scholar]
- Radley J.J., Sisti H.M., Hao J., Rocher A.B., McCall T., et al. Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex. Neuroscience. 2004;125:1–6. doi: 10.1016/j.neuroscience.2004.01.006. [DOI] [PubMed] [Google Scholar]
- Ragozzino M.E. The effects of dopamine D(1) receptor blockade in the prelimbic-infralimbic areas on behavioral flexibility. Learn. Mem. 2002;9:18–28. doi: 10.1101/lm.45802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rainville J.R., Weiss G.L., Evanson N., Herman J.P., Vasudevan N., Tasker J.G. Membrane-initiated nuclear trafficking of the glucocorticoid receptor in hypothalamic neurons. Steroids. 2019;142:55–64. doi: 10.1016/j.steroids.2017.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raio C.M., Hartley C.A., Orederu T.A., Li J., Phelps E.A. Stress attenuates the flexible updating of aversive value. Proc Natl Acad Sci U S A. 2017;114:11241–11246. doi: 10.1073/pnas.1702565114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raio C.M., Konova A.B., Otto A.R. Trait impulsivity and acute stress interact to influence choice and decision speed during multi-stage decision-making. Sci. Rep. 2020;10:7754. doi: 10.1038/s41598-020-64540-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramey T., Regier P.S. Cognitive impairment in substance use disorders. CNS Spectr. 2019;24:102–113. doi: 10.1017/S1092852918001426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rao S.G., Williams G.V., Goldman-Rakic P.S. Destruction and creation of spatial tuning by disinhibition: GABA(A) blockade of prefrontal cortical neurons engaged by working memory. J. Neurosci. 2000;20:485–494. doi: 10.1523/JNEUROSCI.20-01-00485.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Remijnse P.L., Nielen M.M., van Balkom A.J., Cath D.C., van Oppen P., et al. Reduced orbitofrontal-striatal activity on a reversal learning task in obsessive-compulsive disorder. Arch Gen Psychiatry. 2006;63:1225–1236. doi: 10.1001/archpsyc.63.11.1225. [DOI] [PubMed] [Google Scholar]
- Robbins T.W., James M., Owen A.M., Sahakian B.J., Lawrence A.D., et al. A study of performance on tests from the CANTAB battery sensitive to frontal lobe dysfunction in a large sample of normal volunteers: implications for theories of executive functioning and cognitive aging. Cambridge Neuropsychological Test Automated Battery. J. Int. Neuropsychol. Soc. 1998;4:474–490. doi: 10.1017/s1355617798455073. [DOI] [PubMed] [Google Scholar]
- Robbins T.W., Roberts A.C. Differential regulation of fronto-executive function by the monoamines and acetylcholine. Cereb Cortex. 2007;17(Suppl. 1):i151–i160. doi: 10.1093/cercor/bhm066. [DOI] [PubMed] [Google Scholar]
- Robinson E.S., Dalley J.W., Theobald D.E., Glennon J.C., Pezze M.A., et al. Opposing roles for 5-HT2A and 5-HT2C receptors in the nucleus accumbens on inhibitory response control in the 5-choice serial reaction time task. Neuropsychopharmacology. 2008;33:2398–2406. doi: 10.1038/sj.npp.1301636. [DOI] [PubMed] [Google Scholar]
- Robinson E.S., Eagle D.M., Mar A.C., Bari A., Banerjee G., et al. Similar effects of the selective noradrenaline reuptake inhibitor atomoxetine on three distinct forms of impulsivity in the rat. Neuropsychopharmacology. 2008;33:1028–1037. doi: 10.1038/sj.npp.1301487. [DOI] [PubMed] [Google Scholar]
- Rodefer J.S., Nguyen T.N., Karlsson J.J., Arnt J. Reversal of subchronic PCP-induced deficits in attentional set shifting in rats by sertindole and a 5-HT6 receptor antagonist: comparison among antipsychotics. Neuropsychopharmacology. 2008;33:2657–2666. doi: 10.1038/sj.npp.1301654. [DOI] [PubMed] [Google Scholar]
- Rogers R.D., Andrews T.C., Grasby P.M., Brooks D.J., Robbins T.W. Contrasting cortical and subcortical activations produced by attentional set shifting and reversal learning in humans. J Cognitive Neurosci. 2000;12:142–162. doi: 10.1162/089892900561931. [DOI] [PubMed] [Google Scholar]
- Rogers R.D., Andrews T.C., Grasby P.M., Brooks D.J., Robbins T.W. Contrasting cortical and subcortical activations produced by attentional-set shifting and reversal learning in humans. J Cogn Neurosci. 2000;12:142–162. doi: 10.1162/089892900561931. [DOI] [PubMed] [Google Scholar]
- Rogers R.D., Blackshaw A.J., Middleton H.C., Matthews K., Hawtin K., et al. Tryptophan depletion impairs stimulus-reward learning while methylphenidate disrupts attentional control in healthy young adults: implications for the monoaminergic basis of impulsive behaviour. Psychopharmacology (Berl) 1999;146:482–491. doi: 10.1007/pl00005494. [DOI] [PubMed] [Google Scholar]
- Rosenblat J.D., Kakar R., McIntyre R.S. The cognitive effects of antidepressants in major depressive disorder: a systematic review and meta-analysis of randomized clinical trials. Int. J. Neuropsychopharmacol. 2015;19 doi: 10.1093/ijnp/pyv082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saddoris M.P., Sugam J.A., Stuber G.D., Witten I.B., Deisseroth K., Carelli R.M. Mesolimbic dopamine dynamically tracks, and is causally linked to, discrete aspects of value-based decision making. Biol Psychiatry. 2015;77:903–911. doi: 10.1016/j.biopsych.2014.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sakai K., Rowe J.B., Passingham R.E. Active maintenance in prefrontal area 46 creates distractor-resistant memory. Nat. Neurosci. 2002;5:479–484. doi: 10.1038/nn846. [DOI] [PubMed] [Google Scholar]
- Sala-Bayo J., Fiddian L., Nilsson S.R.O., Hervig M.E., McKenzie C., et al. Dorsal and ventral striatal dopamine D1 and D2 receptors differentially modulate distinct phases of serial visual reversal learning. Neuropsychopharmacology. 2020;45:736–744. doi: 10.1038/s41386-020-0612-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salatino A., Miccolis R., Gammeri R., Ninghetto M., Belli F., et al. Improvement of impulsivity and decision making by transcranial direct current stimulation of the dorsolateral prefrontal cortex in a patient with gambling disorder. J. Gambl. Stud. 2022;38:627–634. doi: 10.1007/s10899-021-10050-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez C., Asin K.E., Artigas F. Vortioxetine, a novel antidepressant with multimodal activity: review of preclinical and clinical data. Pharmacol. Ther. 2015;145:43–57. doi: 10.1016/j.pharmthera.2014.07.001. [DOI] [PubMed] [Google Scholar]
- Sanchez C.M., Titus D.J., Wilson N.M., Freund J.E., Atkins C.M. Early life stress exacerbates outcome after traumatic brain injury. J. Neurotrauma. 2021;38:555–565. doi: 10.1089/neu.2020.7267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez E.O., Bangasser D.A. The effects of early life stress on impulsivity. Neurosci. Biobehav. Rev. 2022;137 doi: 10.1016/j.neubiorev.2022.104638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez-Roige S., Stephens D.N., Duka T. Heightened impulsivity: associated with family history of alcohol misuse, and a consequence of alcohol intake. Alcohol Clin. Exp. Res. 2016;40:2208–2217. doi: 10.1111/acer.13184. [DOI] [PubMed] [Google Scholar]
- Sapolsky R.M. Glucocorticoids, the evolution of the stress-response, and the primate predicament. Neurobiol Stress. 2021;14 doi: 10.1016/j.ynstr.2021.100320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sasamori H., Ohmura Y., Yoshida T., Yoshioka M. Noradrenaline reuptake inhibition increases control of impulsive action by activating D(1)-like receptors in the infralimbic cortex. Eur. J. Pharmacol. 2019;844:17–25. doi: 10.1016/j.ejphar.2018.11.041. [DOI] [PubMed] [Google Scholar]
- Sawiak S.J., Jupp B., Taylor T., Caprioli D., Carpenter T.A., Dalley J.W. In vivo gamma-aminobutyric acid measurement in rats with spectral editing at 4.7T. J Magn Reson Imaging. 2016;43:1308–1312. doi: 10.1002/jmri.25093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schippers M.C., Schetters D., De Vries T.J., Pattij T. Differential effects of the pharmacological stressor yohimbine on impulsive decision making and response inhibition. Psychopharmacology (Berl) 2016;233:2775–2785. doi: 10.1007/s00213-016-4337-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoenbaum G., Setlow B., Nugent S.L., Saddoris M.P., Gallagher M. Lesions of orbitofrontal cortex and basolateral amygdala complex disrupt acquisition of odor-guided discriminations and reversals. Learn. Mem. 2003;10:129–140. doi: 10.1101/lm.55203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoofs D., Pabst S., Brand M., Wolf O.T. Working memory is differentially affected by stress in men and women. Behav. Brain Res. 2013;241:144–153. doi: 10.1016/j.bbr.2012.12.004. [DOI] [PubMed] [Google Scholar]
- Schoofs D., Preuss D., Wolf O.T. Psychosocial stress induces working memory impairments in an n-back paradigm. Psychoneuroendocrinology. 2008;33:643–653. doi: 10.1016/j.psyneuen.2008.02.004. [DOI] [PubMed] [Google Scholar]
- Schoofs D., Wolf O.T., Smeets T. Cold pressor stress impairs performance on working memory tasks requiring executive functions in healthy young men. Behav. Neurosci. 2009;123:1066–1075. doi: 10.1037/a0016980. [DOI] [PubMed] [Google Scholar]
- Scoriels L., Jones P.B., Sahakian B.J. Modafinil effects on cognition and emotion in schizophrenia and its neurochemical modulation in the brain. Neuropharmacology. 2013;64:168–184. doi: 10.1016/j.neuropharm.2012.07.011. [DOI] [PubMed] [Google Scholar]
- Sesia T., Temel Y., Lim L.W., Blokland A., Steinbusch H.W., Visser-Vandewalle V. Deep brain stimulation of the nucleus accumbens core and shell: opposite effects on impulsive action. Exp. Neurol. 2008;214:135–139. doi: 10.1016/j.expneurol.2008.07.015. [DOI] [PubMed] [Google Scholar]
- Sessa B., Higbed L., Nutt D. A review of 3,4-methylenedioxymethamphetamine (MDMA)-Assisted psychotherapy. Front Psychiatry. 2019;10:138. doi: 10.3389/fpsyt.2019.00138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shansky R.M., Rubinow K., Brennan A., Arnsten A.F. The effects of sex and hormonal status on restraint-stress-induced working memory impairment. Behav. Brain Funct. 2006;2:8. doi: 10.1186/1744-9081-2-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shields G.S., Trainor B.C., Lam J.C., Yonelinas A.P. Acute stress impairs cognitive flexibility in men, not women. Stress. 2016;19:542–546. doi: 10.1080/10253890.2016.1192603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shilyansky C., Williams L.M., Gyurak A., Harris A., Usherwood T., Etkin A. Effect of antidepressant treatment on cognitive impairments associated with depression: a randomised longitudinal study. Lancet Psychiatr. 2016;3:425–435. doi: 10.1016/S2215-0366(16)00012-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shiroma P.R., Albott C.S., Johns B., Thuras P., Wels J., Lim K.O. Neurocognitive performance and serial intravenous subanesthetic ketamine in treatment-resistant depression. Int. J. Neuropsychopharmacol. 2014;17:1805–1813. doi: 10.1017/S1461145714001011. [DOI] [PubMed] [Google Scholar]
- Shiroma P.R., Velit-Salazar M.R., Vorobyov Y. A systematic review of neurocognitive effects of subanesthetic doses of intravenous ketamine in major depressive disorder, post-traumatic stress disorder, and healthy population. Clin Drug Investig. 2022;42:549–566. doi: 10.1007/s40261-022-01169-z. [DOI] [PubMed] [Google Scholar]
- Short B., Fong J., Galvez V., Shelker W., Loo C.K. Side-effects associated with ketamine use in depression: a systematic review. Lancet Psychiatr. 2018;5:65–78. doi: 10.1016/S2215-0366(17)30272-9. [DOI] [PubMed] [Google Scholar]
- Siddik M.A.B., Fendt M. D-cycloserine rescues scopolamine-induced deficits in cognitive flexibility in rats measured by the attentional set-shifting task. Behav. Brain Res. 2022;431 doi: 10.1016/j.bbr.2022.113961. [DOI] [PubMed] [Google Scholar]
- Siegrist J. Chronic psychosocial stress at work and risk of depression: evidence from prospective studies. Eur. Arch. Psychiatr. Clin. Neurosci. 2008;258(Suppl. 5):115–119. doi: 10.1007/s00406-008-5024-0. [DOI] [PubMed] [Google Scholar]
- Silva-Gomez A.B., Rojas D., Juarez I., Flores G. Decreased dendritic spine density on prefrontal cortical and hippocampal pyramidal neurons in postweaning social isolation rats. Brain Res. 2003;983:128–136. doi: 10.1016/s0006-8993(03)03042-7. [DOI] [PubMed] [Google Scholar]
- Silveira M.M., Wittekindt S.N., Mortazavi L., Hathaway B.A., Winstanley C.A. Investigating serotonergic contributions to cognitive effort allocation, attention, and impulsive action in female rats. J. Psychopharmacol. 2020;34:452–466. doi: 10.1177/0269881119896043. [DOI] [PubMed] [Google Scholar]
- Sinha R. Chronic stress, drug use, and vulnerability to addiction. Ann. N. Y. Acad. Sci. 2008;1141:105–130. doi: 10.1196/annals.1441.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sinkeviciute I., Begemann M., Prikken M., Oranje B., Johnsen E., et al. Efficacy of different types of cognitive enhancers for patients with schizophrenia: a meta-analysis. NPJ Schizophr. 2018;4:22. doi: 10.1038/s41537-018-0064-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith A.G., Neill J.C., Costall B. The dopamine D3/D2 receptor agonist 7-OH-DPAT induces cognitive impairment in the marmoset. Pharmacol. Biochem. Behav. 1999;63:201–211. doi: 10.1016/s0091-3057(98)00230-5. [DOI] [PubMed] [Google Scholar]
- Snyder H.R. Major depressive disorder is associated with broad impairments on neuropsychological measures of executive function: a meta-analysis and review. Psychol. Bull. 2013;139:81–132. doi: 10.1037/a0028727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Snyder K., Wang W.W., Han R., McFadden K., Valentino R.J. Corticotropin-releasing factor in the norepinephrine nucleus, locus coeruleus, facilitates behavioral flexibility. Neuropsychopharmacology. 2012;37:520–530. doi: 10.1038/npp.2011.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sosa J.L.R., Buonomano D., Izquierdo A. The orbitofrontal cortex in temporal cognition. Behav. Neurosci. 2021;135:154–164. doi: 10.1037/bne0000430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spannenburg L., Reed H. Adverse cognitive effects of glucocorticoids: a systematic review of the literature. Steroids. 2023;200 doi: 10.1016/j.steroids.2023.109314. [DOI] [PubMed] [Google Scholar]
- Stalnaker T.A., Franz T.M., Singh T., Schoenbaum G. Basolateral amygdala lesions abolish orbitofrontal-dependent reversal impairments. Neuron. 2007;54:51–58. doi: 10.1016/j.neuron.2007.02.014. [DOI] [PubMed] [Google Scholar]
- Starcke K., Brand M. Effects of stress on decisions under uncertainty: a meta-analysis. Psychol. Bull. 2016;142:909–933. doi: 10.1037/bul0000060. [DOI] [PubMed] [Google Scholar]
- Starkman M.N. Neuropsychiatric findings in Cushing syndrome and exogenous glucocorticoid administration. Endocrinol Metab Clin North Am. 2013;42:477–488. doi: 10.1016/j.ecl.2013.05.010. [DOI] [PubMed] [Google Scholar]
- Starkman M.N., Gebarski S.S., Berent S., Schteingart D.E. Hippocampal formation volume, memory dysfunction, and cortisol levels in patients with Cushing's syndrome. Biol Psychiatry. 1992;32:756–765. doi: 10.1016/0006-3223(92)90079-f. [DOI] [PubMed] [Google Scholar]
- Sterling P., Eyer J. In: Handbook of Life Stress, Cognition and Health. Fisher S., Reason J., editors. John Wiley &Sons; New York: 1988. Allostasis: a new paradigm to explain arousal pathology; pp. 629–649. [Google Scholar]
- Stippl A., Scheidegger M., Aust S., Herrera A., Bajbouj M., et al. Ketamine specifically reduces cognitive symptoms in depressed patients: an investigation of associated neural activation patterns. J. Psychiatr. Res. 2021;136:402–408. doi: 10.1016/j.jpsychires.2021.02.028. [DOI] [PubMed] [Google Scholar]
- Stone D.B., Tesche C.D. Transcranial direct current stimulation modulates shifts in global/local attention. Neuroreport. 2009;20:1115–1119. doi: 10.1097/WNR.0b013e32832e9aa2. [DOI] [PubMed] [Google Scholar]
- Stone E.A., Platt J.E. Brain adrenergic receptors and resistance to stress. Brain Res. 1982;237:405–414. doi: 10.1016/0006-8993(82)90452-8. [DOI] [PubMed] [Google Scholar]
- Street L.J., Sternfeld F., Jelley R.A., Reeve A.J., Carling R.W., et al. Synthesis and biological evaluation of 3-heterocyclyl-7,8,9,10-tetrahydro-(7,10-ethano)-1,2,4-triazolo[3,4-a]phthalazines and analogues as subtype-selective inverse agonists for the GABA(A)alpha5 benzodiazepine binding site. J. Med. Chem. 2004;47:3642–3657. doi: 10.1021/jm0407613. [DOI] [PubMed] [Google Scholar]
- Stuss D.T., Levine B., Alexander M.P., Hong J., Palumbo C., et al. Wisconsin Card Sorting Test performance in patients with focal frontal and posterior brain damage: effects of lesion location and test structure on separable cognitive processes. Neuropsychologia. 2000;38:388–402. doi: 10.1016/s0028-3932(99)00093-7. [DOI] [PubMed] [Google Scholar]
- Sun H., Cocker P.J., Zeeb F.D., Winstanley C.A. Chronic atomoxetine treatment during adolescence decreases impulsive choice, but not impulsive action, in adult rats and alters markers of synaptic plasticity in the orbitofrontal cortex. Psychopharmacology (Berl) 2012;219:285–301. doi: 10.1007/s00213-011-2419-9. [DOI] [PubMed] [Google Scholar]
- Sun J., Jia K., Sun M., Zhang X., Chen J., et al. The GluA1-related BDNF pathway is involved in PTSD-induced cognitive flexibility deficit in attentional set-shifting tasks of rats. J. Clin. Med. 2022;11 doi: 10.3390/jcm11226824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun X., Zhang Y., Li X., Liu X., Qin C. Early-life neglect alters emotional and cognitive behavior in a sex-dependent manner and reduces glutamatergic neuronal excitability in the prefrontal cortex. Front Psychiatry. 2020;11 doi: 10.3389/fpsyt.2020.572224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swaab D.F., Bao A.M. Sex differences in stress-related disorders: major depressive disorder, bipolar disorder, and posttraumatic stress disorder. Handb. Clin. Neurol. 2020;175:335–358. doi: 10.1016/B978-0-444-64123-6.00023-0. [DOI] [PubMed] [Google Scholar]
- Swartz B.E., McDonald C.R., Patel A., Torgersen D. The effects of guanfacine on working memory performance in patients with localization-related epilepsy and healthy controls. Clin. Neuropharmacol. 2008;31:251–260. doi: 10.1097/WNF.0b013e3181633461. [DOI] [PubMed] [Google Scholar]
- Szabo C., Nemeth A., Keri S. Ethical sensitivity in obsessive-compulsive disorder and generalized anxiety disorder: the role of reversal learning. J Behav Ther Exp Psychiatry. 2013;44:404–410. doi: 10.1016/j.jbtep.2013.04.001. [DOI] [PubMed] [Google Scholar]
- Tait D.S., Brown V.J., Farovik A., Theobald D.E., Dalley J.W., Robbins T.W. Lesions of the dorsal noradrenergic bundle impair attentional set-shifting in the rat. Eur. J. Neurosci. 2007;25:3719–3724. doi: 10.1111/j.1460-9568.2007.05612.x. [DOI] [PubMed] [Google Scholar]
- Tanaka S.C., Doya K., Okada G., Ueda K., Okamoto Y., Yamawaki S. Prediction of immediate and future rewards differentially recruits cortico-basal ganglia loops. Nat. Neurosci. 2004;7:887–893. doi: 10.1038/nn1279. [DOI] [PubMed] [Google Scholar]
- Tank A.W., Lee Wong D. Peripheral and central effects of circulating catecholamines. Compr. Physiol. 2015;5:1–15. doi: 10.1002/cphy.c140007. [DOI] [PubMed] [Google Scholar]
- Tarantino I.S., Sharp R.F., Geyer M.A., Meves J.M., Young J.W. Working memory span capacity improved by a D2 but not D1 receptor family agonist. Behav. Brain Res. 2011;219:181–188. doi: 10.1016/j.bbr.2010.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tedford S.E., Persons A.L., Napier T.C. Dopaminergic lesions of the dorsolateral striatum in rats increase delay discounting in an impulsive choice task. PLoS One. 2015;10 doi: 10.1371/journal.pone.0122063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terry A.V., Jr., Gearhart D.A., Warner S., Hohnadel E.J., Middlemore M.L., et al. Protracted effects of chronic oral haloperidol and risperidone on nerve growth factor, cholinergic neurons, and spatial reference learning in rats. Neuroscience. 2007;150:413–424. doi: 10.1016/j.neuroscience.2007.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terry A.V., Jr., Gearhart D.A., Warner S.E., Zhang G., Bartlett M.G., et al. Oral haloperidol or risperidone treatment in rats: temporal effects on nerve growth factor receptors, cholinergic neurons, and memory performance. Neuroscience. 2007;146:1316–1332. doi: 10.1016/j.neuroscience.2007.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thai C.A., Zhang Y., Howland J.G. Effects of acute restraint stress on set-shifting and reversal learning in male rats. Cogn Affect Behav Neurosci. 2013;13:164–173. doi: 10.3758/s13415-012-0124-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomassin H., Flavin M., Espinas M.L., Grange T. Glucocorticoid-induced DNA demethylation and gene memory during development. EMBO J. 2001;20:1974–1983. doi: 10.1093/emboj/20.8.1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson S.M., Josey M., Holmes A., Brigman J.L. Conditional loss of GluN2B in cortex and hippocampus impairs attentional set formation. Behav. Neurosci. 2015;129:105–112. doi: 10.1037/bne0000045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson S.M., Kallarackal A.J., Kvarta M.D., Van Dyke A.M., LeGates T.A., Cai X. An excitatory synapse hypothesis of depression. Trends in Neuroscience. 2015;38:279–294. doi: 10.1016/j.tins.2015.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiemensma J., Andela C.D., Biermasz N.R., Romijn J.A., Pereira A.M. Mild cognitive deficits in patients with primary adrenal insufficiency. Psychoneuroendocrinology. 2016;63:170–177. doi: 10.1016/j.psyneuen.2015.09.029. [DOI] [PubMed] [Google Scholar]
- Torregrossa M.M., Xie M., Taylor J.R. Chronic corticosterone exposure during adolescence reduces impulsive action but increases impulsive choice and sensitivity to yohimbine in male Sprague-Dawley rats. Neuropsychopharmacology. 2012;37:1656–1670. doi: 10.1038/npp.2012.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tozzi L., Goldstein-Piekarski A.N., Korgaonkar M.S., Williams L.M. Connectivity of the cognitive control network during response inhibition as a predictive and response biomarker in major depression: evidence from a randomized clinical trial. Biol Psychiatry. 2020;87:462–472. doi: 10.1016/j.biopsych.2019.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsigos C., Kyrou I., Kassi E., Chrousos G.P. In: Stress: Endocrine Physiology and Pathophysiology in Endotext. Feingold K.R., Anawalt B., Blackman M.R., Boyce A., Chrousos G., et al., editors. 2000. South Dartmouth (MA) [PubMed] [Google Scholar]
- Turner D.C., Clark L., Dowson J., Robbins T.W., Sahakian B.J. Modafinil improves cognition and response inhibition in adult attention-deficit/hyperactivity disorder. Biol Psychiatry. 2004;55:1031–1040. doi: 10.1016/j.biopsych.2004.02.008. [DOI] [PubMed] [Google Scholar]
- Turner K.M., Simpson C.G., Burne T.H.J. Touchscreen-based visual discrimination and reversal tasks for mice to test cognitive flexibility. Bio Protoc. 2017;7 doi: 10.21769/BioProtoc.2583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tuscher J.J., Taxier L.R., Fortress A.M., Frick K.M. Chemogenetic inactivation of the dorsal hippocampus and medial prefrontal cortex, individually and concurrently, impairs object recognition and spatial memory consolidation in female mice. Neurobiol. Learn. Mem. 2018;156:103–116. doi: 10.1016/j.nlm.2018.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tytherleigh M.Y., Vedhara K., Lightman S.L. Mineralocorticoid and glucocorticoid receptors and their differential effects on memory performance in people with Addison's disease. Psychoneuroendocrinology. 2004;29:712–723. doi: 10.1016/S0306-4530(03)00103-3. [DOI] [PubMed] [Google Scholar]
- Uribe-Marino A., Gassen N.C., Wiesbeck M.F., Balsevich G., Santarelli S., et al. Prefrontal cortex corticotropin-releasing factor receptor 1 conveys acute stress-induced executive dysfunction. Biol Psychiatry. 2016;80:743–753. doi: 10.1016/j.biopsych.2016.03.2106. [DOI] [PubMed] [Google Scholar]
- Vaiana A.M., Asher A.M., Tapia K., Morilak D.A. Vortioxetine reverses impairment of visuospatial memory and cognitive flexibility induced by degarelix as a model of androgen deprivation therapy in rats. Neuroendocrinology. 2023 doi: 10.1159/000535365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Schaaf M.E., van Schouwenburg M.R., Geurts D.E., Schellekens A.F., Buitelaar J.K., et al. Establishing the dopamine dependency of human striatal signals during reward and punishment reversal learning. Cereb Cortex. 2014;24:633–642. doi: 10.1093/cercor/bhs344. [DOI] [PubMed] [Google Scholar]
- Verharen J.P.H., Adan R.A.H., Vanderschuren L. Differential contributions of striatal dopamine D1 and D2 receptors to component processes of value-based decision making. Neuropsychopharmacology. 2019;44:2195–2204. doi: 10.1038/s41386-019-0454-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viola T.W., Creutzberg K.C., Zaparte A., Kestering-Ferreira E., Tractenberg S.G., et al. Acute neuroinflammation elicited by TLR-3 systemic activation combined with early life stress induces working memory impairments in male adolescent mice. Behav. Brain Res. 2019;376 doi: 10.1016/j.bbr.2019.112221. [DOI] [PubMed] [Google Scholar]
- Vogel S., Fernandez G., Joels M., Schwabe L. Cognitive adaptation under stress: a case for the mineralocorticoid receptor. Trends Cogn Sci. 2016;20:192–203. doi: 10.1016/j.tics.2015.12.003. [DOI] [PubMed] [Google Scholar]
- Volkow N.D., Chang L., Wang G.J., Fowler J.S., Ding Y.S., et al. Low level of brain dopamine D2 receptors in methamphetamine abusers: association with metabolism in the orbitofrontal cortex. Am J Psychiatry. 2001;158:2015–2021. doi: 10.1176/appi.ajp.158.12.2015. [DOI] [PubMed] [Google Scholar]
- Volkow N.D., Fowler J.S., Wang G.J., Hitzemann R., Logan J., et al. Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse. 1993;14:169–177. doi: 10.1002/syn.890140210. [DOI] [PubMed] [Google Scholar]
- Voon V., Irvine M.A., Derbyshire K., Worbe Y., Lange I., et al. Measuring "waiting" impulsivity in substance addictions and binge eating disorder in a novel analogue of rodent serial reaction time task. Biol Psychiatry. 2014;75:148–155. doi: 10.1016/j.biopsych.2013.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace A., Pehrson A.L., Sanchez C., Morilak D.A. Vortioxetine restores reversal learning impaired by 5-HT depletion or chronic intermittent cold stress in rats. Int. J. Neuropsychopharmacol. 2014;17:1695–1706. doi: 10.1017/S1461145714000571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X.D., Chen Y., Wolf M., Wagner K.V., Liebl C., et al. Forebrain CRHR1 deficiency attenuates chronic stress-induced cognitive deficits and dendritic remodeling. Neurobiol. Dis. 2011;42:300–310. doi: 10.1016/j.nbd.2011.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warburton E.C., Brown M.W. Neural circuitry for rat recognition memory. Behav. Brain Res. 2015;285:131–139. doi: 10.1016/j.bbr.2014.09.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe Y., Gould E., McEwen B.S. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Res. 1992;588:341–345. doi: 10.1016/0006-8993(92)91597-8. [DOI] [PubMed] [Google Scholar]
- Watanabe Y., Stone E., McEwen B.S. Induction and habituation of c-fos and zif/268 by acute and repeated stressors. Neuroreport. 1994;5:1321–1324. [PubMed] [Google Scholar]
- Weerda R., Muehlhan M., Wolf O.T., Thiel C.M. Effects of acute psychosocial stress on working memory related brain activity in men. Hum. Brain Mapp. 2010;31:1418–1429. doi: 10.1002/hbm.20945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei J., Yuen E.Y., Liu W., Li X., Zhong P., et al. Estrogen protects against the detrimental effects of repeated stress on glutamatergic transmission and cognition. Mol Psychiatry. 2014;19:588–598. doi: 10.1038/mp.2013.83. [DOI] [PubMed] [Google Scholar]
- Weidacker K., Johnston S.J., Mullins P.G., Boy F., Dymond S. Impulsive decision-making and gambling severity: the influence of gamma-amino-butyric acid (GABA) and glutamate-glutamine (Glx) Eur. Neuropsychopharmacol. 2020;32:36–46. doi: 10.1016/j.euroneuro.2019.12.110. [DOI] [PubMed] [Google Scholar]
- Wellman C.L. Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration. J. Neurobiol. 2001;49:245–253. doi: 10.1002/neu.1079. [DOI] [PubMed] [Google Scholar]
- Wellman C.L., Bollinger J.L., Moench K.M. Effects of stress on the structure and function of the medial prefrontal cortex: insights from animal models. Int. Rev. Neurobiol. 2020;150:129–153. doi: 10.1016/bs.irn.2019.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wieland L., Ebrahimi C., Katthagen T., Panitz M., Luettgau L., et al. Acute stress alters probabilistic reversal learning in healthy male adults. Eur. J. Neurosci. 2023;57:824–839. doi: 10.1111/ejn.15916. [DOI] [PubMed] [Google Scholar]
- Williams G.V., Castner S.A. Under the curve: critical issues for elucidating D1 receptor function in working memory. Neuroscience. 2006;139:263–276. doi: 10.1016/j.neuroscience.2005.09.028. [DOI] [PubMed] [Google Scholar]
- Williams G.V., Goldman-Rakic P.S. Modulation of memory fields by dopamine D1 receptors in prefrontal cortex. Nature. 1995;376:572–575. doi: 10.1038/376572a0. [DOI] [PubMed] [Google Scholar]
- Williams G.V., Rao S.G., Goldman-Rakic P.S. The physiological role of 5-HT2A receptors in working memory. J. Neurosci. 2002;22:2843–2854. doi: 10.1523/JNEUROSCI.22-07-02843.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams L.M. Precision psychiatry: a neural circuit taxonomy for depression and anxiety. Lancet Psychiatr. 2016;3:472–480. doi: 10.1016/S2215-0366(15)00579-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willner P. Chronic mild stress (CMS) revisited: consistency and behavioural-neurobiological concordance in the effects of CMS. Neuropsychobiology. 2005;52:90–110. doi: 10.1159/000087097. [DOI] [PubMed] [Google Scholar]
- Willner P., Towell A., Sampson D., Sophokleous S., Muscat R. Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology. 1987;93:358–364. doi: 10.1007/BF00187257. [DOI] [PubMed] [Google Scholar]
- Wilner A.P., de Varennes B., Gregoire P.A., Lupien S., Pruessner J.C. Glucocorticoids and hippocampal atrophy after heart transplantation. Ann. Thorac. Surg. 2002;73:1965–1967. doi: 10.1016/s0003-4975(01)03502-0. [DOI] [PubMed] [Google Scholar]
- Wingenfeld K., Otte C. Mineralocorticoid receptor function and cognition in health and disease. Psychoneuroendocrinology. 2019;105:25–35. doi: 10.1016/j.psyneuen.2018.09.010. [DOI] [PubMed] [Google Scholar]
- Wingenfeld K., Wolf S., Krieg J.C., Lautenbacher S. Working memory performance and cognitive flexibility after dexamethasone or hydrocortisone administration in healthy volunteers. Psychopharmacology (Berl) 2011;217:323–329. doi: 10.1007/s00213-011-2286-4. [DOI] [PubMed] [Google Scholar]
- Winstanley C.A., Baunez C., Theobald D.E., Robbins T.W. Lesions to the subthalamic nucleus decrease impulsive choice but impair autoshaping in rats: the importance of the basal ganglia in Pavlovian conditioning and impulse control. Eur. J. Neurosci. 2005;21:3107–3116. doi: 10.1111/j.1460-9568.2005.04143.x. [DOI] [PubMed] [Google Scholar]
- Winstanley C.A., Dalley J.W., Theobald D.E., Robbins T.W. Fractionating impulsivity: contrasting effects of central 5-HT depletion on different measures of impulsive behavior. Neuropsychopharmacology. 2004;29:1331–1343. doi: 10.1038/sj.npp.1300434. [DOI] [PubMed] [Google Scholar]
- Winstanley C.A., Eagle D.M., Robbins T.W. Behavioral models of impulsivity in relation to ADHD: translation between clinical and preclinical studies. Clin. Psychol. Rev. 2006;26:379–395. doi: 10.1016/j.cpr.2006.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Winstanley C.A., Theobald D.E., Cardinal R.N., Robbins T.W. Contrasting roles of basolateral amygdala and orbitofrontal cortex in impulsive choice. J. Neurosci. 2004;24:4718–4722. doi: 10.1523/JNEUROSCI.5606-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Winstanley C.A., Theobald D.E., Dalley J.W., Robbins T.W. Interactions between serotonin and dopamine in the control of impulsive choice in rats: therapeutic implications for impulse control disorders. Neuropsychopharmacology. 2005;30:669–682. doi: 10.1038/sj.npp.1300610. [DOI] [PubMed] [Google Scholar]
- Winstanley C.A., Theobald D.E.H., Dalley J.W., Glennon J.C., Robbins T.W. 5-HT2A and 5-HT2C receptor antagonists have opposing effects on a measure of impulsivity: interactions with global 5-HT depletion. Psychopharmacology. 2004;176:376–385. doi: 10.1007/s00213-004-1884-9. [DOI] [PubMed] [Google Scholar]
- Witkin J.M., Lippa A., Smith J.L., Jin X., Ping X., et al. The imidazodiazepine, KRM-II-81: an example of a newly emerging generation of GABAkines for neurological and psychiatric disorders. Pharmacol. Biochem. Behav. 2022;213 doi: 10.1016/j.pbb.2021.173321. [DOI] [PubMed] [Google Scholar]
- Wood G.E., Young L.T., Reagan L.P., McEwen B.S. Acute and chronic restraint stress alter the incidence of social conflict in male rats. Horm. Behav. 2003;43:205–213. doi: 10.1016/s0018-506x(02)00026-0. [DOI] [PubMed] [Google Scholar]
- Woolley B. Growing off-label neuro-cognitive uses for guanfacine? An informal review of publications from 2022 with discussion about two clinical trials scheduled to conclude in 2023-2024. Issues Ment. Health Nurs. 2023;44:923–925. doi: 10.1080/01612840.2023.2242236. [DOI] [PubMed] [Google Scholar]
- Worbe Y., Savulich G., Voon V., Fernandez-Egea E., Robbins T.W. Serotonin depletion induces 'waiting impulsivity' on the human four-choice serial reaction time task: cross-species translational significance. Neuropsychopharmacology. 2014;39:1519–1526. doi: 10.1038/npp.2013.351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright R.L., Conrad C.D. Enriched environment prevents chronic stress-induced spatial learning and memory deficits. Behav. Brain Res. 2008;187:41–47. doi: 10.1016/j.bbr.2007.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yalin N., Kempton M.J., Mazibuko N., Mehta M.A., Young A.H., Stokes P.R. Mifepristone enhances the neural efficiency of human visuospatial memory encoding and recall. Psychoneuroendocrinology. 2021;125 doi: 10.1016/j.psyneuen.2020.105116. [DOI] [PubMed] [Google Scholar]
- Yao Y., Silver R. Mutual shaping of circadian body-wide synchronization by the suprachiasmatic nucleus and circulating steroids. Front. Behav. Neurosci. 2022;16 doi: 10.3389/fnbeh.2022.877256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yaple Z.A., Yu R. Fractionating adaptive learning: a meta-analysis of the reversal learning paradigm. Neurosci. Biobehav. Rev. 2019;102:85–94. doi: 10.1016/j.neubiorev.2019.04.006. [DOI] [PubMed] [Google Scholar]
- Yates J.R., Bardo M.T. Effects of intra-accumbal administration of dopamine and ionotropic glutamate receptor drugs on delay discounting performance in rats. Behav. Neurosci. 2017;131:392–405. doi: 10.1037/bne0000214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yates J.R., Batten S.R., Bardo M.T., Beckmann J.S. Role of ionotropic glutamate receptors in delay and probability discounting in the rat. Psychopharmacology (Berl) 2015;232:1187–1196. doi: 10.1007/s00213-014-3747-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoon J.H., Grandelis A., Maddock R.J. Dorsolateral prefrontal cortex GABA concentration in humans predicts working memory load processing capacity. J. Neurosci. 2016;36:11788–11794. doi: 10.1523/JNEUROSCI.1970-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young A.H., Gallagher P., Watson S., Del-Estal D., Owen B.M., Ferrier I.N. Improvements in neurocognitive function and mood following adjunctive treatment with mifepristone (RU-486) in bipolar disorder. Neuropsychopharmacology. 2004;29:1538–1545. doi: 10.1038/sj.npp.1300471. [DOI] [PubMed] [Google Scholar]
- Yu M., Zhang Y., Chen X., Zhang T. Antidepressant-like effects and possible mechanisms of amantadine on cognitive and synaptic deficits in a rat model of chronic stress. Stress. 2016;19:104–113. doi: 10.3109/10253890.2015.1108302. [DOI] [PubMed] [Google Scholar]
- Yuen E.Y., Liu W., Karatsoreos I.N., Feng J., McEwen B.S., Yan Z. Acute stress enhances glutamatergic transmission in prefrontal cortex and facilitates working memory. Proc Natl Acad Sci U S A. 2009;106:14075–14079. doi: 10.1073/pnas.0906791106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuen E.Y., Liu W., Karatsoreos I.N., Ren Y., Feng J., et al. Mechanisms for acute stress-induced enhancement of glutamatergic transmission and working memory. Mol Psychiatry. 2011;16:156–170. doi: 10.1038/mp.2010.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zaehle T., Sandmann P., Thorne J.D., Jancke L., Herrmann C.S. Transcranial direct current stimulation of the prefrontal cortex modulates working memory performance: combined behavioural and electrophysiological evidence. BMC Neurosci. 2011;12:2. doi: 10.1186/1471-2202-12-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zanos P., Gould T.D. Mechanisms of ketamine action as an antidepressant. Mol Psychiatry. 2018;23:801–811. doi: 10.1038/mp.2017.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang M.W., Ho R.C. Controversies of the effect of ketamine on cognition. Front Psychiatry. 2016;7:47. doi: 10.3389/fpsyt.2016.00047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou J., Yang L., Yang G., Gao J. The efficacy and safety of some new GABAkines for treatment of depression: a systematic review and meta-analysis from randomized controlled trials. Psychiatry Res. 2023;328 doi: 10.1016/j.psychres.2023.115450. [DOI] [PubMed] [Google Scholar]