Abstract
The burden of anxiety disorders is growing, but the efficacy of available anxiolytic treatments remains inadequate. Cognitive behavioural therapy for anxiety disorders focuses on identifying and modifying maladaptive patterns of thinking and behaving, and has a testable analogue in rodents in the form of fear extinction. A large preclinical literature has amassed in recent years describing the neural and molecular basis of fear extinction in rodents. In this review, we discuss how this work is being harnessed to foster translational research on anxiety disorders and facilitate the search for new anxiolytic treatments. We begin by summarizing the anatomical and functional connectivity of a medial prefrontal cortex (mPFC)–amygdala circuit that subserves fear extinction, including new insights from optogenetics. We then cover some of the approaches that have been taken to model impaired fear extinction and associated impairments with mPFC–amygdala dysfunction. The principal goal of the review is to evaluate evidence that various neurotransmitter and neuromodulator systems mediate fear extinction by modulating the mPFC–amygdala circuitry. To that end, we describe studies that have tested how fear extinction is impaired or facilitated by pharmacological manipulations of dopamine, noradrenaline, 5-HT, GABA, glutamate, neuropeptides, endocannabinoids and various other systems, which either directly target the mPFC–amygdala circuit, or produce behavioural effects that are coincident with functional changes in the circuit. We conclude that there are good grounds to be optimistic that the progress in defining the molecular substrates of mPFC–amygdala circuit function can be effectively leveraged to identify plausible candidates for extinction-promoting therapies for anxiety disorders.
Linked Articles
This article is part of a themed section on Animal Models in Psychiatry Research. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-20
Introduction
Prevalence and treatment of anxiety disorders
Anxiety disorders constitute some of the most widely known and commonly diagnosed neuropsychiatric problems, affecting a significant number of people around the world (Kessler et al., 2011; Wittchen et al., 2011). The broad diagnostic category of anxiety disorders encompasses a range of conditions, including generalized anxiety disorder, panic disorder (PD), various types of phobias and post-traumatic stress disorder (PTSD) (DSM-5, 2013; World Health Organisation, 1994). This categorization has been revised somewhat in the most recent revision of the Diagnostic and Statistical Manual of Mental Disorders, which now separates anxiety disorders into three main subcategories: anxiety disorders, obsessive–compulsive and related disorders, and trauma- and stressor-related disorders (DSM-5, 2013).
Despite modifications in the way anxiety disorders are diagnosed, which is based primarily on symptomatology, there remains considerable overlap in the medications used to treat the different disorders. The main Food and Drug Administration-approved anxiolytic treatments are the benzodiazepines (e.g. clonazepam, lorazepam) and β-blockers (e.g. propranolol), and the tricyclic (e.g. imipramine, clomipramine), MAO A inhibitor (e.g. phenelzine, isocarboxazid) and 5-HT and noradrenaline re-uptake inhibitors (e.g. fluoxetine, escitalopram, venlafaxine) classes of antidepressants. It is noteworthy that none of these drugs were developed for anxiety based on a biological hypothesis of their mechanism of action, but are in use today largely because of serendipitous discoveries of their beneficial clinical effects.
Anxiety disorders are also often treated with various forms of psychotherapy. One commonly employed psychological approach is cognitive behavioural therapy (CBT), which focuses on identifying and modifying maladaptive patterns of thinking and behaving. Recent meta-analysis shows that CBT has efficacy across anxiety disorders, including PTSD, PD and generalized anxiety disorder (Stewart and Chambless, 2009). With the goal of improving long-term therapeutic outcome, patients are often treated with a combination of CBT, or some other psychotherapy, and a regimen of chronic drug treatment. Compelling evidence that such combinations are more effective than either approach alone is, however, lacking (Barlow et al., 2000; Otto et al., 2010; Rodrigues et al., 2011). Indeed, the notion that the combination of drugs and CBT should work to enhance therapeutic efficacy is predicated on the idea that the two interventions will act in an additive or synergistic manner. While intuitively appealing, this view is not often grounded in an understanding of the brain processes by which CBT acts to alleviate anxiety. Nor does it account for how adjunctive drug treatments might affect these processes, either beneficially or deleteriously, to determine the net therapeutic impact of a combination of treatments.
Towards mechanism-based treatments
A number of authors have argued that the development of successful pharmacological adjuncts to CBT must stem from a ‘mechanism-up’ approach built on a deep understanding of the neural circuit underlying anxiety, and the identification of drugs that target these circuits (Myers and Davis, 2007; Ressler and Mayberg, 2007; Holmes and Quirk, 2010; Graham et al., 2011; Steckler and Risbrough, 2012). Particularly amenable to this approach are those anxiety disorders, such as PTSD and phobias, that typically result from identifiable traumatic events and that are triggered by clear, definable environmental reminders of the trauma. CBT often focuses on severing the cognitive link between environmental cues and trauma by repeated exposure to these cues. The generation and maintenance of trauma-like memories can be readily modelled in rodents using well-established paradigms based on classical conditioning, in which discrete cues or contexts are paired with an aversive outcome (e.g. footshock) to generate a ‘fear’ memory that is measurable by expression of defensive behaviour (e.g. freezing). Once formed, the capacity to extinguish a fear memory can also be assayed, by measuring the degree to which fear is reduced as a result of repeated presentation of a conditioned stimulus without concomitant footshock. Fear extinction has been employed by a growing number of preclinical anxiety studies, and strongly benefits from having a strong clinical parallel in the form of exposure-based CBT.
Recent years have seen significant progress in elucidating the neural basis of conditioned fear and extinction in rodents. A goal of the current review was to discuss how work is being harnessed to foster translational research on anxiety disorders and facilitate the search for new anxiolytic treatments. To keep the review manageable, we limit our focus to fear extinction and largely avoid studies on fear conditioning, even though those studies would sometimes be informative to the discussion. We first offer an overview of the anatomical and functional connectivity of a medial prefrontal cortex (mPFC)–amygdala circuit that is most strongly implicated in learned fear and extinction – given this is the foundation for mechanism-based modelling and drug discovery. A growing number of rodent models of impaired extinction have been developed. We review the general approaches that have been taken to model extinction and discuss evidence linking some models to mPFC–amygdala dysfunction. We then turn to the chief focus of the review – a summary of studies that have examined how various neurotransmitter and neuromodulator systems might mediate fear extinction via modulation of the mPFC–amygdala circuit. Finally, we consider the potential for moving preclinical targets forward into eventual therapeutic use and the challenges that would need to be overcome.
Functional circuitry of fear and extinction
Anatomical connections between the mPFC and amygdala
Our understanding of the neural circuitry mediating fear and extinction in rodents rests on an increasingly detailed description of the anatomical connections between the PFC and the amygdala. Glutamatergic afferents to the amygdala, arising from cortical pyramidal cell layers 2 and 5 (Gabbott et al., 2005; Little and Carter, 2012; 2013), course ventrally through the striatum or stria terminalis. Tract-tracing studies in the rat show that axons originating in the infralimbic cortex of the mPFC terminate most densely in the ventromedial lateral nucleus, the rostral part of the accessory basal amygdala, lateral capsular subdivision of the central nucleus and the superficial nuclei (lateral olfactory tract, periamygdaloid cortex and cortical nuclei) (Cassell and Wright, 1986; McDonald et al., 1996; McDonald, 1998; Pinard et al., 2012). Neurons in the more caudal areas of the infralimbic subregion also project to the medial and intermediate subdivisions of the central nucleus (Hurley et al., 1991; McDonald, 1998).
The prelimbic cortex of the mPFC is located dorsally adjacent to the infralimbic subregion and it has a different pattern of connectivity with the amygdala. Prelimbic cortex neurons target the basal nucleus of the amygdala (BA), primarily the dorsomedial portion (McDonald, 1991; 1998; McDonald et al., 1996; Vertes, 2004), while caudal prelimbic cortex neurons concentrate inputs in the medial parvicellular basal nucleus (Sesack et al., 1989). In turn, baso-lateral amygdala (BLA) neurons project back to both the prelimbic cortex and infralimbic subregion, creating a feedback loop (Krettek and Price, 1977; Shinonaga et al., 1994; Conde et al., 1995; Gabbott et al., 2006; Hoover and Vertes, 2007). These BLA inputs to the mPFC exert an inhibitory influence over cortical pyramidal cells, probably via engagement of local interneurons, although excitatory modulation (at least in the prelimbic cortex) is also reported (Perez-Jaranay and Vives, 1991; Ishikawa and Nakamura, 2003; Sotres-Bayon et al., 2012; Sun and Laviolette, 2012; Dilgen et al., 2013).
Anatomical tracing studies show that the infralimbic subregion and prelimbic cortex only sparsely innervate the main, fear-generating, output nucleus of amygdala – the medial nucleus of the central amygdala (CeM) – raising the question of how mPFC neurons modulate fear. One influential model posits that this connection is bridged by infralimbic subregion inputs to the intercalated cell nuclei (ICN) of the amygdala (Royer et al., 1999; Quirk et al., 2003; Pare et al., 2004; Amano et al., 2010; Amir et al., 2011; Li et al., 2011). The ICNs comprise narrow clusters of densely packed, mainly GABAergic, neurons associated with the fibre bundles that lie between the BA/lateral amygdala (LA) and the central lateral (CeL)/central medial (CeM) nuclei, but have a molecular phenotype more akin to striatal neurons (Millhouse, 1986; Nitecka and Ben-Ari, 1987; McDonald and Augustine, 1993; McDonald, 1998; Kaoru et al., 2010; Manko et al., 2011). This model has recently been refined to emphasize the heterogeneity of different ICNs. The medial paracapsular and main ICN nucleus (IN) show a high degree of interconnectivity, but also exhibit diverse electrophysiological and molecular profiles and are differentially activated by fear and fear extinction (Geracitano et al., 2007; Kaoru et al., 2010; Busti et al., 2011).
Infralimbic subregion axons also project to a network of local ICN dendrites located in a region termed the capsular infralimbic subregion target zone (CITZ) (Cassell and Wright, 1986; McDonald et al., 1996; Vertes, 2004; Marowsky et al., 2005; Pinto and Sesack, 2008; Busti et al., 2011; Pinard et al., 2012). It is worth noting, however, that although the densest mPFC projections to the CITZ arise from the infralimbic subregion, there are also some projections from the prelimbic cortex to the CITZ (McDonald et al., 1996). A final point to bear in mind is, in contrast to the mPFC and major amygdala nuclei, which have analogues in the human brain (note, for an excellent recent review of putatively analogous circuits in humans, see Vanelzakker et al., 2014), the literature on the CITZ and ICNs is largely based on rodents and it remains to be shown whether these structures are similarly integral to the corticoamygdala circuitry in the primate and human brain. In this context, at least one study has reported mPFC (subgenual cortex/area 25) projections to the ICNs in non-human primates (Freedman et al., 2000).
Functional mPFC–amygdala interactions – new insights from optogenetics
Delineating the functional contribution of these mPFC–amygdala circuits to fear in rodents has been the subject of intense recent study using techniques including lesioning and transient inactivation, analysis of patterns of gene expression, and in vivo and ex vivo neural recordings. This extensive literature has been covered in many excellent reviews (see following citations) and we will not overburden the current paper by retreading this ground. To distill some of the principal conclusions – the infralimbic subregion clearly plays a role in fear inhibition and fear extinction (Milad and Quirk, 2002; Berretta et al., 2005; Likhtik et al., 2005; Sierra-Mercado et al., 2006; Knapska and Maren, 2009; Busti et al., 2011; Knapska et al., 2012), whereas the prelimbic cortex has been implicated in the generation and maintenance of fear (Burgos-Robles et al., 2009; Laurent and Westbrook, 2009a; Sierra-Mercado et al., 2011; Knapska et al., 2012; Li et al., 2012; Sotres-Bayon et al., 2012; Courtin et al., 2014a; Fenton et al., 2014). With regard to the amygdala, the LA, BA, CeL and CeM have all been found to be critical to the formation and/or expression of fear memories (Herry et al., 2010; Pape and Pare, 2010; Orsini and Maren, 2012), while the BA, basomedial amygdala and ICNs are important for extinction (Repa et al., 2001; Anglada-Figueroa and Quirk, 2005; Herry et al., 2008; Likhtik et al., 2008; Knapska and Maren, 2009; Amano et al., 2011; Busti et al., 2011; Lesting et al., 2011; Sierra-Mercado et al., 2011; Livneh and Paz, 2012; Courtin et al., 2014b; Trouche et al., 2013). It should be borne in mind throughout this review that the acquisition, consolidation and retrieval of extinction are separable processes that are under the control of different brain regions and neural systems (Plendl and Wotjak, 2010). However, for the sake of readability, we will refer to studies of extinction without detailing whether the focus was on one or more of these processes.
As in other areas of neuroscience, studies aimed at delineating the neural basis of fear and extinction have begun to take advantage of some powerful optogenetic tools, that allow for precise temporal, and in some instances, molecularly defined, control of specific circuits (Johansen et al., 2012; Lammel et al., 2014). By replacing footshock with optogenetic stimulation of LA pyramidal neurons during cue presentations in a pseudo-conditioning session, Johansen and colleagues were able to generate a mild auditory fear memory (Johansen et al., 2010). Conversely, optogenetic inhibition of the LA/BA during conditioning leads to severe impairments in cued and context fear learning (Goshen et al., 2011). Interestingly, stimulating a subpopulation of pyramidal neurons in the BA expressing Thy1 also produced impaired fear learning, but actually strengthened fear extinction – demonstrating how the effects of optogenetic manipulations in this region will depend on the cell type targeted (Jasnow et al., 2013). With regards to the central nucleus of the amygdala (CeA), optogenetically stimulating neurons in CeM evoked unconditioned fear, consistent with this nuclei’s function as the system’s major output station (Ciocchi et al., 2010). The activity of the CeM is regulated by distinct subpopulations of CeL neurons that are either excited or inhibited by conditioned fear stimuli (Haubensak et al., 2010; Duvarci et al., 2011). Optogenetic control of these different subpopulations bi-directionally regulates conditioned fear, revealing an intricate microcircuit in which ‘CeL on’ cells inhibit ‘CeL off’ cells to disinhibit CeM output and increase conditioned fear (Haubensak et al., 2010). It will be interesting to extend these observations to the analysis of extinction.
Optogenetics has been employed in a number of fear studies focusing on the mPFC and the region’s links to the amygdala. One example examined the effects of optogenetic inhibition of the anterior cingulate cortex (ACC) on time-dependent fear memory recall, and confirming the findings of lesion/inactivation studies (Bontempi et al., 1999), showed that the ACC was necessary for remotely, but not recently, retrieved contextual fear memory (Goshen et al., 2011). Another study applied sustained optogenetic activation of infralimbic subregion/prelimbic cortex pyramidal cells, but not parvalbumin-positive interneurons, during fear conditioning and found that this produced reductions in later fear expression (Yizhar et al., 2011). More in-depth analysis of the mPFC parvalbumin-positive interneurons finds that these cells do exert a major influence on fear extinction. Courtin and colleagues identified a subpopulation of parvalbumin interneurons in the mPFC (primarily prelimbic cortex) that showed high firing during low fear states, suggestive of a fear-inhibiting function (Courtin et al., 2014a). Supporting this suggestion, optogenetically inhibiting these cells produced increased fear and reversed the fear suppressing effects of extinction, likely by disinhibiting the activity of prelimbic cortex pyramidal neurons innervating the BLA (Courtin et al., 2014a).
Complimenting these findings, another recent study sought to elucidate how the functional connections between the mPFC and amygdala changed with extinction. Here, mPFC-originating (infralimbic subregion or prelimbic cortex) fibres in the BLA were optogenetically stimulated to reveal how extinction decreased the strength of excitatory mPFC projections to pyramidal neurons in the BLA (Cho et al., 2013), an effect analogous to the decreased excitability of electrically mPFC-activated BLA neurons after extinction (Vouimba and Maroun, 2011). The authors suggest the decrease in the excitatory influence of the mPFC over the BLA may lessen BLA drive of CeM output and rebalance the circuitry in favour of mPFC inputs to CeM-inhibiting ICNs, thereby enabling extinction (Cho et al., 2013). In an illustration of how extinction leads to bidirectional changes in the mPFC–amygdala circuit, Senn et al. used an elegant combination of viral tools to electrophysiologically record from and optogenetically manipulate neurons projecting from the BA to the subregions of the mPFC (Senn et al., 2014). Using this approach, they found not only that BA neurons projecting to prelimbic cortex and infralimbic subregion are activated during fear and extinction, respectively, but that optogenetically inhibiting BA-prelimbic cortex neurons promoted extinction, whereas silencing BA-infralimbic subregion projections impaired extinction (Senn et al., 2014).
These early studies have already highlighted how optogenetics can prove very useful for establishing the diverse roles of components of the mPFC–amygdala circuit to fear and extinction. The technique even has the power to isolate the contributions of specific neural ensembles within the mPFC–amygdala circuitry. This is illustrated by recent work in which optogenetically reactivating only those cells in the dentate gyrus region of the hippocampus recruited during contextual fear conditioning was able to elicit an ‘artificial’ fear response without context re-exposure (Liu et al., 2012; Ramirez et al., 2013). Using optogenetic techniques such as these, it is likely that the field will soon gain some new important insights into the neural circuitry mediating extinction.
Rodent models of impaired extinction and mPFC–amygdala dysfunction
Various rodent models of impaired extinction have been developed and tested for abnormalities in the structure and function of mPFC–amygdala circuitry. These models generally fall into one of three broad conceptual categories. One set of models exploits differences in extinction that emerge within a rodent population or between different rodent strains, while another is based around explicit exposure to environmental insults such as stress or a drug of abuse (Holmes and Singewald, 2013). The third encompasses models of engineered mutations in specific genes, and we will consider these later, within the context of the relevant neurotransmitter systems.
In an early illustration of the utility of segregating a population of C57BL/6 mice based on extinction performance as a means to reveal underlying mechanisms, deficient extinction associated with reduced neuronal activation in mPFC (note, throughout the review, we will refer to ‘mPFC’ in most cases where infralimbic subregion and prelimbic cortex were not specified) and BLA as well as abnormal mPFC synaptic plasticity (Herry and Mons, 2004). Along similar lines, sorting Sprague Dawley rats into good and poor extinguishers showed that poor extinction is related to increased firing of neurons in the prelimbic cortex and attenuated burst firing of infralimbic subregion neurons (Burgos-Robles et al., 2007; 2009). This approach has also revealed how rats with relatively poor extinction exhibit less activation of infralimbic subregion inputs to the LA, but hyperactivation of prelimbic cortex inputs to this same region of the amygdala (Knapska et al., 2012).
An alternative to the post hoc sorting of subjects based on extinction performance is to select mice, a priori, for anxiety-related traits and breed these traits into phenotypically divergent subpopulations. In this manner, rats bred for stable high anxiety-like behaviour exhibit deficient extinction associated with the hypoactivation of neurons in the infralimbic subregion and BLA and the hyperactivation of cells in the CeM (Muigg et al., 2008). Functional variation in the mPFC–amygdala circuit also underpins differences in fear extinction found across different genetically inbred mouse strains (for an in-depth discussion, see Holmes and Singewald, 2013). For instance, deficient extinction in the 129S1/SvImJ mouse strain is coupled to a range of mPFC–amygdala abnormalities, including infralimbic subregion hypoactivation, prelimbic cortex hyperactivation and neuronal hyperactivity, and at the level of the amygdala, hypoactivation and dendritic hypertrophy in the BLA and CeL, as well as CeM hyperactivation (Hefner et al., 2008; Whittle et al., 2010; Camp et al., 2012; Fitzgerald et al., 2014).
Disturbances to the mPFC–amygdala circuitry are also related to the impaired extinction produced by certain environmental insults. For instance, chronic exposure to alcohol causes extinction deficits tied to the down-regulation of NMDA receptors (for nomenclature see Alexander et al., 2013a) in the mPFC and the loss of infralimbic subregion neuronal firing (Bertotto et al., 2006; Holmes et al., 2012). An even larger body of literature demonstrates marked effects of stress on fear extinction and brain function. Early work found that extinction is highly sensitive to deleterious effects of exposure to stressors including forced swim and restraint (Izquierdo et al., 2006; Miracle et al., 2006). The observation of a deleterious effect on extinction has since been extended to a wide range of stressors, both acute and chronic. These range from maternal separation to social defeat to elevated platform exposure (Matsumoto et al., 2008; 2013; Yamamoto et al., 2008; 2009; Goswami et al., 2010; Judo et al., 2010; Andero et al., 2011; Green et al., 2011; Wilber et al., 2011; Chauveau et al., 2012; Dubreucq et al., 2012; Ishikawa et al., 2012; Knox et al., 2012a; Long and Fanselow, 2012; Toledo-Rodriguez et al., 2012; Deschaux et al., 2013; Ganon-Elazar and Akirav, 2013; Saito et al., 2012; 2013; Segev et al., 2014; Wilson et al., 2013; Zhang and Rosenkranz, 2013; Zheng et al., 2013). Manipulation of stress-related signalling by exogenous corticosterone administration (Gourley et al., 2009; Bingham et al., 2013), blockade of corticosterone synthesis (via metyrapone) (Blundell et al., 2011) or forebrain deletion of the mineralocorticoid receptor (Ter Horst et al., 2012), also impair extinction – although there are other examples in which corticosterone administration can facilitate extinction (Cai et al., 2006; Brinks et al., 2009).
The stress-impairing effects on extinction can be traced to an array of functional alterations in the mPFC–amygdala circuit. These include infralimbic subregion dendritic hypotrophy, prelimbic cortex neuronal hyperactivity, increased BLA synaptic excitability and neuronal spine density, mPFC glucocorticoid receptor up-regulation and NMDA receptor, L-α-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid ionotropic (AMPA) receptor and ERK down-regulation, and impaired synaptic plasticity both in the mPFC and reciprocal pathways interconnecting the mPFC and the amygdala (Maroun and Richter-Levin, 2003; Izquierdo et al., 2006; Maroun, 2006; Gourley et al., 2009; Wilber et al., 2009; 2011; Judo et al., 2010; Chauveau et al., 2012; Ishikawa et al., 2012; Knox et al., 2012b; Toledo-Rodriguez et al., 2012; Bingham et al., 2013; Maroun et al., 2013). The role of the infralimbic subregion is further demonstrated by the finding that infralimbic subregion lesions occlude stress-induced impairments of extinction (Farrell et al., 2010). Also of note, performing extinction training soon after conditioning impairs extinction (the ‘immediate extinction effect’) (Maren and Chang, 2006; Myers et al., 2006; Macpherson et al., 2013; Maren, 2014). Although the immediate extinction procedure was not explicitly designed as a stressor, the effect could possibly reflect the stress of fear conditioning temporarily impairing mPFC function and thereby hampering extinction (Maren, 2014). Indeed, immediate extinction deficits are linked to functional correlates in the mPFC that are reminiscent of those produced by various stressors, including reductions in mPFC neuronal bursting and hyperactivation of the prelimbic cortex (Chang et al., 2010; Kim et al., 2010; Stafford et al., 2013).
Stress effects on extinction are strongly influenced by the age of subjects. Extinction in younger rodents at pre-weaning age produces more robust reductions in fear than seen in adults (Callaghan et al., 2013). This form of juvenile extinction parallels the stage of development at which extracellular matrix structures known as perineuronal nets have not yet fully developed around parvalbumin-positive interneurons in the BLA (Gogolla et al., 2009; Karpova et al., 2012). It is also related to the loss of synaptic plasticity and dendritic spine density in mPFC caused by haploinsufficiency of the extracellular matrix protein, reelin (Ammassari-Teule et al., 2009; Iafrati et al., 2013).
Post-natal stress has recently been shown to expedite the development of the adult-like form of extinction in pre-weaning rats, and may do so by catalysing the development of BLA perineuronal nets or mPFC–amygdala connectivity into the adult form, although this remains to be formally tested (Callaghan and Richardson, 2012; Cowan et al., 2013). In contrast to pre-weaning rodents, adolescent rats and mice show a resistance to extinction (as compared with adults) that is associated with a lack of neuronal activation and synaptic plasticity in the infralimbic subregion (McCallum et al., 2010; Kim et al., 2011; Pattwell et al., 2012), and may reflect the immaturity of the infralimbic subregion and a functional bias towards the fear-promoting prelimbic cortex at this ontogenic time point (Chan et al., 2011; Li et al., 2012).
These findings clearly show how age is an important factor moderating mPFC–amygdala mediation of fear extinction. More generally, the consistent finding to emerge from a diverse set of rodent models is that impaired extinction is closely linked with the functional deficiencies at certain nodes in the mPFC circuit (particularly the infralimbic subregion, BLA and CeL), and a corresponding over-engagement of other areas (notably the prelimbic cortex and CeM).
Neurotransmitters and neuromodulators in mPFC–amygdala-mediated extinction
Our growing understanding of the mPFC–amygdala circuitry subserving extinction, together with the availability of models of deficient extinction linked to circuit dysfunction, offers rich opportunities for identifying novel anxiety treatments. A reasonable and potentially tractable route is to develop anxiolytic drugs that target ‘druggable’ neurotransmitter and molecular systems known to modulate the functions of the mPFC–amygdala circuit. In the present section, we review some of the major systems studied to date that may have offered such candidates.
Dopamine
There are prominent dopamine projections arising from the ventral midbrain to both the mPFC and amygdala (Pinard et al., 2008; Pinto and Sesack, 2008), and a number of authors have suggested this neurotransmitter could exert a major influence on fear extinction (for an excellent recent review, see Abraham et al., 2014). In vitro, the activation of dopamine receptors (via apomorphine), or stimulation of D1-like receptors (for nomenclature see Alexander et al., 2013a) specifically (via SKF3892 or SKF81297), attenuates mPFC-evoked inhibition of BLA pyramidal neuronal firing, possibly by stimulating local inhibitory interneurons (Rosenkranz and Grace, 1999; 2002). Another source of BLA modulation by dopamine comes from the midbrain dopaminergic neurons that synapse onto BLA pyramidal, and predominantly parvalbumin-positive interneurons (Brinley-Reed and McDonald, 1999; Pinard et al., 2008; Pinto and Sesack, 2008; Muller et al., 2009). These inputs probably underlie the increases in LA neuronal oscillatory inhibitory network activity that can be produced by dopamine, mimicked by D1-like agonists (dihydrexidine, SK81297) and blocked by D1-like (using SCH23390), but not the D2-like (via sulpiride) antagonists (Bissiere et al., 2003; Loretan et al., 2004; Kroner et al., 2005). These effects of dopamine could potentially exert strong effects on extinction in view of the importance of amygdala network activity for fear (Rainnie et al., 2006; Pape and Pare, 2010).
Dopamine’s effects on extinction circuitry are not limited to the BLA. The CeA and ICNs are also innervated by dopamine, with D1-like receptors showing particularly rich expression on the ICNs (Asan, 1998; Fuxe et al., 2003; Marowsky et al., 2005; Jacobsen et al., 2006; Pinto and Sesack, 2008; Pinard et al., 2012). Dopamine suppresses the excitability of ICNs in a manner that is mimicked by D1-like (dihydrexidine), but not D2-like (quinpirole), receptor agonism, and prevented by D1-like (SCH23390), but not D2-like (sulpiride), receptor antagonism (Marowsky et al., 2005; Manko et al., 2011). Acting through D1 receptors in this way, dopamine could depress ICN activity and augment amygdala output, especially under conditions of high dopamine release, such as fear and stress (Marowsky et al., 2005). In turn, the actions of D1 receptors will be influenced by various factors regulating dopamine availability, such as the dopamine-clearing organic cation transporter 3, which is expressed on the ICNs and is linked to the regulation of stress (Baganz et al., 2011; Hill and Gasser, 2013). Collectively, these anatomical and electrophysiological data suggest that dopamine is well positioned to regulate amygdala-mediated fear extinction at any one of a number of levels. Although most of these data implicate D1-like receptors in these effects, a contribution of D2-like receptors should not be discounted. In vitro application of a D2-like agonist (quinpirole), but not a D1-like agonist (SKF38393) results in the amplification of LA neuronal excitability (Rosenkranz and Grace, 1999), suppression of LA interneuronal feedforward inhibition and the release of synaptic plasticity at pyramidal neurons (Bissiere et al., 2003). Moreover, in vivo, systemic activation of D2-like receptors (again using quinpirole) attenuates BLA-driven suppression of neuron firing in the mPFC (Floresco and Tse, 2007). Thus, D2-like receptors can excite both the BLA and mPFC and it would be unlikely that this would not translate into changes in fear extinction under at least some conditions.
Given dopamine has such profound effects on the mPFC–amygdala circuitry, how do genetic and pharmacological manipulations of dopamine and its receptors influence extinction? Constitutive gene deletion of the D1 receptor produces impaired extinction (El-Ghundi et al., 2001), and systemic administration of drugs that increase dopamine (using the dopamine re-uptake inhibitors methamphetamine, d-amphetamine or cocaine), or activate D1-like receptors (via SKF38393), also either impair extinction or have no effect on the behaviour (Miczek and Luttinger, 1978; Borowski and Kokkinidis, 1998; Mueller et al., 2009; Carmack et al., 2010). The effects of systemically targeting D2-like receptors are also somewhat mixed. Systemic D2-like agonism (with quinpirole) disrupts extinction (Nader and LeDoux, 1999), whereas blockade of D2-like receptors (with haloperidol, sulpiride or raclopride) can either facilitate or impair extinction (Nader and LeDoux, 1999; Ponnusamy et al., 2005; Holtzman-Assif et al., 2010; Mueller et al., 2010). In part, these discrepancies may be attributable to the lack of selectivity for specific subtypes within the D1-like (comprising the D1 and D5 subtypes) and D2-like (made up of the D2, D3 and D4 subtypes) receptor families, given the roles of individual subtypes are only now being uncovered (Holmes et al., 2004). Of note in this context, studies using selective D4 receptor agonists (PD-168,077) and antagonists (L-741,741) suggest that D4 receptors promote BLA-driven mPFC neuronal excitation (Laviolette et al., 2005; Floresco and Tse, 2007), and that mPFC D4 receptor inhibition (via L-741,741) is sufficient to impair extinction (Pfeiffer and Fendt, 2006). Further studies that make use of selective pharmacological probes at other dopamine receptor subtypes, once these become available, will be essential to refining our understanding of dopamine’s role in extinction.
Another issue that could contribute to the apparent variability in effects produced by systemic dopamine manipulations is the potential for complex and even opposing actions of dopamine at different regions within the broader mPFC–amygdala circuitry. Site-specific infusions and electrophysiological recordings have been one way to broach this issue. Infusing either a D1-like (SCH23390) or D2-like (raclopride) receptor antagonist directly into the infralimbic subregion impairs fear extinction, whereas systemic D2-like inactivation (also using raclopride) reduces the firing of infralimbic subregion neurons during extinction (Hikind and Maroun, 2008; Mueller et al., 2010; Fiorenza et al., 2012). Similar, extinction impairing, effects are produced by blocking D1 receptors in the BLA, as demonstrated by intra-BLA infusion of a D1-like antagonist (SCH23390) (Hikind and Maroun, 2008; Fiorenza et al., 2012). Thus, the effect of inactivating D1 receptors is actually quite consistent across these two brain regions. This then poses the questions of whether augmenting dopamine availability at D1 receptors would exert a pro-extinction profile.
As already noted, there are examples of ‘dopamine boosters,’ including psychostimulant drugs, that have detrimental or few effects on extinction. More positively, however, a recent study reported the facilitation of extinction in humans and mice following systemic administration of the dopamine precursor L-DOPA or a dopamine (and noradrenaline) re-uptake inhibitor (methylphenidate) (Abraham et al., 2012; Haaker et al., 2013). These pro-extinction effects of L-DOPA are associated with increased neuronal activation in the mPFC and a depression of CeM amygdala output in mice, as well as increased midbrain–mPFC functional coupling in human volunteers (Haaker et al., 2013). This pattern of activation tentatively suggests that the behavioural effect of L-DOPA may occur by promoting dopamine activity in the mPFC. In support of this scheme, mPFC dopamine levels are found to rise during extinction, while depleting dopamine in the mPFC (via locally applied 6-hydroxdopamine) impairs extinction (Morrow et al., 1999; Fernandez Espejo, 2003; Hugues et al., 2007; Saito et al., 2013). However, further work will be needed to clarify the precise mechanisms underlying L-DOPA’s effects on extinction. These include elucidating the contribution of dopamine from that of noradrenaline, a neurotransmitter we consider in the next section.
Noradrenaline
Various lines of evidence implicate the ascending noradrenaline system in extinction, with indications that both the mPFC and BLA may be involved. For instance, extinction produces increases in endogenous levels of noradrenaline in the mPFC (Hugues et al., 2007), where it may produce neuronal excitement (Mueller et al., 2008). Furthermore, infusing noradrenaline into the infralimbic subregion or BLA leads to an enhancement or impairment of extinction respectively (Berlau and McGaugh, 2006; Fiorenza et al., 2012). Improvements in extinction can also be produced by systemic administration of drugs (e.g. methylphenidate) that increase levels of noradrenaline (as well as dopamine), presumably in both the mPFC and amygdala (and elsewhere) (Abraham et al., 2012). Much of the research aimed at extending these observations has focused on delineating the specific noradrenaline receptors involved.
There is accumulating support for both α- and β-adrenoceptors in extinction. Systemic blockade of α2-adrenoceptors (using yohimbine) facilitates extinction (Cain et al., 2004; Morris and Bouton, 2007), although it is unclear whether this drug’s effects can be attributable solely to the α2-adrenoceptor because the behavioural effect is not mimicked by a more selective antagonist (atipamezole) (Davis et al., 2008) (for further discussion, see Holmes and Quirk, 2010). Systemic treatment with a β-adrenoceptor agonist (isoprenaline) (Do Monte et al., 2010) also facilitates extinction, whereas an antagonist at this receptor (propranolol) disrupts extinction in some studies, but only in other studies if given repeatedly or used to oppose the pro-extinction effects of a NMDA receptor partial agonist (D-cycloserine) (Cain et al., 2004; Ouyang and Thomas, 2005; Rodriguez-Romaguera et al., 2009; Do Monte et al., 2010; Yamada et al., 2011; Archbold et al., 2013). These findings indicate that targeting β-adrenoceptors can affect extinction, but not always in a straightforward manner. A similar inference can be made from studies that have examined the consequences of regional infusions of drugs acting on β-adrenoceptors.
On the one hand, stimulating β-adrenoceptors (using isoprenaline) specifically within the infralimbic subregion facilitates extinction, but on the other hand, infusions given into the BLA after fear is reactivated lead to impairments in extinction (Do Monte et al., 2010; Debiec et al., 2011). Conversely, blocking β-adrenoceptors (with propranolol) in the infralimbic subregion impairs extinction, while intra-BLA infusion of another β-adrenoceptor blocker (timolol) enhances extinction (Mueller et al., 2008; Fiorenza et al., 2012). Taken together, these findings generally fit a scheme in which increasing noradrenaline signalling through β-adrenoceptors in the infralimbic subregion favours extinction, whereas promoting such signalling in the BLA opposes extinction. Contrary to this scheme, there is one report of enhanced extinction after intra-mPFC β-adrenoceptor antagonist (timolol) administration, the mPFC-wide nature of this infusion would have encompassed regions outside of the infralimbic subregion (Fiorenza et al., 2012). Nonetheless, additional studies are warranted before any firm conclusions can be made and noradrenaline-acting drugs considered for possible clinical development as adjuncts to exposure therapy. Of note in this regard, treatment with propranolol has been proposed as a potential exposure-adjunct and prophylactic treatment for PTSD when given soon after trauma, but has received limited or discouraging clinical support (Orr et al., 2006; Bos et al., 2012; Hoge et al., 2012; Soeter and Kindt, 2012). Preliminary results using yohimbine as an adjunct to exposure therapy have been somewhat more promising (Powers et al., 2009), but as noted, it remains questionable whether its effects can be solely attributable to actions on the noradrenaline system.
5-HT
The 5-hydroxytryptaminergic system is of special interest as a target for extinction-modulating drugs in view of the use of 5-hydroxytryptaminergic-acting drugs, such as the selective 5-HT re-uptake inhibitors (SSRIs), to treat anxiety disorders, sometimes in combination with exposure therapy (Schneier et al., 2012; Yang et al., 2012; Bui et al., 2013). A number of studies have examined the extinction-related effects of genetically ablating or pharmacologically inhibiting the primary target of SSRIs, the 5-HT transporter (SERT, see Alexander et al., 2013b). This has shown that mutant mice and rats lacking SERT are extinction impaired and exhibit abnormalities in the dendritic morphology and spine density of pyramidal neurons in the infralimbic subregion and BLA (Wellman et al., 2007; Nietzer et al., 2011; Hartley et al., 2012; Nonkes et al., 2012; Riddle et al., 2013). Electrophysiological recordings find that SERT-deficient mutants also show increased synchronization of θ wave activity between the LA and mPFC during extinction (Narayanan et al., 2011), which is reminiscent of the altered functional mPFC–amygdala coupling reported in humans with a loss-of-function mutation in the SERT gene (Heinz et al., 2005; Pezawas et al., 2005). Thus, genetic loss of the SERT may cause a functional rebalancing of the mPFC–amygdala circuitry that favours fear over extinction.
While the SERT is blocked by SSRIs, it is clear that the effects of pharmacological inhibition are not synonymous with those produced by genetic disturbances, probably because of lasting genetic influences on brain development (for discussion, see Caspi et al., 2010). Indeed, fear and extinction are sensitive to disruptions of the developing 5-hydroxytryptaminergic system, for example by deleting Pet-1, a transcription factor critically involved in 5-hydroxytryptaminergic neuron development (Wellman et al., 2013). In contrast to the extinction-impairing effects of SERT gene mutation, a number of studies find that systemic chronic fluoxetine treatment facilitates extinction under most experimental conditions (c.f., caloric restriction) (Norcross et al., 2008; Spennato et al., 2008; Deschaux et al., 2011; 2013; Camp et al., 2012; Karpova et al., 2012; Fitzgerald et al., 2014; Riddle et al., 2013). Systemic treatment with a non-selective MAO inhibitor, methylene blue, also improves extinction and increases cytochrome oxidase activity in the mPFC (Gonzalez-Lima and Bruchey, 2004; Wrubel et al., 2007). However, while a similar extinction-facilitating effect is seen with certain other monoaminergic antidepressants (e.g. venlafaxine) (Yang et al., 2012), the opposite (i.e. extinction impairing) effect is seen after treatment with the SERT citalopram (Burghardt and Bauer, 2013).
One culprit for these discrepancies may be the differences in the pharmacological profiles and downstream actions of SSRIs. The 5-hydroxytryptaminergic system is notoriously complex and has a high number (over 14) of receptor subtypes, many expressed in the mPFC and amygdala (Holmes, 2008). Citalopram is a relatively specific inhibitor of SERT, but down-regulates BLA expression of the NMDA receptor subtype 2B (GluN2B) subunit, which as discussed later, would be predicted to impair extinction (Burghardt and Bauer, 2013; Burghardt et al., 2013). Fluoxetine has a number of ‘off-target’ effects, that include antagonist actions at the 5-HT2C receptor subtype, but it remains unclear whether these direct pharmacological actions or certain downstream changes underlie the drug’s facilitatory effects on extinction. Of relevance here, fluoxetine decreases the number of perineuronal nets around parvalbumin-positive interneurons in the BLA, which as discussed earlier, is linked to the superior extinction shown by juvenile rodents (Karpova et al., 2012). This suggests at least one potential mechanism for the drug’s extinction effects, although it would be valuable to show that citalopram, for example, failed to affect a change in BLA perineuronal nets. It would also be useful to have a clearer picture of the contribution of specific 5-HT receptors to fear extinction. Unfortunately, there remains a paucity of literature on this topic. One recent study shows that systemic administration of a 5-HT1A receptor partial agonist (tandospirone) ameliorated extinction deficits and associated deficits in mPFC synaptic plasticity generated in a model of juvenile stress (Saito et al., 2013), although the effects are attributed to increases in mPFC dopamine levels, rather than changes in 5-hydroxytryptaminergic transmission per se.
Various 5-HT receptors, including the 5-HT1A, 5-HT2A and 5-HT3A subtypes, are well expressed in the BLA, suggesting another possible site whereby 5-HT could affect extinction (Mascagni and McDonald, 2007; McDonald and Mascagni, 2007). In vitro recordings show that 5-HT inhibits glutamate-induced excitation of BLA pyramidal neurons possibly, via activation of GABAergic interneurons, in a manner that is mimicked by a 5-HT2 receptor agonist (α-methyl-5-HT), but not a 5-HT1A receptor agonist (8-OH-DPAT) (Rainnie, 1999; Stutzmann and LeDoux, 1999). 5-HT2 modulation of BLA neuronal activity could account for the pro-extinction effects recently reported after systemic administration of a 5-HT2A receptor agonist (TCB-2), but this remains speculative in lieu of more directed experiments (Zhang and Rosenkranz, 2013; Zhang et al., 2013). Also intriguing, but preliminary, is the finding that systemic blockade of 5-HT3 receptors (using granisetron) improves extinction, while constitutively deleting the 5-HT3A gene impairs extinction (Park and Williams, 2012; Kondo et al., 2013). Establishing a potential link with the amygdala and the GABA system, the pro-extinction effects of 5-HT3 receptor blockade were paralleled by increases in the amygdala expression of gephyrin, a GABAA receptor clustering protein (Park and Williams, 2012). The GABA system will be the focus in the next section.
In summary, there currently is a surprising dearth of research on 5-HT’s role in extinction, given the dominance of 5-HT-acting drugs in anxiolytic drug market. A priority for the field going forward will be both to elucidate the extinction-related effects of targeting specific 5-HT receptor subtypes and clarify the mechanisms of action by which first-line anxiolytic treatments, such as fluoxetine, promote extinction in preclinical assays.
GABA
By providing the major source of inhibitory neurotransmission in the mPFC and amygdala, GABA exerts a powerful influence on a range of fear- and anxiety-related behaviours, including fear extinction (for a detailed discussion, see Ehrlich et al., 2009; Makkar et al., 2010; Pape and Pare, 2010; Courtin et al., 2013). The effect of stimulating GABAA receptors can be so robust that GABAA receptor agonists (such as muscimol) are often used an experimental tool to temporally inactivate a specific brain region and thereby probe its contribution to extinction (for receptor nomenclature see Alexander et al., 2013c). Temporary inactivations have been used to help establish necessary contribution of the infralimbic subregion or BLA (but not prelimbic cortex) to extinction (Sierra-Mercado et al., 2006; 2011; Laurent and Westbrook, 2008; 2009a; 2010; Laurent et al., 2008; Parkes and Westbrook, 2010; Sotres-Bayon et al., 2012; Holmes and Singewald, 2013; Holmes et al., 2013). Disruptions to extinction (possibly involving effects on memory reconsolidation) after augmenting GABAA receptor signalling is also achieved by delivering benzodiazepine agonists (diazepam, chlordiazepoxide, midazolam) systemically or directly into the BLA (Kamano, 1972; Goldman, 1977; Pereira et al., 1989; Bouton et al., 1990; Bustos et al., 2009; Hart et al., 2009; 2010).
GABAA receptor drug manipulations have bidirectional effects on extinction. For example, delivering a GABAA receptor antagonist (picrotoxin) either systemically or directly into the infralimbic subregion facilitates extinction (McGaugh et al., 1990; Thompson et al., 2010; Chang and Maren, 2011; Fitzgerald et al., 2014). This behavioural effect could stem from the disinhibition of infralimbic subregion projections to the amygdala, which would be in line with the observation that the ICNs and LA neurons are activated by intra-infralimbic subregion GABAA receptor blockade (using picrotoxin) (Berretta et al., 2005). Disinhibition of amygdala neuronal activity could also possibly explain the improvement in extinction produced by infusion of a GABAA receptor antagonist (bicuculline) directly into the BLA (Berlau and McGaugh, 2006). Another route by which GABAA receptors in the amygdala might modulate extinction is at the level of the ICNs. GABAA receptors containing the α2/α3 subunits are expressed on ICNs projecting to the CeA and are physiologically sensitive to application of a benzodiazepine agonist (diazepam), GABA potentiator (zolpidem) and specific α3 subunit agonist (TP003) (Marowsky et al., 2005; Geracitano et al., 2012). The behavioural consequence of pharmacologically targeting these receptors is an interesting question that has not been addressed at the current time.
As the complex pattern of receptor expression suggests, the role of GABA in extinction is more nuanced than simply turning a brain region on or off with a GABAA receptor agonist or antagonist. Extinction testing up-regulates the infralimbic subregion and BLA expression of glutamic acid decarboxylase isoform (GAD67), an enzyme that controls the synthesis of GABA in the brain (Heldt and Ressler, 2007; Sangha et al., 2012) and promotes the binding and clustering of GABAA receptors in the BLA (Chhatwal et al., 2005b; Heldt and Ressler, 2007; Lin et al., 2009a). Extinction is impaired either by BLA viral knockdown of GAD67 or by constitutive deletion of the other GAD isoform, GAD65 (Sangha et al., 2009; Heldt et al., 2012). Thus, effective extinction appears to recruit and require GABAergic signalling in the mPFC–amygdala circuitry, perhaps to shape the plastic changes in circuitry that underlies extinction. This may go some way to explain ostensibly paradoxical cases whereby infusion of a GABAA receptor agonist (muscimol) into either the infralimbic subregion or BLA facilitates extinction (Akirav et al., 2006), and where systemic administration of a GABA signalling-reducing benzodiazepine partial inverse agonist (FG 7142) impairs extinction (Harris and Westbrook, 1998; Kim and Richardson, 2007; 2009).
These behavioural findings beg the question of how GABAergic neurotransmission contributes, mechanistically, to extinction. Some authors emphasize a role for GABA in decreasing BLA pyramidal neuronal activity and actively reversing some of the plastic changes produced by fear learning (Lin et al., 2003a,b). In addition, a number of recent studies provide compelling evidence for the integral role played by GABAergic parvalbumin interneurons in the BLA and mPFC. First is the aforementioned study by Courtin et al. showing that parvalbumin interneurons exert an outsized influence in controlling the activity of prelimbic cortex/ACC pyramidal neurons and maintain reduced fear after extinction by suppressing prelimbic cortex excitatory inputs to the amygdala (Courtin et al., 2014a). The dysregulation of prelimbic cortex pyramidal cells could explain why extinction is impaired in mutants with genetically induced loss of parvalbumin-positive interneurons in the mPFC, although this genetic insult was not restricted to the prelimbic cortex in these models (Pitts et al., 2012; Bissonette et al., 2014). Second, Cho et al. have found that excitatory mPFC input to parvalbumin interneurons in the BLA decreases with extinction, which they posit results in the disinhibition of a population of BLA pyramidal neurons and a net shift in favour of the amygdala output-inhibiting mPFC connections to the ICNs, which appear functionally unaltered after extinction (Cho et al., 2013). Cho et al. also observed that fear extinction led to an increase in the inhibition auditory cortical inputs to the BLA that was reversed by a GABAB receptor blocker (CGP52432) (Cho et al., 2013). A contribution of GABAB receptors to extinction-induced amygdala plasticity echoes the observation that extinction increases amygdala expression of the GABAB2 receptor subunit (Heaney et al., 2012). It is also noteworthy, in light of behavioural studies, that systemic delivery of a GABAB receptor antagonist (baclofen) or gene deletion of the GABAB1 receptor subunit lead to impaired extinction, although other studies report no effect of other systemically delivered GABAB receptor antagonists (phaclofen or CGP52432) or positive allosteric modulators (GS39783) (Jacobson et al., 2006; Heaney et al., 2012; Sweeney et al., 2013).
In summary, the available evidence indicates that GABA signalling has important, but nuanced role in extinction. On the one hand, GABA system is recruited during extinction, probably in the service of plasticity mechanisms that reshaping neuronal networks underlying extinction. On the other hand, commonly prescribed drugs such as benzodiazepines, which increase GABAergic tone and effectively alleviate many of the acute symptoms of anxiety, would be expected to limit the efficacy of extinction-based therapies by interfering with the activation of the mPFC–amygdala circuitry. It would appear, therefore, that GABA-targeting pharmacological adjuncts to exposure therapy would need to strike a delicate balance between maintaining GABA activity without overactivating the system.
Glutamate
The involvement of glutamatergic neurotransmission in extinction has been demonstrating by experimental manipulations of the AMPA, metabotropic glutamate (mGlu) and NMDA receptors.
In terms of AMPA receptors, a handful of studies have found that pharmacologically blocking the receptors (via 6-cyano-7-nitroquinoxaline-2,3-dione) in the BLA does not alter extinction (Falls et al., 1992; Lin et al., 2003c; Zimmerman and Maren, 2010). However, activating AMPA receptors, via systemic treatment with an AMPA receptor agonist 2-[2,6-difluoro-4-[[2-[(phenylsulfonyl)amino]ethyl]thio]phenoxy]acetamide (PEPA) facilitates extinction in various rodent models (Zushida et al., 2007; Yamada et al., 2009; 2011), although not in severely extinction-impaired animals (Whittle et al., 2013). Data obtained from combining direct drug infusions (of PEPA) into the mPFC or BLA, with ex vivo electrophysiological analysis of changes in neuronal activity, has identified the mPFC as a possible locus of these drug effects (Zushida et al., 2007).
The precise mechanistic basis for pro-extinction effects of AMPA receptor stimulation is not wholly clear, but may involve alterations in synaptic plasticity, AMPA receptor internalization and intracellular signalling, given evidence that extinction leads to alterations in the phosphorylation and cell-surface expression of AMPA receptors, as well as decoupling from synaptic scaffolding proteins including post-synaptic density 95 (Lin et al., 2003c; Mao et al., 2006; 2008; 2013; Kim et al., 2007b; Lee et al., 2013). In this context, systemic or intra-BLA infusion of a synthetic peptide that blocks activity-dependent internalization of the AMPA receptor subunit, GluA2, impairs extinction (Kim et al., 2007b; Dalton et al., 2008; Lin et al., 2010), while interference with phosphorylation of the AMPA receptor subunit 1 (GluA1) bolsters at least some measures of extinction (Lee et al., 2013). The behavioural actions of AMPA receptor manipulations are also linked to the effects of two signalling molecules downstream of AMPA: calcineurin and PKA. Transgenic or pharmacological inhibition of calcineurin facilitates extinction (Lin et al., 2003a,b; Havekes et al., 2008), whereas mutant or drug-independent inhibition of PKA has the opposite effect (Isiegas et al., 2006; Nijholt et al., 2008). Targeting these downstream molecules could potentially offer alternative approaches to modulating AMPA receptors to affect changes in extinction.
Another route to modifying extinction through glutamate-targeting drugs is through mGlu receptors. Blocking the mGlu1 receptor subtype (using CPCCOEt) disrupts AMPA receptor-mediated synaptic depotentiation in the BLA and produces impairments in extinction (Kim et al., 2007a,b). The mGlu1 receptor is also notable for its expression on neurons innervating in some ICNs, although the possible role of this population of receptors in extinction is unknown (Busti et al., 2011). Of the various other mGlu subtypes, mGlu5 and mGlu7 have been quite well studied for their effects on extinction. Gene deletion of either of these subtypes results in deficits in extinction (Callaerts-Vegh et al., 2006; Goddyn et al., 2008; Xu et al., 2009), suggesting that augmenting function at these subunits could promote extinction. Indeed, pro-extinction effects have been reported with a mGlu7 receptor agonist (AMN082) given either systemically or directly into the BLA, not mPFC (Fendt et al., 2008; Morawska and Fendt, 2012; Toth et al., 2012a; Dobi et al., 2013; Whittle et al., 2013). These effects have been traced to the localization of mGlu7 receptors in the proximity of the ICNs and on local and thalamic glutamatergic inputs in the BLA (Dobi et al., 2013). In terms of mGlu5, results to date show that extinction is disrupted by systemic, intra- infralimbic subregion or intra-BLA blockade of the subtype [via 2-methyl-6-(phenylethynyl)pyridine], but in some instances, only if extinction training is sufficiently deepened with prolonged training (Fontanez-Nuin et al., 2011; Toth et al., 2012a; Mao et al., 2013). These extinction-impairing effects of mGluA5 antagonists have been tied to reduced infralimbic subregion neuronal bursting and the disruption of synaptic plasticity and synaptic insertion of the AMPA GluA2 subunit (Fontanez-Nuin et al., 2011; Sepulveda-Orengo et al., 2013).
In comparison with AMPA and mGlu receptors, NMDA receptors have been extensively studied for their role in fear extinction. Systemic, intra-BLA or intra-mPFC delivery of NMDA receptor blockers (e.g. via MK-801 or AP5) reliably produce deficits in extinction (Falls et al., 1992; Baker and Azorlosa, 1996; Lee and Kim, 1998; Santini et al., 2001; Lin et al., 2003c; Lee et al., 2006; Burgos-Robles et al., 2007; Laurent and Westbrook, 2008; 2009b; Laurent et al., 2008; Chan and McNally, 2009; Parsons et al., 2010; Zimmerman and Maren, 2010; Parkes and Westbrook, 2011; Fiorenza et al., 2012; Holmes et al., 2012). Moreover, NMDA receptors in the BLA are permissive for the extinction-related effects of drugs targeting other systems, such as glucocorticoids. Systemic administration or intra-BLA delivery of a synthetic glucocorticoid (dexamethasone) or glucocorticoid agonist (RU28362) enhances extinction, whereas systemic inhibition of glucocorticoid synthesis (using metyrapone) or intra-BLA glucocorticoid receptor blockade (with mifepristone) impairs extinction (Yang et al., 2006; 2007). The pro-extinction effects of systemic glucocorticoid receptor agonism (using dexamethasone) are occluded by blocking NMDA receptors in the BLA (using MK-801 or DL-AP5), whereas the deficiency in extinction caused by the glucocorticoid synthesis inhibitor (via metyrapone) are reversed by systemic NMDA receptor partial agonist (D-cycloserine) administration (Yang et al., 2007). Achieving a better understanding of these interactions may have clinical implications down the line. A randomized, double-blind, placebo-controlled study of acrophobics found that adjunctive cortisol treatment increased the efficacy of exposure therapy (de Quervain et al., 2011), echoing earlier support for hydrocortisone augmentation of exposure therapy in PTSD (Yehuda and LeDoux, 2007; Surís et al., 2010). The preclinical literature would suggest that consideration should be given to augmenting the effectiveness of such treatment with NMDA receptor-acting drugs.
There has been initial progress in delineating the contribution of specific NMDA receptor subunits to extinction. Transgenic overexpression of the GluN2B subunit facilitates extinction (Tang et al., 1999), while GluN2B-selective antagonism (using ifenprodil or Ro 25-6981) either systemically or specifically within the mPFC or BLA (but not the hippocampus) disrupts extinction learning or relearning (Sotres-Bayon et al., 2007; 2009; Laurent and Westbrook, 2008; Laurent et al., 2008; Dalton et al., 2012; Leaderbrand et al., 2014). Systemic treatment with a GluN2B-selective antagonist (Ro 25-891) also reverses an enhanced extinction phenotype in reelin haploin-sufficient mutants, that is typically only seen in young rodents, to a more adult-like form (Iafrati et al., 2013). The role of other NMDA receptor subunits is an important, but unresolved question. One recent study demonstrated that potentiating the actions of the NMDA receptor subtype 2C/D (GluN2C/D) subunit in the BLA (using CIQ) enhances extinction (Ogden et al., 2014). Another way to achieve nuanced pharmacological modulation of the NMDA receptor is to target the glycine binding site located on the GluN1 subunit. Enhancements in extinction are achieved by intra-BLA inhibition of glycine reuptake (via NFPS) (Mao et al., 2009), or by more direct stimulation of the glycine site by systemic or intra-BLA administration of D-serine or D-cycloserine (Walker et al., 2002; Ledgerwood et al., 2003; 2005; Yang and Lu, 2005; Lee et al., 2006; Mao et al., 2006; 2008; Woods and Bouton, 2006; Weber et al., 2007; Bouton et al., 2008; Matsuda et al., 2010; McCallum et al., 2010; Yamada et al., 2011; Baker et al., 2012; Fiorenza et al., 2012; Toth et al., 2012a). The pro-extinction effects of D-cycloserine extend to models of environmentally induced extinction impairment, including those produced by stress (Matsumoto et al., 2008; Yamamoto et al., 2008; Akirav et al., 2009; Judo et al., 2010), sleep deprivation (Silvestri and Root, 2008) and chronic alcohol exposure (Bertotto et al., 2006). D-cycloserine does not, however, promote extinction under all conditions. For example, systemic D-cycloserine is ineffective when extinction occurs soon after conditioning (Chang and Maren, 2011), is preceded by footshock (Langton and Richardson, 2010) or has to be relearned (Langton and Richardson, 2010). The extinction-related effects of D-cycloserine are also obscured if subjects exhibit poor basal extinction (Tomilenko and Dubrovina, 2007; Weber et al., 2007; Bouton et al., 2008; Hefner et al., 2008; Whittle et al., 2013) or have a history of chronic treatment with D-cycloserine or certain antidepressant (Parnas et al., 2005; Werner-Seidler and Richardson, 2007).
Under conditions where they do manifest, the extinction improving effects of D-cycloserine are probably not solely because of modulation of NMDA receptor function. Rather, these effects can be linked back in part to some of the changes in AMPA receptors and ERK1/2 signalling discussed earlier. D-cycloserine increases BLA AMPA receptor internalization (Mao et al., 2006; 2008) and blocking peptidergically inhibiting the GluA2 subunit occludes the pro-extinction effects produced by intra-BLA D-cycloserine (Lin et al., 2010). Similarly, systemic or intra-BLA administration of ERK1/2 blockers (PD98059, U0-126 or SL327) also reverses the pro-extinction effects of systemic D-cycloserine (Yang and Lu, 2005; Matsuda et al., 2010). Tangentially, ERK1/2 inhibition (via PD98059) also occludes the extinction enhancing effects of Ginkgo biloba extract (EGb761) (Yang et al., 2009) and brain-derived neurotrophic factor (BDNF)-induced phosphorylation of stathmin, a cytoskeleton regulator that, when deleted, promotes extinction and BLA neuronal activation (Shumyatsky et al., 2005; Martel et al., 2012). These findings serve to illustrate the intimate functional interplay between not only different glutamate receptors, but also neurotrophins and intracellular signalling molecules. As such, they underscore how the extinction-related effects of a given compound will typically result from multiple molecular changes that extend beyond the drug’s primary pharmacological target.
Neuropeptides
Various neuropeptide systems have long attracted interest as potential anxiolytics, given their modulatory actions on emotional processes (Holmes et al., 2003). Neuropeptides expressed in the mPFC–amygdala circuit represent a number of interesting potential candidates for novel pharmacological approaches to promoting fear extinction. Two good examples are neuropeptide Y (NPY) and neuropeptide S (NPS), which are localized in the BLA on GABAergic (McDonald and Pearson, 1989) and glutamatergic (Xu et al., 2007) neurons respectively. Extinction is facilitated by i.c.v. infusion of NPY, and this effect is blocked by systemic antagonism of the Y1 receptor subtype (via BIBO3304) (Gutman et al., 2008; Lach and de Lima, 2013). The locus of these effects remains to be precisely established, but might entail the actions of NPY on GABA interneurons in the BLA, given the finding that specifically antagonizing Y1 receptor (again using BIBO3304) in the BLA, or constitutively deleting the subunit, impairs extinction (Gutman et al., 2008; Verma et al., 2012).
The mechanism underlying the modulation of fear extinction by NPS is somewhat clearer, and involves a key role for the amygdala. Intra-BLA infusion of NPS is sufficient to exert a pro-extinction effect and blocking BLA NPS receptors is enough to disrupt extinction (Jungling et al., 2008; Chauveau et al., 2012). These effects might occur through presynaptic NPS receptors on BLA pyramidal neurons; NPS has been shown to increase glutamatergic inputs to (medial paracapsular) ICNs and could thereby dampen CeA output and augment extinction (Jungling et al., 2008). Although these findings strongly implicate the amygdala, NPS could also affect extinction at the level of the mPFC, given the finding that i.c.v. infusion of the peptide increases extracellular levels of dopamine (not 5-HT) in the mPFC (Si et al., 2010).
A number of recent findings have revealed an interesting, albeit complex, role for opioid peptides in extinction. Systemic blockade of κ-opioid receptors (KOP receptors) [via norbinaltorphimine (nor-BNI) ] or gene deletion of dynorphin (but not encephalin or β-endorphin) leads to increased fear and/or poorer extinction, in association with reduced neuronal activity in the BLA and mPFC (Bilkei-Gorzo et al., 2012). However, successful fear extinction correlates with a reduction in the mRNA expression of KOP receptors in the BLA (Knoll et al., 2011) and i.c.v. antagonism (via nor-BNI) can reduce fear renewal after extinction (Cole et al., 2011; 2013) [infusion of a KOP receptor agonist (U50,488) into the nucleus accumbens is also without effect Muschamp et al., 2011]. It is unclear, therefore, whether KOP receptors primarily work to promote or disrupt fear extinction.
With respect to other opioid receptor subtypes, systemic treatment with a subtype non-selective opioid receptor antagonist with preferential binding for μ-opioid receptors (MOP receptors) (naloxone) impairs extinction (McNally and Westbrook, 2003). Blocking MOP receptors specifically within the BLA (again via naloxone) failed to affect extinction (Parsons et al., 2010), suggesting the effect of systemic antagonism may be localized to MOP receptors elsewhere in the brain. In this context inhibiting MOP receptors (again via naloxone) in the periaqueductal grey is sufficient to impair extinction (McNally et al., 2004; Parsons et al., 2010), and this effect is recapitulated by a more selective MOP receptor blocker [CTAP (D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2)], but not selective KOP (with nor-BNI) or δ-opioid receptor (naltrindole) antagonists (McNally, 2005). These various observations show that both KOP and MOP receptors influence extinction, but the precise locus of these effects remains to be determined. One site of particular interest to future work is the ICNs, which express high levels of MOP receptors and extinction is impaired by ablating ICNs using a MOP receptor agonist (demorphin) conjugated to a toxin (Likhtik et al., 2008; Busti et al., 2011; Geracitano et al., 2012; Pinard et al., 2012).
Somatostatin and oxytocin play prominent roles in fear and extinction that have been convincingly linked to the mPFC–amygdala circuit. Fear learning increases excitatory input to somatostatin-positive neurons in the mouse CeL, which could act to dampen CeL inhibitory control of CeM output and thereby release fear and oppose extinction (Li et al., 2013) (c.f. Amano et al., 2012). Correspondingly, optogenetic stimulation of somatostatin-positive CeL neurons produces increases in conditioned fear (Li et al., 2013) (akin to the ‘CeL on’ cells discussed earlier Haubensak et al., 2010), and would be predicted to oppose extinction. Another potential link between somatostatin and extinction is through regulation of AC by the somatostatin receptor-3 subtype, given the finding that gene deletion of the type 3 AC produces extinction of impairment (Wang et al., 2011). The pro-fear function of the CeL’s somatostatin-positive cells contrasts with neighbouring somatostatin-negative cells, which are also GABAergic, but are defined by PKC-δ expression, and reduce CeA output and fear, consistent with the ‘CeL off’ cells discussed earlier (Haubensak et al., 2010; Amano et al., 2012).
Another feature of ‘CeL off’ cells is the expression of oxytocin receptors and modulation by oxytocin (Huber et al., 2005). Local application of oxytocin to the CeL, or optogenetic stimulation of oxytocin-labelled inputs to the CeL from the hypothalamus reduces CeM excitability and attenuates fear (Viviani et al., 2011; Knobloch et al., 2012). This raises the question of whether a similar mechanism may also be recruited during extinction to suppress CeM-driven fear. However, pre-extinction i.c.v. administration of oxytocin has been found to disrupt, rather than promote, extinction, and to do so in a manner that is occluded by oxytocin receptor antagonism [via desGly-NH2,d(CH2)5[Tyr(Me)2,Thr4]OVT; Toth et al., 2012b]. This might very likely reflect opposing extinction-related effects of oxytocin acting in different brain regions. This hypothesis is borne out by the finding that infusion of oxytocin (but not an agonist, [Thr4,Gly7]-oxytocin) into the BLA or dorsal raphe nucleus impairs extinction, whereas infusion into the infralimbic subregion (but not CeA) improves extinction (Kovacs et al., 1979; Lahoud and Maroun, 2013). Such region-based, opposite effects make it difficult to predict with certainty how a systemically delivered oxytocin-acting drug might influence extinction in a clinical setting. Notwithstanding, there is preliminary evidence that oxytocin, given intranasally, can improve extinction in healthy humans (Acheson et al., 2013).
There is nascent support for two functionally related peptides, gastrin and cholecystokinin (CCK), in fear extinction. Deletion of gastrin-releasing peptide (GRP) receptors alters extinction-related neuronal activation in the mPFC (activity is decreased) and BLA (activity is increased) and promotes BLA synaptic plasticity, but appears to have rather mixed effects on extinction, with only one of two studies reporting an impairment (Chaperon et al., 2012; Martel et al., 2012). The endogenous ligand of GRP receptors, GRP, stimulates gastrin, which in turn binds the CCK2 receptor (previously known as CCK-B) subtype. Infusion of a CCK2 receptor agonist (pentagastrin), i.c.v., impairs extinction (Chhatwal et al., 2009), an effect that is occluded by systemic or intra-BLA CCK2 blockade (via CR2945) (Chhatwal et al., 2009). Furthermore, systemic or intra-BLA antagonism of CCK2 receptors (with CR2945) reverses the fear extinction deficits produced by systemic antagonism of CB1 cannabinoid receptors (via SR141716) (Chhatwal et al., 2009). Together, these observations demonstrate that activating CCK2 receptors in the BLA, either through direct agonism or via loss of CB1 receptor-mediated inhibitory control over CCK release (Mascagni and McDonald, 2003), is deleterious to extinction, and, conversely, that blocking this receptor may enable extinction.
Finally, despite being one of the most widely investigated neuropeptides in stress and anxiety, surprisingly, few studies have examined the contribution of corticotropin-releasing factor (CRF) to fear extinction. Infusion of a CRF antagonist (α-helical CRF9-41), i.c.v., prevents fear recovery after extinction (Waddell et al., 2008). This effect could conceivably stem from the potent modulatory actions of CRF on GABA transmission in the amygdala (Rainnie et al., 2004). In this regard, deleting GABAA-α1 receptors solely on CRF neurons is sufficient to impair extinction in a manner reversible by blocking CRF1 receptors (via R121919) systemically or in the bed nucleus of the stria terminalis (Gafford et al., 2012). Nonetheless, there remains much still to understand about the role of CRF in fear extinction.
Endocannabinoids
Already mentioned in the context of a link with CCK system, CB1 receptors are further implicated by a compelling literature tying endocannabinoids with fear extinction (for more detailed recent reviews, see Riebe et al., 2012; Gunduz-Cinar et al., 2013). Numerous reports have shown that gene knockout or systemic blockade (via SR141716A) of CB1 receptors impairs extinction (Marsicano et al., 2002), whereas extinction is promoted by systemically activating CB1 receptors (via WIN55,212-2, arachidonyl-2-chloroethylamide or cannabidiol) or increasing endocannabinoids by blocking re-uptake (via AM404) or inhibiting the anandamide-degrading enzyme fatty acid amide hydrolase (FAAH) (Kathuria et al., 2003) (using AM3506 or URB597) (Marsicano et al., 2002; Cannich et al., 2004; Suzuki et al., 2004; Chhatwal et al., 2005a; Pamplona et al., 2006; 2008; Bitencourt et al., 2008; Kamprath et al., 2009; Gunduz-Cinar et al., 2013; Laricchiuta et al., 2013; Reich et al., 2013; Segev et al., 2014). The mPFC–BLA circuit is implicated as a site of these actions by a number of observations. First, endocannabinoids regulate synaptic plasticity in the BLA and fear extinction increases endocannabinoid levels and recruits various extinction-related intracellular signalling cascades in this region (Marsicano et al., 2002; Cannich et al., 2004; Gunduz-Cinar et al., 2013). Second, CB1 receptor agonism (via WIN55,212-2 or cannabidiol), re-uptake inhibition (using AM404) or FAAH inhibition (with AM3506), either in the BLA, or in at least some studies, the mPFC or infralimbic subregion, promotes fear extinction and prevents stress-induced extinction deficits (Lin et al., 2006; 2009b; Do Monte et al., 2013; Ganon-Elazar and Akirav, 2013; Gunduz-Cinar et al., 2013). Along similar lines, CB1 receptor antagonism (via AM251) limited to the mPFC also impairs extinction (Kuhnert et al., 2013).
The basis for CB1 receptor-mediated control of extinction has being further delineated in studies using sophisticated gene mutant techniques. This work has shown that deletion of CB1 receptors on forebrain or cortical glutamatergic cells or dopamine D1 receptor-expressing neurons is sufficient to disrupt extinction (Kamprath et al., 2009; Terzian et al., 2011). Extinction is also impaired if CB1 receptors are absent on all, but a subpopulation of forebrain glutamatergic neurons (Ruehle et al., 2013), although this same population of CB1 receptors may not mediate stress effects on extinction (Dubreucq et al., 2012). In a more focused examination of CB1 receptor function acting within the mPFC–amygdala circuit, the pro-extinction actions CB1 receptors have been localized to a subset of neurons within the BA (Trouche et al., 2013). Trouche and colleagues found that the excitability of fear-activated BA pyramidal neurons is diminished following extinction in association with an increased inhibitory input from parvalbumin- and CCK-positive, CB1 receptor-expressing, interneurons (Trouche et al., 2013). Taken together, the weight of these and other preclinical findings point to the potential therapeutic benefit of activating CB1 receptors. Encouragingly, initial clinical data show that stimulating cannabinoid receptors with synthetic constituents of cannabis (cannabidiol, dronabinol) promotes extinction in healthy volunteers in some, although not all studies, and increases (blood oxygen level-dependent fMRI) activity within the human mPFC (Klumpers et al., 2012; Das et al., 2013; Rabinak et al., 2013; 2014).
Miscellaneous: from ACh to epigenetics
The intense interest in fear extinction has led researchers to explore the role of myriad neurotransmitter and molecular systems in the process. Covering all of these leads is out of the scope of the current review, but we will touch on some of them in this section.
We have concentrated on the major amine, excitatory and inhibitory neurotransmitters, but there is a more nascent body work concerning a number of other transmitters in fear extinction. One example is the histamine system, which has been found to exert bidirectional effects on mPFC–amygdala-mediated extinction. A histamine H2 receptor antagonist (ranitidine) delivered into the BLA or mPFC impairs extinction, while enhancing histamine in these regions by inhibiting the histamine-metabolizing enzyme, N-methyltransferase (via SKF91488) improves extinction (Fiorenza et al., 2012). Another neurotransmitter, ACh, has a long been established as a mediator of learning is memory, and more recently, identified as a modulator fear extinction. Systemic blockade of muscarinic receptors (scopolamine) enhances extinction by rendering the process context-independent (Zelikowsky et al., 2013). Stimulating muscarinic receptors in the mPFC (via cevimeline) enhances extinction, whereas blocking the receptors systemically or in the mPFC (via scopolamine) impairs extinction (Santini et al., 2012), possibly by attenuating neuronal excitability modulated by the M-type potassium channel (Santini and Porter, 2010). Although there may or may not be a link with muscarinic receptors, or with cholinergic signalling, there is evidence implicating another type of ion channel, the L-type voltage-gated calcium channel (CaV1.x; for nomenclature see Alexander et al., 2013d), in extinction. Blocking (but not stimulating, with BayK) CaV1.x (using nifedipine or verapamil) either systemically or specifically within the BLA (but not i.c.v.) impairs extinction and prevents increases BLA mitogen-activated kinase activation, with the systemic effect being dependent upon the CaV1.2 channel isoform (Busquet et al., 2008; Waltereit et al., 2008; Davis and Bauer, 2012).
Beyond the major neurotransmitters, neurotrophins are currently a focus in many fields of learning and memory. A particularly well-studied neurotrophic system in the context of fear extinction is BDNF. Extinction is impaired in mice that are haploinsufficient for BDNF or carry a BDNF gene variant that also impairs extinction in human subjects (Soliman et al., 2010; Psotta et al., 2013). Virus-mediated deletion of BDNF or the BDNF trkB receptor (for receptor nomenclature see Alexander et al., 2013e) within BLA (but not prelimbic cortex) also disrupts extinction (Chhatwal et al., 2006; Choi et al., 2010). Conversely, systemic administration of a trkB receptor agonist (7,8-dihydoxyflavone) increases mPFC BDNF expression, BLA trkB activity and facilitates extinction, as does infusion of BDNF into hippocampal inputs to the infralimbic subregion (Peters et al., 2010; Andero et al., 2011; Baker-Andresen et al., 2013). Increasing brain levels of magnesium (via treatment with magnesium-l-threonate) also improves extinction in association with increased mPFC, but in this case not BLA, BDNF expression and related enhancements in mPFC synaptic plasticity (Abumaria et al., 2011). Increases in mPFC BDNF were not, however, related to the enhanced extinction produced by a cute or chronic systemic administration of an angiotensin receptor type 1 antagonist (losartan) (Marvar et al., 2014).
Increased BDNF signalling might promote extinction by increasing the number of parvalbumin-positive synapses around BLA fear neurons and thereby increasing inhibitory control of excitatory, fear-promoting, neurons (Kohara et al., 2007; Gittis et al., 2011) – a mechanism analogous to that discussed earlier involving CCK and CB1 receptors (Trouche et al., 2013). In addition to BDNF, fibroblast growth factor-2 (FGF-2), a mitogen with assorted effects on neuroplasticity, neurogenesis and various molecular signalling pathways involved in memory, is another neurotrophin acting within the mPFC–amygdala circuit to modify extinction. A series of studies by Graham, Richardson and colleagues found that FGF-2, delivered systemically or directly into the BLA, facilitates extinction (Graham and Richardson, 2009; 2010; 2011a,b).
A connection has emerged between extinction, BDNF and epigenetic changes in the mPFC–amygdala circuit. Systemic treatment with valproic acid, a commonly prescribed anticonvulsant and mood-stabilizing medication, facilitates extinction in association with increases in the mPFC expression of BDNF and an epigenetic change (increased histone H4 acetylation) around the BDNF gene promoter (Bredy et al., 2007; Bredy and Barad, 2008; Heinrichs et al., 2013; Whittle et al., 2013). Extinction-promoting effects are not limited to valproic acid, but extend to other manipulations with histone deacetylase (HDAC)-inhibiting properties, including treatment with sodium butyrate, vorinostat or trichostatin A, and dietary zinc depletion (Lattal et al., 2007; Whittle et al., 2010; Fujita et al., 2012; Matsumoto et al., 2013). Suggesting that at least some of these behavioural effects may work through the mPFC–amygdala circuit, improvements in fear extinction produced by zinc depletion correlated with the normalization of aberrant activation of multiple mPFC and amygdala subregions (Whittle et al., 2010). Of further relevance in this regard, inhibiting the activity of another transcriptional modifier of histone acetylation, the histone acetyltransferase (HAT) p300 in the infralimbic subregion (using C646 or a combined p300/cAMP-responsive element-binding protein-binding protein inhibitor, PCAF) strengthened extinction and enhanced synaptic plasticity within the infralimbic subregion (Marek et al., 2011; Wei et al., 2012).
These findings have fostered the investigation of specific subtypes of class I HDACs in extinction. Infusion of a HDAC1/HDAC3 inhibitor (MS-275) into the hippocampus (but not when given systemically; Whittle et al., 2013) disrupts extinction, while viral-mediated hippocampal overexpression HDAC1 has the opposite effect (Bahari-Javan et al., 2012). Conversely, and more akin to the effects of subtype non-specific HDAC inhibitors, extinction is facilitated by gene knockout of HDAC2 (not HDAC1) on forebrain neurons (Morris et al., 2013). Given HDAC constrains gene expression and synaptic plasticity (Sharma, 2010), the pro-extinction effects of HDAC inhibitors can be framed in terms of the release of extinction-mediating plasticity in the mPFC–amygdala circuitry. However, the finding that inhibition of HDAC or HAT does not uniformly promote extinction suggests that there are additional factors at play that have not yet been adequately illuminated (Marek et al., 2011). Clarifying these mechanisms will be important to inform the clinical potential of this interesting approach to modifying extinction (for an excellent recent review, see Whittle and Singewald, 2014.
Concluding remarks
Our goal here was to provide a comprehensive, although not exhaustive, update on a large and ever-expanding body of preclinical research that connects fear extinction with the function and dysfunction of a neural circuit comprising various regions of the mPFC and amygdala. Fear extinction deficits associated with the mPFC–amygdala circuit abnormalities observed across rodent subpopulation differences or produced by environmental, genetic or pharmacological manipulations, are summarized in Table 1. Table 2 lists pharmacologically induced rescue or facilitation of fear extinction that can be attributed to functional alterations in the mPFC or amygdala. There are good grounds to be optimistic that real progress can be made in further defining the neural basis of fear extinction, and using this knowledge base as a platform to identify plausible candidates for extinction-promoting therapeutics.
Table 1.
Category of model | Link to mPFC–amygdala circuit | Reference |
---|---|---|
Subpopulation or strain | ||
Poor extinction C57BL/6 mice | mPFC/BLA hypoactivation, prolonged mPFC long-term depression | Herry and Mons, 2004 |
Poor extinction Sprague Dawley rats | Increased prelimbic cortex neuronal firing, reduced infralimbic cortex neuronal bursting | Burgos-Robles et al., 2007; 2009 |
Poor extinction transgenic rats | Hypoactivation of infralimbic cortex inputs to BLA, hyperactivation of prelimbic cortex inputs to BLA | Knapska et al., 2012 |
129S1/SvImJ inbred strain | Infralimbic cortex/BLA/CeL hypoactivation, prelimbic cortex/CeM hyperactivation, prelimbic cortex/infralimbic cortex neuronal hyperactivity | Hefner et al., 2008; Whittle et al., 2010; Camp et al., 2012; Fitzgerald et al., 2014 |
High-anxiety behavior rats | Infralimbic cortex/BLA hypoactivity, CeM hyperactivity | Muigg et al., 2008 |
Adolescent rats and mice | Infralimbic cortex hypoactivation, impaired infralimbic cortex synaptic plasticity | Kim et al., 2011; Pattwell et al., 2012 |
Environmental insult | ||
Chronic alcohol | Infralimbic cortex dendritic hypertrophy, reduced infralimbic cortex NMDA receptor function, infralimbic cortex neuronal hypoactivity | Holmes et al., 2012 |
Forced swim stress | Infralimbic cortex dendritic hypotrophy | Izquierdo et al., 2006 |
Restraint stress | Infralimbic cortex neuronal hypoactivity, prelimbic cortex neuronal hyperactivity, BLA synaptic hyperactivity | Wilber et al., 2011; Chauveau et al., 2012 |
Elevated platform stress | BLA dendritic hypotrophy/spinogenesis, infralimbic cortex GR up-regulation | Maroun and Richter-Levin, 2003; Maroun, 2006; Deschaux et al., 2013; Maroun et al., 2013 |
Social defeat stress | Increased mPFC 2-AG levels | Dubreucq et al., 2012 |
Chronic corticosterone | mPFC NMDA/AMPA receptor down-regulation | Gourley et al., 2009 |
Prenatal stress or corticosterone | Infralimbic cortex GR down-regulation | Bingham et al., 2013 |
Maternal separation | Infralimbic cortex GR up-regulation/NMDA receptor down-regulation | Wilber et al., 2009 |
Post-natal footshock stress | Ishikawa et al., 2012 | |
Adolescent stress | Prelimbic cortex/BA hyperactivation, impaired mPFC synaptic plasticity | Judo et al., 2010; Toledo-Rodriguez et al., 2012; Saito et al., 2013 |
‘Immediate extinction effect’ | Reduced mPFC neuronal bursting, mPFC neuronal hypoactivity, prelimbic cortex hyperactivity | Chang et al., 2010; Kim et al., 2010; Stafford et al., 2013 |
Genetic manipulation | ||
Reelin haploinsufficient | Abnormal mPFC synaptic plasticity/spine density | Iafrati et al., 2013 |
5-HT transporter knockout | Infralimbic cortex dendritic hypertrophy, increased BLA spine density, increased LA-mPFC theta activity | Wellman et al., 2007; Narayanan et al., 2011; Nietzer et al., 2011 |
Pet-1 knockout | BLA dendritic hypertrophy | Wellman et al., 2013 |
GAD67 knockdown | BLA specific | Heldt et al., 2012 |
Plaur knockout | Reduced mPFC interneurons | Bissonette et al., 2014 |
Dynorphin knockout | Infralimbic cortex/BLA hypoactivation | Bilkei-Gorzo et al., 2012 |
GRP receptor knockout | mPFC hypoactivation, BLA hyperactivation and increased synaptic plasticity | Martel et al., 2012 |
TrkB knockdown | BLA specific knockdown | Chhatwal et al., 2006 |
Stathmin knockout | mPFC hyperactivation, BLA hypoactivation | Martel et al., 2012 |
Pharmacological manipulation | ||
6-hydroxdopamine | mPFC dopamine depletion | Morrow et al., 1999; Fernandez Espejo, 2003; Hugues et al., 2007; Saito et al., 2013 |
Dopamine D1 antagonist | Intra-BLA or intra-infralimbic cortex infusion | Hikind and Maroun, 2008; Fiorenza et al., 2012 |
Dopamine D2 antagonist | Intra-infralimbic cortex infusion | Mueller et al., 2010 |
Dopamine D4 antagonist | Intra-mPFC infusion | Pfeiffer and Fendt, 2006 |
Noradrenaline | Intra-BLA infusion | Berlau and McGaugh, 2006; Fiorenza et al., 2012 |
β-adrenoceptor agonist | Intra-mPFC or intra-BLA infusion | Fiorenza et al., 2012 |
β-adrenoceptor antagonist | Intra-infralimbic cortex infusion | Mueller et al., 2008 |
GABAA agonist | Intra-infralimbic cortex or intra-BLA infusion | Sierra-Mercado et al., 2006; 2011; Laurent and Westbrook, 2008; 2009a; 2010; Laurent et al., 2008; Parkes and Westbrook, 2010; Sotres-Bayon et al., 2012; Holmes and Singewald, 2013; Holmes et al., 2013 |
Benzodiazepine agonist | Intra-BLA infusion | Hart et al., 2009; 2010 |
AMPA receptor internalization inhibition | Intra-BLA infusion | Kim et al., 2007b; Dalton et al., 2008 |
mGluA1 antagonist | Intra-BLA infusion | Kim et al., 2007a |
mGluA5 antagonist | Intra-BLA infusion | Fontanez-Nuin et al., 2011; Sepulveda-Orengo et al., 2013 |
NMDA receptor antagonist | Intra-BLA or intra-mPFC infusion | Falls et al., 1992; Lee and Kim, 1998; Lin et al., 2003c; Burgos-Robles et al., 2007; Laurent and Westbrook, 2008; 2009b; Laurent et al., 2008; Zimmerman and Maren, 2010; Parkes and Westbrook, 2011; Fiorenza et al., 2012; Holmes et al., 2012 |
GluN2B antagonist | Intra-BLA or intra-mPFC infusion | Sotres-Bayon et al., 2007; 2009; Laurent and Westbrook, 2008; Laurent et al., 2008; Dalton et al., 2012 |
Glucocorticoid receptor antagonist | Intra-BLA | Yang et al., 2006 |
NPY-Y1 antagonist | Intra-BLA infusion | Gutman et al., 2008 |
NPS antagonist | Intra-BLA infusion | Jungling et al., 2008 |
KOP receptor antagonism | mPFC and BLA hypoactivation | Bilkei-Gorzo et al., 2012 |
MOP receptor ablation | ICN specific | Likhtik et al., 2008 |
Oxytocin | Intra-BLA infusion | Likhtik et al., 2008 |
CCK2 receptor agonist | BLA CCK-B receptor dependent | Chhatwal et al., 2009 |
CB1 receptor antagonist | Intra-BLA infusion | Kuhnert et al., 2013 |
H2 receptor antagonist | Intra-BLA or mPFC infusion | Fiorenza et al., 2012 |
CaV1.x antagonist | Intra-BLA infusion, BLA MAPK hypoactivation | Davis and Bauer, 2012 |
Table 2.
Pharmacological manipulation | Relevance to mPFC–amygdala circuit | Reference |
---|---|---|
L-DOPA | mPFC activation, CeM deactivation | Haaker et al., 2013 |
Noradrenaline | Intra-BLA infusion | Fiorenza et al., 2012 |
β-adrenoceptor agonist | Intra-infralimbic cortex infusion | Do Monte et al., 2010 |
5-HT re-uptake inhibition | Decreased BLA perineuronal nets | Karpova et al., 2012 |
Methylene blue | Increased mPFC activity | Wrubel et al., 2007 |
5-HT1A agonism | Increased mPFC dopamine | Saito et al., 2013 |
GABAA agonism | Intra-infralimbic cortex or intra-BLA infusion | Akirav et al., 2006 |
GABAA antagonism | Intra-infralimbic cortex or intra-BLA infusion | Berlau and McGaugh, 2006; Thompson et al., 2010; Chang and Maren, 2011; Fitzgerald et al., 2014 |
AMPA receptor agonist | Intra-mPFC or intra-BLA infusion | Zushida et al., 2007 |
mGluA7 agonism | Intra-BLA infusion | Morawska and Fendt, 2012; Dobi et al., 2013 |
mGluA5 antagonism | Intra-BLA infusion | Mao et al., 2013 |
GluN2B antagonism | Normalized mPFC synaptic plasticity | Iafrati et al., 2013 |
GluN2C/D potentiator | Intra-BLA infusion | Ogden et al., 2014 |
Glycine re-uptake inhibition | Intra-BLA infusion | Mao et al., 2009 |
NMDA receptor partial agonism | Intra-BLA infusion | Walker et al., 2002; Ledgerwood et al., 2003; Lee et al., 2006; Akirav et al., 2009; Baker et al., 2012; Toth et al., 2012a |
Ginkgo biloba extract | Intra-BLA | Yang et al., 2009 |
Glucocorticoid agonism | Intra-BLA | Yang et al., 2006 |
NPS | Intra-BLA infusion, increased ICN inputs | Jungling et al., 2008; Chauveau et al., 2012 |
Oxytocin | Intra-infralimbic cortex infusion | Likhtik et al., 2008 |
CB1 receptor agonism | Intra-BLA or intra-mPFC infusion | Lin et al., 2006; 2009b; Do Monte et al., 2013; Ganon-Elazar and Akirav, 2013 |
FAAH inhibition | Intra-BLA infusion, enhanced BLA synaptic plasticity | Gunduz-Cinar et al., 2013 |
TrkB agonism, BDNF | Increased mPFC BDNF expression, BLA TrkB activity, BDNF inputs to infralimbic cortex | Peters et al., 2010; Andero et al., 2011; Baker-Andresen et al., 2013 |
Brain magnesium increaser | Enhanced mPFC synaptic plasticity | Abumaria et al., 2011 |
FGF-2 | Intra-BLA | Graham and Richardson, 2011b |
Dietary zinc deficiency | Normalized infralimbic cortex/BLA/CeL/CeM activation | Whittle et al., 2010 |
Valproic acid | Increased mPFC BDNF expression | Bredy and Barad, 2008 |
HAT inhibition | Intra-infralimbic cortex infusion, enhanced mPFC synaptic plasticity | Marek et al., 2011 |
Histamine metabolism inhibitor | Intra-BLA or mPFC infusion | Fiorenza et al., 2012 |
Muscarinic agonism | Intra-mPFC infusion | Santini et al., 2012 |
However, despite the wealth of data that has amassed in recent years and the increasing knowledge base this provides, there remain major challenges to developing anxiolytic medications from preclinical findings in rodents. Some of the issues are common to any psychotropic drug development effort, and include the design of safe, brain-penetrant molecules with limited adverse side effects. Individual differences in treatment tolerability and efficacy, caused by genetic variation, previous medication history or sex/gender, is another concern; in fact, there is now good evidence that fear extinction in rodents and humans is strongly influenced by the oestrus/menstrual cycle and gonadal hormones because of modulation of the mPFC–amygdala circuit (Lebron-Milad and Milad, 2012). The careful dissection of the brain regions mediating fear extinction has also shown that some systems can have both extinction-facilitating and -impairing effects, depending on the region where they are acting. An example is the β-adrenoceptor, blockade of which interferes with extinction when limited to the infralimbic subregion, but promotes extinction when the BLA is targeted – hence it is difficult to predict the net effect of medicating a patient with a β-adrenceptor blocker or agonist during exposure therapy. A lesson here is that despite the availability of powerful tools for manipulation, the brain in ever more atomized ways, pharmacological studies using simple systemic treatments still have an important place in translational neuroscience. There is also room for developing new behaviour-based interventions, alongside drug treatments, to improve the effectiveness of exposure-based therapies, with promising developments in applying simple behavioural techniques to facilitate extinction (Auber et al., 2013).
Acknowledgments
We thank A.J. McDonald for critical reading of manuscript and NIAAA intramural funding for support.
Glossary
- ACC
anterior cingulate cortex
- BA
basal nucleus of the amygdala
- BLA
basolateral amygdala
- CBT
cognitive behavioural therapy
- CCK
cholecystokinin
- CeA
central nucleus of amygdala
- CeL
lateral nucleus of the central amygdala
- CeM
medial nucleus of the central amygdala
- CITZ
capsular infralimbic subregion target zone
- CRF
corticotropin-releasing factor
- FAAH
fatty acid amide hydrolase
- GluA1
AMPA receptor subunit 1
- GluA2
AMPA receptor subunit 2
- GluN2B
NMDA receptor subtype 2B
- GluN2C/D
NMDA receptor subtype 2C/D
- GRP
gastrin-releasing peptide
- HDAC
histone deacetylase
- HAT
histone acetyltransferase ICN, intercalated cell nuclei
- IN
main intercalated nucleus
- KOP
κ-opioid
- LA
lateral amygdala
- mGlu
metabotropic glutamate receptor
- mImp
medial paracapsular intercalated nucleus
- MOP
μ-opioid
- mPFC
medial prefrontal cortex
- NPS
neuropeptide S
- NPY
neuropeptide Y
- PD
panic disorder
- PEPA
2-[2,6-difluoro-4-[[2-[(phenylsulfonyl)amino]ethyl]thio]phenoxy]acetamide
- PTSD
post-traumatic stress disorder
- SERT
serotonin transporter
- SSRIs
selective serotonin re-uptake inhibitors
Conflict of interest
None.
References
- Abraham AD, Cunningham CL, Lattal KM. Methylphenidate enhances extinction of contextual fear. Learn Mem. 2012;19:67–72. doi: 10.1101/lm.024752.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abraham AD, Neve KA, Lattal KM. Dopamine and extinction: a convergence of theory with fear and reward circuitry. Neurobiol Learn Mem. 2014;108:65–77. doi: 10.1016/j.nlm.2013.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abumaria N, Yin B, Zhang L, Li XY, Chen T, Descalzi G, et al. Effects of elevation of brain magnesium on fear conditioning, fear extinction, and synaptic plasticity in the infralimbic prefrontal cortex and lateral amygdala. J Neurosci. 2011;31:14871–14881. doi: 10.1523/JNEUROSCI.3782-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Acheson D, Feifel D, de Wilde S, McKinney R, Lohr J, Risbrough V. The effect of intranasal oxytocin treatment on conditioned fear extinction and recall in a healthy human sample. Psychopharmacology (Berl) 2013;229:199–208. doi: 10.1007/s00213-013-3099-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akirav I, Raizel H, Maroun M. Enhancement of conditioned fear extinction by infusion of the GABA(A) agonist muscimol into the rat prefrontal cortex and amygdala. Eur J Neurosci. 2006;23:758–764. doi: 10.1111/j.1460-9568.2006.04603.x. [DOI] [PubMed] [Google Scholar]
- Akirav I, Segev A, Motanis H, Maroun M. D-cycloserine into the BLA reverses the impairing effects of exposure to stress on the extinction of contextual fear, but not conditioned taste aversion. Learn Mem. 2009;16:682–686. doi: 10.1101/lm.1565109. [DOI] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: G Protein-Coupled Receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: Transporters. Br J Pharmacol. 2013b;170:1706–1790. doi: 10.1111/bph.12450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: Ligand-gated ion channels. Br J Pharmacol. 2013c;170:1582–1603. doi: 10.1111/bph.12446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: Ion channels. Br J Pharmacol. 2013d;170:1607–1646. doi: 10.1111/bph.12447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: Catalytic receptors. Br J Pharmacol. 2013e;170:1676–1703. doi: 10.1111/bph.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amano T, Unal CT, Pare D. Synaptic correlates of fear extinction in the amygdala. Nat Neurosci. 2010;13:489–494. doi: 10.1038/nn.2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amano T, Duvarci S, Popa D, Pare D. The fear circuit revisited: contributions of the basal amygdala nuclei to conditioned fear. J Neurosci. 2011;31:15481–15489. doi: 10.1523/JNEUROSCI.3410-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amano T, Amir A, Goswami S, Pare D. Morphology, PKC-δ expression, and synaptic responsiveness of different types of rat central lateral amygdala neurons. J Neurophysiol. 2012;108:3196–3205. doi: 10.1152/jn.00514.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amir A, Amano T, Pare D. Physiological identification and infralimbic responsiveness of rat intercalated amygdala neurons. J Neurophysiol. 2011;105:3054–3066. doi: 10.1152/jn.00136.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ammassari-Teule M, Sgobio C, Biamonte F, Marrone C, Mercuri NB, Keller F. Reelin haploinsufficiency reduces the density of PV + neurons in circumscribed regions of the striatum and selectively alters striatal-based behaviors. Psychopharmacology (Berl) 2009;204:511–521. doi: 10.1007/s00213-009-1483-x. [DOI] [PubMed] [Google Scholar]
- Andero R, Heldt SA, Ye K, Liu X, Armario A, Ressler KJ. Effect of 7,8-dihydroxyflavone, a small-molecule TrkB agonist, on emotional learning. Am J Psychiatry. 2011;168:163–172. doi: 10.1176/appi.ajp.2010.10030326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anglada-Figueroa D, Quirk GJ. Lesions of the basal amygdala block expression of conditioned fear but not extinction. J Neurosci. 2005;25:9680–9685. doi: 10.1523/JNEUROSCI.2600-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Archbold GE, Dobbek N, Nader K. Temporal dynamics of recovery from extinction shortly after extinction acquisition. Learn Mem. 2013;20:395–398. doi: 10.1101/lm.028225.112. [DOI] [PubMed] [Google Scholar]
- Asan E. The catecholaminergic innervation of the rat amygdala. Adv Anat Embryol Cell Biol. 1998;142:1–118. doi: 10.1007/978-3-642-72085-7. [DOI] [PubMed] [Google Scholar]
- Auber A, Tedesco V, Jones CE, Monfils MH, Chiamulera C. Post-retrieval extinction as reconsolidation interference: methodological issues or boundary conditions? Psychopharmacology (Berl) 2013;226:631–647. doi: 10.1007/s00213-013-3004-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baganz N, Horton R, Martin K, Holmes A, Daws LC. Repeated swim impairs serotonin clearance via a corticosterone-sensitive mechanism: organic cation transporter 3, the smoking gun. J Neurosci. 2011;30:15185–15195. doi: 10.1523/JNEUROSCI.2740-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bahari-Javan S, Maddalena A, Kerimoglu C, Wittnam J, Held T, Bahr M, et al. HDAC1 regulates fear extinction in mice. J Neurosci. 2012;32:5062–5073. doi: 10.1523/JNEUROSCI.0079-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baker JD, Azorlosa JL. The NMDA antagonist MK-801 blocks the extinction of Pavlovian fear conditioning. Behav Neurosci. 1996;110:618–620. doi: 10.1037//0735-7044.110.3.618. [DOI] [PubMed] [Google Scholar]
- Baker KD, McNally GP, Richardson R. D-cycloserine does not facilitate fear extinction by reducing conditioned stimulus processing or promoting conditioned inhibition to contextual cues. Learn Mem. 2012;19:461–469. doi: 10.1101/lm.026674.112. [DOI] [PubMed] [Google Scholar]
- Baker-Andresen D, Flavell CR, Li X, Bredy TW. Activation of BDNF signaling prevents the return of fear in female mice. Learn Mem. 2013;20:237–240. doi: 10.1101/lm.029520.112. [DOI] [PubMed] [Google Scholar]
- Barlow DH, Gorman JM, Shear MK, Woods SW. Cognitive-behavioral therapy, imipramine, or their combination for panic disorder: a randomized controlled trial. JAMA. 2000;283:2529–2536. doi: 10.1001/jama.283.19.2529. [DOI] [PubMed] [Google Scholar]
- Berlau DJ, McGaugh JL. Enhancement of extinction memory consolidation: the role of the noradrenergic and GABAergic systems within the basolateral amygdala. Neurobiol Learn Mem. 2006;86:123–132. doi: 10.1016/j.nlm.2005.12.008. [DOI] [PubMed] [Google Scholar]
- Berretta S, Pantazopoulos H, Caldera M, Pantazopoulos P, Pare D. Infralimbic cortex activation increases c-Fos expression in intercalated neurons of the amygdala. Neuroscience. 2005;132:943–953. doi: 10.1016/j.neuroscience.2005.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertotto ME, Bustos SG, Molina VA, Martijena ID. Influence of ethanol withdrawal on fear memory: effect of D-cycloserine. Neuroscience. 2006;142:979–990. doi: 10.1016/j.neuroscience.2006.07.013. [DOI] [PubMed] [Google Scholar]
- Bilkei-Gorzo A, Erk S, Schurmann B, Mauer D, Michel K, Boecker H, et al. Dynorphins regulate fear memory: from mice to men. J Neurosci. 2012;32:9335–9343. doi: 10.1523/JNEUROSCI.1034-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bingham BC, Sheela Rani CS, Frazer A, Strong R, Morilak DA. Exogenous prenatal corticosterone exposure mimics the effects of prenatal stress on adult brain stress response systems and fear extinction behavior. Psychoneuroendocrinology. 2013;38:2746–2757. doi: 10.1016/j.psyneuen.2013.07.003. [DOI] [PubMed] [Google Scholar]
- Bissiere S, Humeau Y, Luthi A. Dopamine gates LTP induction in lateral amygdala by suppressing feedforward inhibition. Nat Neurosci. 2003;6:587–592. doi: 10.1038/nn1058. [DOI] [PubMed] [Google Scholar]
- Bissonette GB, Bae MH, Suresh T, Jaffe DE, Powell EM. Prefrontal cognitive deficits in mice with altered cerebral cortical GABAergic interneurons. Behav Brain Res. 2014;259:143–151. doi: 10.1016/j.bbr.2013.10.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bitencourt RM, Pamplona FA, Takahashi RN. Facilitation of contextual fear memory extinction and anti-anxiogenic effects of AM404 and cannabidiol in conditioned rats. Eur Neuropsychopharmacol. 2008;18:849–859. doi: 10.1016/j.euroneuro.2008.07.001. [DOI] [PubMed] [Google Scholar]
- Blundell J, Blaiss CA, Lagace DC, Eisch AJ, Powell CM. Block of glucocorticoid synthesis during re-activation inhibits extinction of an established fear memory. Neurobiol Learn Mem. 2011;95:453–460. doi: 10.1016/j.nlm.2011.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bontempi B, Laurent-Demir C, Destrade C, Jaffard R. Time-dependent reorganization of brain circuitry underlying long-term memory storage. Nature. 1999;400:671–675. doi: 10.1038/23270. [DOI] [PubMed] [Google Scholar]
- Borowski TB, Kokkinidis L. The effects of cocaine, amphetamine, and the dopamine D1 receptor agonist SKF 38393 on fear extinction as measured with potentiated startle: implications for psychomotor stimulant psychosis. Behav Neurosci. 1998;112:952–965. doi: 10.1037//0735-7044.112.4.952. [DOI] [PubMed] [Google Scholar]
- Bos MG, Beckers T, Kindt M. The effects of noradrenergic blockade on extinction in humans. Biol Psychol. 2012;89:598–605. doi: 10.1016/j.biopsycho.2012.01.007. [DOI] [PubMed] [Google Scholar]
- Bouton ME, Kenney FA, Rosengard C. State-dependent fear extinction with two benzodiazepine tranquilizers. Behav Neurosci. 1990;104:44–55. doi: 10.1037//0735-7044.104.1.44. [DOI] [PubMed] [Google Scholar]
- Bouton ME, Vurbic D, Woods AM. D-cycloserine facilitates context-specific fear extinction learning. Neurobiol Learn Mem. 2008;90:504–510. doi: 10.1016/j.nlm.2008.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bredy TW, Barad M. The histone deacetylase inhibitor valproic acid enhances acquisition, extinction, and reconsolidation of conditioned fear. Learn Mem. 2008;15:39–45. doi: 10.1101/lm.801108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bredy TW, Wu H, Crego C, Zellhoefer J, Sun YE, Barad M. Histone modifications around individual BDNF gene promoters in prefrontal cortex are associated with extinction of conditioned fear. Learn Mem. 2007;14:268–276. doi: 10.1101/lm.500907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brinks V, de Kloet ER, Oitzl MS. Corticosterone facilitates extinction of fear memory in BALB/c mice but strengthens cue related fear in C57BL/6 mice. Exp Neurol. 2009;216:375–382. doi: 10.1016/j.expneurol.2008.12.011. [DOI] [PubMed] [Google Scholar]
- Brinley-Reed M, McDonald AJ. Evidence that dopaminergic axons provide a dense innervation of specific neuronal subpopulations in the rat basolateral amygdala. Brain Res. 1999;850:127–135. doi: 10.1016/s0006-8993(99)02112-5. [DOI] [PubMed] [Google Scholar]
- Bui E, Orr SP, Jacoby RJ, Keshaviah A, LeBlanc NJ, Milad MR, et al. Two weeks of pretreatment with escitalopram facilitates extinction learning in healthy individuals. Hum Psychopharmacol. 2013;28:447–456. doi: 10.1002/hup.2330. [DOI] [PubMed] [Google Scholar]
- Burghardt NS, Bauer EP. Acute and chronic effects of selective serotonin reuptake inhibitor treatment on fear conditioning: implications for underlying fear circuits. Neuroscience. 2013;247:253–272. doi: 10.1016/j.neuroscience.2013.05.050. [DOI] [PubMed] [Google Scholar]
- Burghardt NS, Sigurdsson T, Gorman JM, McEwen BS, LeDoux JE. Chronic antidepressant treatment impairs the acquisition of fear extinction. Biol Psychiatry. 2013;73:1078–1086. doi: 10.1016/j.biopsych.2012.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burgos-Robles A, Vidal-Gonzalez I, Santini E, Quirk GJ. Consolidation of fear extinction requires NMDA receptor-dependent bursting in the ventromedial prefrontal cortex. Neuron. 2007;53:871–880. doi: 10.1016/j.neuron.2007.02.021. [DOI] [PubMed] [Google Scholar]
- Burgos-Robles A, Vidal-Gonzalez I, Quirk GJ. Sustained conditioned responses in prelimbic prefrontal neurons are correlated with fear expression and extinction failure. J Neurosci. 2009;29:8474–8482. doi: 10.1523/JNEUROSCI.0378-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busquet P, Hetzenauer A, Sinnegger-Brauns MJ, Striessnig J, Singewald N. Role of L-type Ca2+ channel isoforms in the extinction of conditioned fear. Learn Mem. 2008;15:378–386. doi: 10.1101/lm.886208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busti D, Geracitano R, Whittle N, Dalezios Y, Manko M, Kaufmann W, et al. Different fear states engage distinct networks within the intercalated cell clusters of the amygdala. J Neurosci. 2011;31:5131–5144. doi: 10.1523/JNEUROSCI.6100-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bustos SG, Maldonado H, Molina VA. Disruptive effect of midazolam on fear memory reconsolidation: decisive influence of reactivation time span and memory age. Neuropsychopharmacology. 2009;34:446–457. doi: 10.1038/npp.2008.75. [DOI] [PubMed] [Google Scholar]
- Cai WH, Blundell J, Han J, Greene RW, Powell CM. Postreactivation glucocorticoids impair recall of established fear memory. J Neurosci. 2006;26:9560–9566. doi: 10.1523/JNEUROSCI.2397-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cain CK, Blouin AM, Barad M. Adrenergic transmission facilitates extinction of conditional fear in mice. Learn Mem. 2004;11:179–187. doi: 10.1101/lm.71504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callaerts-Vegh Z, Beckers T, Ball SM, Baeyens F, Callaerts PF, Cryan JF, et al. Concomitant deficits in working memory and fear extinction are functionally dissociated from reduced anxiety in metabotropic glutamate receptor 7-deficient mice. J Neurosci. 2006;26:6573–6582. doi: 10.1523/JNEUROSCI.1497-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callaghan BL, Richardson R. The effect of adverse rearing environments on persistent memories in young rats: removing the brakes on infant fear memories. Transl Psychiatry. 2012;2:e138. doi: 10.1038/tp.2012.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callaghan BL, Graham BM, Li S, Richardson R. From resilience to vulnerability: mechanistic insights into the effects of stress on transitions in critical period plasticity. Front Psychiatry. 2013;4:90. doi: 10.3389/fpsyt.2013.00090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camp MC, Macpherson KP, Lederle L, Graybeal C, Gaburro S, Debrouse LM, et al. Genetic strain differences in learned fear inhibition associated with variation in neuroendocrine, autonomic, and amygdala dendritic phenotypes. Neuropsychopharmacology. 2012;37:1534–1547. doi: 10.1038/npp.2011.340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cannich A, Wotjak CT, Kamprath K, Hermann H, Lutz B, Marsicano G. CB1 cannabinoid receptors modulate kinase and phosphatase activity during extinction of conditioned fear in mice. Learn Mem. 2004;11:625–632. doi: 10.1101/lm.77904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carmack SA, Wood SC, Anagnostaras SG. Amphetamine and extinction of cued fear. Neurosci Lett. 2010;468:18–22. doi: 10.1016/j.neulet.2009.10.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caspi A, Hariri AR, Holmes A, Uher R, Moffitt TE. Genetic sensitivity to the environment: the case of the serotonin transporter gene and its implications for studying complex diseases and traits. Am J Psychiatry. 2010;167:509–527. doi: 10.1176/appi.ajp.2010.09101452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cassell MD, Wright DJ. Topography of projections from the medial prefrontal cortex to the amygdala in the rat. Brain Res Bull. 1986;17:321–333. doi: 10.1016/0361-9230(86)90237-6. [DOI] [PubMed] [Google Scholar]
- Chan T, Kyere K, Davis BR, Shemyakin A, Kabitzke PA, Shair HN, et al. The role of the medial prefrontal cortex in innate fear regulation in infants, juveniles, and adolescents. J Neurosci. 2011;31:4991–4999. doi: 10.1523/JNEUROSCI.5216-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan WY, McNally GP. Conditioned stimulus familiarity determines effects of MK-801 on fear extinction. Behav Neurosci. 2009;123:303–314. doi: 10.1037/a0014988. [DOI] [PubMed] [Google Scholar]
- Chang CH, Maren S. Medial prefrontal cortex activation facilitates re-extinction of fear in rats. Learn Mem. 2011;18:221–225. doi: 10.1101/lm.2070111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang CH, Berke JD, Maren S. Single-unit activity in the medial prefrontal cortex during immediate and delayed extinction of fear in rats. PLoS ONE. 2010;5:e11971. doi: 10.1371/journal.pone.0011971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chaperon F, Fendt M, Kelly PH, Lingenhoehl K, Mosbacher J, Olpe HR, et al. Gastrin-releasing peptide signaling plays a limited and subtle role in amygdala physiology and aversive memory. PLoS ONE. 2012;7:e34963. doi: 10.1371/journal.pone.0034963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chauveau F, Lange MD, Jungling K, Lesting J, Seidenbecher T, Pape HC. Prevention of stress-impaired fear extinction through neuropeptide S action in the lateral amygdala. Neuropsychopharmacology. 2012;37:1588–1599. doi: 10.1038/npp.2012.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chhatwal JP, Davis M, Maguschak KA, Ressler KJ. Enhancing cannabinoid neurotransmission augments the extinction of conditioned fear. Neuropsychopharmacology. 2005a;30:516–524. doi: 10.1038/sj.npp.1300655. [DOI] [PubMed] [Google Scholar]
- Chhatwal JP, Myers KM, Ressler KJ, Davis M. Regulation of gephyrin and GABAA receptor binding within the amygdala after fear acquisition and extinction. J Neurosci. 2005b;25:502–506. doi: 10.1523/JNEUROSCI.3301-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chhatwal JP, Stanek-Rattiner L, Davis M, Ressler KJ. Amygdala BDNF signaling is required for consolidation but not encoding of extinction. Nat Neurosci. 2006;9:870–872. doi: 10.1038/nn1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chhatwal JP, Gutman AR, Maguschak KA, Bowser ME, Yang Y, Davis M, et al. Functional interactions between endocannabinoid and CCK neurotransmitter systems may be critical for extinction learning. Neuropsychopharmacology. 2009;34:509–521. doi: 10.1038/npp.2008.97. [DOI] [PubMed] [Google Scholar]
- Cho JH, Deisseroth K, Bolshakov VY. Synaptic encoding of fear extinction in mPFC–amygdala circuits. Neuron. 2013;80:1491–1507. doi: 10.1016/j.neuron.2013.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi DC, Maguschak KA, Ye K, Jang SW, Myers KM, Ressler KJ. Prelimbic cortical BDNF is required for memory of learned fear but not extinction or innate fear. Proc Natl Acad Sci U S A. 2010;107:2675–2680. doi: 10.1073/pnas.0909359107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ciocchi S, Herry C, Grenier F, Wolff SB, Letzkus JJ, Vlachos I, et al. Encoding of conditioned fear in central amygdala inhibitory circuits. Nature. 2010;468:277–282. doi: 10.1038/nature09559. [DOI] [PubMed] [Google Scholar]
- Cole S, Richardson R, McNally GP. Kappa opioid receptors mediate where fear is expressed following extinction training. Learn Mem. 2011;18:88–95. doi: 10.1101/lm.2049511. [DOI] [PubMed] [Google Scholar]
- Cole S, Richardson R, McNally GP. Ventral hippocampal kappa opioid receptors mediate the renewal of fear following extinction in the rat. PLoS ONE. 2013;8:e58701. doi: 10.1371/journal.pone.0058701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conde F, Maire-Lepoivre E, Audinat E, Crepel F. Afferent connections of the medial frontal cortex of the rat. II. Cortical and subcortical afferents. J Comp Neurol. 1995;352:567–593. doi: 10.1002/cne.903520407. [DOI] [PubMed] [Google Scholar]
- Courtin J, Bienvenu TC, Einarsson EO, Herry C. Medial prefrontal cortex neuronal circuits in fear behavior. Neuroscience. 2013;240:219–242. doi: 10.1016/j.neuroscience.2013.03.001. [DOI] [PubMed] [Google Scholar]
- Courtin J, Chaudun F, Rozeske RR, Karalis N, Gonzalez-Campo C, Wurtz H, et al. Prefrontal parvalbumin interneurons shape neuronal activity to drive fear expression. Nature. 2014a;505:92–96. doi: 10.1038/nature12755. [DOI] [PubMed] [Google Scholar]
- Courtin J, Karalis N, Gonzalez-Campo C, Wurtz H, Herry C. Persistence of amygdala gamma oscillations during extinction learning predicts spontaneous fear recovery. Neurobiol Learn Mem. 2014b;113:82–89. doi: 10.1016/j.nlm.2013.09.015. [DOI] [PubMed] [Google Scholar]
- Cowan CS, Callaghan BL, Richardson R. Acute early-life stress results in premature emergence of adult-like fear retention and extinction relapse in infant rats. Behav Neurosci. 2013;127:703–711. doi: 10.1037/a0034118. [DOI] [PubMed] [Google Scholar]
- Dalton GL, Wang YT, Floresco SB, Phillips AG. Disruption of AMPA receptor endocytosis impairs the extinction, but not acquisition of learned fear. Neuropsychopharmacology. 2008;33:2416–2426. doi: 10.1038/sj.npp.1301642. [DOI] [PubMed] [Google Scholar]
- Dalton GL, Wu DC, Wang YT, Floresco SB, Phillips AG. NMDA GluN2A and GluN2B receptors play separate roles in the induction of LTP and LTD in the amygdala and in the acquisition and extinction of conditioned fear. Neuropharmacology. 2012;62:797–806. doi: 10.1016/j.neuropharm.2011.09.001. [DOI] [PubMed] [Google Scholar]
- Das RK, Kamboj SK, Ramadas M, Yogan K, Gupta V, Redman E, et al. Cannabidiol enhances consolidation of explicit fear extinction in humans. Psychopharmacology (Berl) 2013;226:781–792. doi: 10.1007/s00213-012-2955-y. [DOI] [PubMed] [Google Scholar]
- Davis AR, Shields AD, Brigman JL, Norcross M, McElligott ZA, Holmes A, et al. Yohimbine impairs extinction of cocaine-conditioned place preference in an α2-adrenergic receptor independent process. Learn Mem. 2008;15:667–676. doi: 10.1101/lm.1079308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis SE, Bauer EP. L-type voltage-gated calcium channels in the basolateral amygdala are necessary for fear extinction. J Neurosci. 2012;32:13582–13586. doi: 10.1523/JNEUROSCI.0809-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Debiec J, Bush DE, LeDoux JE. Noradrenergic enhancement of reconsolidation in the amygdala impairs extinction of conditioned fear in rats–a possible mechanism for the persistence of traumatic memories in PTSD. Depress Anxiety. 2011;28:186–193. doi: 10.1002/da.20803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deschaux O, Spennato G, Moreau JL, Garcia R. Chronic treatment with fluoxetine prevents the return of extinguished auditory-cued conditioned fear. Psychopharmacology (Berl) 2011;215:231–237. doi: 10.1007/s00213-010-2134-y. [DOI] [PubMed] [Google Scholar]
- Deschaux O, Zheng X, Lavigne J, Nachon O, Cleren C, Moreau JL, et al. Post-extinction fluoxetine treatment prevents stress-induced reemergence of extinguished fear. Psychopharmacology (Berl) 2013;225:209–216. doi: 10.1007/s00213-012-2806-x. [DOI] [PubMed] [Google Scholar]
- Dilgen J, Tejeda HA, O’Donnell P. Amygdala inputs drive feedforward inhibition in the medial prefrontal cortex. J Neurophysiol. 2013;110:221–229. doi: 10.1152/jn.00531.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Do Monte FH, Kincheski GC, Pavesi E, Sordi R, Assreuy J, Carobrez AP. Role of beta-adrenergic receptors in the ventromedial prefrontal cortex during contextual fear extinction in rats. Neurobiol Learn Mem. 2010;94:318–328. doi: 10.1016/j.nlm.2010.07.004. [DOI] [PubMed] [Google Scholar]
- Do Monte FH, Souza RR, Bitencourt RM, Kroon JA, Takahashi RN. Infusion of cannabidiol into infralimbic cortex facilitates fear extinction via CB1 receptors. Behav Brain Res. 2013;250:23–27. doi: 10.1016/j.bbr.2013.04.045. [DOI] [PubMed] [Google Scholar]
- Dobi A, Sartori SB, Busti D, Van der Putten H, Singewald N, Shigemoto R, et al. Neural substrates for the distinct effects of presynaptic group III metabotropic glutamate receptors on extinction of contextual fear conditioning in mice. Neuropharmacology. 2013;66:274–289. doi: 10.1016/j.neuropharm.2012.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DSM-5. Diagnostic and Statistical Manual of Mental Disorders. 4th edn. Washington, DC: APA Press; 2013. [Google Scholar]
- Dubreucq S, Matias I, Cardinal P, Haring M, Lutz B, Marsicano G, et al. Genetic dissection of the role of cannabinoid type-1 receptors in the emotional consequences of repeated social stress in mice. Neuropsychopharmacology. 2012;37:1885–1900. doi: 10.1038/npp.2012.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duvarci S, Popa D, Pare D. Central amygdala activity during fear conditioning. J Neurosci. 2011;31:289–294. doi: 10.1523/JNEUROSCI.4985-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehrlich I, Humeau Y, Grenier F, Ciocchi S, Herry C, Luthi A. Amygdala inhibitory circuits and the control of fear memory. Neuron. 2009;62:757–771. doi: 10.1016/j.neuron.2009.05.026. [DOI] [PubMed] [Google Scholar]
- El-Ghundi M, O’Dowd BF, George SR. Prolonged fear responses in mice lacking dopamine D1 receptor. Brain Res. 2001;892:86–93. doi: 10.1016/s0006-8993(00)03234-0. [DOI] [PubMed] [Google Scholar]
- Falls WA, Miserendino MJ, Davis M. Extinction of fear-potentiated startle: blockade by infusion of an NMDA antagonist into the amygdala. J Neurosci. 1992;12:854–863. doi: 10.1523/JNEUROSCI.12-03-00854.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farrell MR, Sayed JA, Underwood AR, Wellman CL. Lesion of infralimbic cortex occludes stress effects on retrieval of extinction but not fear conditioning. Neurobiol Learn Mem. 2010;94:240–246. doi: 10.1016/j.nlm.2010.06.001. [DOI] [PubMed] [Google Scholar]
- Fendt M, Schmid S, Thakker DR, Jacobson LH, Yamamoto R, Mitsukawa K, et al. mGluR7 facilitates extinction of aversive memories and controls amygdala plasticity. Mol Psychiatry. 2008;13:970–979. doi: 10.1038/sj.mp.4002073. [DOI] [PubMed] [Google Scholar]
- Fenton GE, Pollard AK, Halliday DM, Mason R, Bredy TW, Stevenson CW. Persistent prelimbic cortex activity contributes to enhanced learned fear expression in females. Learn Mem. 2014;21:55–60. doi: 10.1101/lm.033514.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernandez Espejo E. Prefrontocortical dopamine loss in rats delays long-term extinction of contextual conditioned fear, and reduces social interaction without affecting short-term social interaction memory. Neuropsychopharmacology. 2003;28:490–498. doi: 10.1038/sj.npp.1300066. [DOI] [PubMed] [Google Scholar]
- Fiorenza NG, Rosa J, Izquierdo I, Myskiw JC. Modulation of the extinction of two different fear-motivated tasks in three distinct brain areas. Behav Brain Res. 2012;232:210–216. doi: 10.1016/j.bbr.2012.04.015. [DOI] [PubMed] [Google Scholar]
- Fitzgerald PJ, Whittle N, Flynn SM, Graybeal C, Pinard C, Gunduz-Cinar O, et al. Prefrontal single-unit firing associated with deficient extinction in mice. Neurobiol Learn Mem. 2014;113:69–81. doi: 10.1016/j.nlm.2013.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Floresco SB, Tse MT. Dopaminergic regulation of inhibitory and excitatory transmission in the basolateral amygdala–prefrontal cortical pathway. J Neurosci. 2007;27:2045–2057. doi: 10.1523/JNEUROSCI.5474-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fontanez-Nuin DE, Santini E, Quirk GJ, Porter JT. Memory for fear extinction requires mGluR5-mediated activation of infralimbic neurons. Cereb Cortex. 2011;21:727–735. doi: 10.1093/cercor/bhq147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Freedman LJ, Insel TR, Smith Y. Subcortical projections of area 25 (subgenual cortex) of the macaque monkey. J Comp Neurol. 2000;421:172–188. [PubMed] [Google Scholar]
- Fujita Y, Morinobu S, Takei S, Fuchikami M, Matsumoto T, Yamamoto S, et al. Vorinostat, a histone deacetylase inhibitor, facilitates fear extinction and enhances expression of the hippocampal NR2B-containing NMDA receptor gene. J Psychiatr Res. 2012;46:635–643. doi: 10.1016/j.jpsychires.2012.01.026. [DOI] [PubMed] [Google Scholar]
- Fuxe K, Jacobsen KX, Hoistad M, Tinner B, Jansson A, Staines WA, et al. The dopamine D1 receptor-rich main and paracapsular intercalated nerve cell groups of the rat amygdala: relationship to the dopamine innervation. Neuroscience. 2003;119:733–746. doi: 10.1016/s0306-4522(03)00148-9. [DOI] [PubMed] [Google Scholar]
- Gabbott PL, Warner TA, Jays PR, Salway P, Busby SJ. Prefrontal cortex in the rat: projections to subcortical autonomic, motor, and limbic centers. J Comp Neurol. 2005;492:145–177. doi: 10.1002/cne.20738. [DOI] [PubMed] [Google Scholar]
- Gabbott PL, Warner TA, Busby SJ. Amygdala input monosynaptically innervates parvalbumin immunoreactive local circuit neurons in rat medial prefrontal cortex. Neuroscience. 2006;139:1039–1048. doi: 10.1016/j.neuroscience.2006.01.026. [DOI] [PubMed] [Google Scholar]
- Gafford GM, Guo JD, Flandreau EI, Hazra R, Rainnie DG, Ressler KJ. Cell-type specific deletion of GABA(A)α1 in corticotropin-releasing factor-containing neurons enhances anxiety and disrupts fear extinction. Proc Natl Acad Sci U S A. 2012;109:16330–16335. doi: 10.1073/pnas.1119261109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganon-Elazar E, Akirav I. Cannabinoids and traumatic stress modulation of contextual fear extinction and GR expression in the amygdala-hippocampal-prefrontal circuit. Psychoneuroendocrinology. 2013;38:1675–1687. doi: 10.1016/j.psyneuen.2013.01.014. [DOI] [PubMed] [Google Scholar]
- Geracitano R, Kaufmann WA, Szabo G, Ferraguti F, Capogna M. Synaptic heterogeneity between mouse paracapsular intercalated neurons of the amygdala. J Physiol. 2007;585:117–134. doi: 10.1113/jphysiol.2007.142570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geracitano R, Fischer D, Kasugai Y, Ferraguti F, Capogna M. Functional expression of the GABA(A) receptor α2 and α3 subunits at synapses between intercalated medial paracapsular neurons of mouse amygdala. Front Neural Circuits. 2012;6:32. doi: 10.3389/fncir.2012.00032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gittis AH, Hang GB, LaDow ES, Shoenfeld LR, Atallah BV, Finkbeiner S, et al. Rapid target-specific remodeling of fast-spiking inhibitory circuits after loss of dopamine. Neuron. 2011;71:858–868. doi: 10.1016/j.neuron.2011.06.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goddyn H, Callaerts-Vegh Z, Stroobants S, Dirikx T, Vansteenwegen D, Hermans D, et al. Deficits in acquisition and extinction of conditioned responses in mGluR7 knockout mice. Neurobiol Learn Mem. 2008;90:103–111. doi: 10.1016/j.nlm.2008.01.001. [DOI] [PubMed] [Google Scholar]
- Gogolla N, Caroni P, Luthi A, Herry C. Perineuronal nets protect fear memories from erasure. Science. 2009;325:1258–1261. doi: 10.1126/science.1174146. [DOI] [PubMed] [Google Scholar]
- Goldman MS. Effect of chlordiazepoxide administered early in extinction on subsequent extinction of a conditioned emotional response in rats: implications for human clinical use. Psychol Rep. 1977;40:783–786. doi: 10.2466/pr0.1977.40.3.783. [DOI] [PubMed] [Google Scholar]
- Gonzalez-Lima F, Bruchey AK. Extinction memory improvement by the metabolic enhancer methylene blue. Learn Mem. 2004;11:633–640. doi: 10.1101/lm.82404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goshen I, Brodsky M, Prakash R, Wallace J, Gradinaru V, Ramakrishnan C, et al. Dynamics of retrieval strategies for remote memories. Cell. 2011;147:678–689. doi: 10.1016/j.cell.2011.09.033. [DOI] [PubMed] [Google Scholar]
- Goswami S, Cascardi M, Rodriguez-Sierra OE, Duvarci S, Pare D. Impact of predatory threat on fear extinction in Lewis rats. Learn Mem. 2010;17:494–501. doi: 10.1101/lm.1948910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gourley SL, Kedves AT, Olausson P, Taylor JR. A history of corticosterone exposure regulates fear extinction and cortical NR2B, GluR2/3, and BDNF. Neuropsychopharmacology. 2009;34:707–716. doi: 10.1038/npp.2008.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graham BM, Richardson R. Acute systemic fibroblast growth factor-2 enhances long-term extinction of fear and reduces reinstatement in rats. Neuropsychopharmacology. 2009;34:1875–1882. doi: 10.1038/npp.2009.14. [DOI] [PubMed] [Google Scholar]
- Graham BM, Richardson R. Fibroblast growth factor-2 enhances extinction and reduces renewal of conditioned fear. Neuropsychopharmacology. 2010;35:1348–1355. doi: 10.1038/npp.2010.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graham BM, Richardson R. Fibroblast growth factor-2 alters the nature of extinction. Learn Mem. 2011a;18:80–84. doi: 10.1101/lm.2006511. [DOI] [PubMed] [Google Scholar]
- Graham BM, Richardson R. Intraamygdala infusion of fibroblast growth factor 2 enhances extinction and reduces renewal and reinstatement in adult rats. J Neurosci. 2011b;31:14151–14157. doi: 10.1523/JNEUROSCI.3014-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graham BM, Langton JM, Richardson R. Pharmacological enhancement of fear reduction: preclinical models. Br J Pharmacol. 2011;164:1230–1247. doi: 10.1111/j.1476-5381.2010.01175.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green MK, Rani CS, Joshi A, Soto-Pina AE, Martinez PA, Frazer A, et al. Prenatal stress induces long term stress vulnerability, compromising stress response systems in the brain and impairing extinction of conditioned fear after adult stress. Neuroscience. 2011;192:438–451. doi: 10.1016/j.neuroscience.2011.06.041. [DOI] [PubMed] [Google Scholar]
- Gunduz-Cinar O, Macpherson KP, Cinar R, Gamble-George J, Sugden K, Williams B, et al. Convergent translational evidence of a role for anandamide in amygdala-mediated fear extinction, threat processing and stress-reactivity. Mol Psychiatry. 2013;18:813–823. doi: 10.1038/mp.2012.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gutman AR, Yang Y, Ressler KJ, Davis M. The role of neuropeptide Y in the expression and extinction of fear-potentiated startle. J Neurosci. 2008;28:12682–12690. doi: 10.1523/JNEUROSCI.2305-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haaker J, Gaburro S, Sah A, Gartmann N, Lonsdorf TB, Meier K, et al. Single dose of L-DOPA makes extinction memories context-independent and prevents the return of fear. Proc Natl Acad Sci U S A. 2013;110:E2428–E2436. doi: 10.1073/pnas.1303061110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harris JA, Westbrook RF. Evidence that GABA transmission mediates context-specific extinction of learned fear. Psychopharmacology (Berl) 1998;140:105–115. doi: 10.1007/s002130050745. [DOI] [PubMed] [Google Scholar]
- Hart G, Harris JA, Westbrook RF. Systemic or intra-amygdala injection of a benzodiazepine (midazolam) impairs extinction but spares re-extinction of conditioned fear responses. Learn Mem. 2009;16:53–61. doi: 10.1101/lm.1154409. [DOI] [PubMed] [Google Scholar]
- Hart G, Harris JA, Westbrook RF. Systemic or intra-amygdala infusion of the benzodiazepine, midazolam, impairs learning, but facilitates re-learning to inhibit fear responses in extinction. Learn Mem. 2010;17:210–220. doi: 10.1101/lm.1682410. [DOI] [PubMed] [Google Scholar]
- Hartley CA, McKenna MC, Salman R, Holmes A, Casey BJ, Phelps EA, et al. Serotonin transporter polyadenylation polymorphism modulates the retention of fear extinction memory. Proc Natl Acad Sci U S A. 2012;109:5493–5498. doi: 10.1073/pnas.1202044109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haubensak W, Kunwar PS, Cai H, Ciocchi S, Wall NR, Ponnusamy R, et al. Genetic dissection of an amygdala microcircuit that gates conditioned fear. Nature. 2010;468:270–276. doi: 10.1038/nature09553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Havekes R, Nijholt IM, Visser AK, Eisel UL, Van der Zee EA. Transgenic inhibition of neuronal calcineurin activity in the forebrain facilitates fear conditioning, but inhibits the extinction of contextual fear memories. Neurobiol Learn Mem. 2008;89:595–598. doi: 10.1016/j.nlm.2007.08.003. [DOI] [PubMed] [Google Scholar]
- Heaney CF, Bolton MM, Murtishaw AS, Sabbagh JJ, Magcalas CM, Kinney JW. Baclofen administration alters fear extinction and GABAergic protein levels. Neurobiol Learn Mem. 2012;98:261–271. doi: 10.1016/j.nlm.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hefner K, Whittle N, Juhasz J, Norcross M, Karlsson RM, Saksida LM, et al. Impaired fear extinction learning and cortico-amygdala circuit abnormalities in a common genetic mouse strain. J Neurosci. 2008;28:8074–8085. doi: 10.1523/JNEUROSCI.4904-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heinrichs SC, Leite-Morris KA, Rasmusson AM, Kaplan GB. Repeated valproate treatment facilitates fear extinction under specific stimulus conditions. Neurosci Lett. 2013;552:108–113. doi: 10.1016/j.neulet.2013.07.035. [DOI] [PubMed] [Google Scholar]
- Heinz A, Braus DF, Smolka MN, Wrase J, Puls I, Hermann D, et al. Amygdala–prefrontal coupling depends on a genetic variation of the serotonin transporter. Nat Neurosci. 2005;8:20–21. doi: 10.1038/nn1366. [DOI] [PubMed] [Google Scholar]
- Heldt SA, Ressler KJ. Training-induced changes in the expression of GABAA-associated genes in the amygdala after the acquisition and extinction of Pavlovian fear. Eur J Neurosci. 2007;26:3631–3644. doi: 10.1111/j.1460-9568.2007.05970.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heldt SA, Mou L, Ressler KJ. In vivo knockdown of GAD67 in the amygdala disrupts fear extinction and the anxiolytic-like effect of diazepam in mice. Transl Psychiatry. 2012;2:e181. doi: 10.1038/tp.2012.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herry C, Mons N. Resistance to extinction is associated with impaired immediate early gene induction in medial prefrontal cortex and amygdala. Eur J Neurosci. 2004;20:781–790. doi: 10.1111/j.1460-9568.2004.03542.x. [DOI] [PubMed] [Google Scholar]
- Herry C, Ciocchi S, Senn V, Demmou L, Muller C, Luthi A. Switching on and off fear by distinct neuronal circuits. Nature. 2008;454:600–606. doi: 10.1038/nature07166. [DOI] [PubMed] [Google Scholar]
- Herry C, Ferraguti F, Singewald N, Letzkus JJ, Ehrlich I, Luthi A. Neuronal circuits of fear extinction. Eur J Neurosci. 2010;31:599–612. doi: 10.1111/j.1460-9568.2010.07101.x. [DOI] [PubMed] [Google Scholar]
- Hikind N, Maroun M. Microinfusion of the D1 receptor antagonist, SCH23390 into the IL but not the BLA impairs consolidation of extinction of auditory fear conditioning. Neurobiol Learn Mem. 2008;90:217–222. doi: 10.1016/j.nlm.2008.03.003. [DOI] [PubMed] [Google Scholar]
- Hill JE, Gasser PJ. Organic cation transporter 3 is densely expressed in the intercalated cell groups of the amygdala: anatomical evidence for a stress hormone-sensitive dopamine clearance system. J Chem Neuroanat. 2013;52:36–43. doi: 10.1016/j.jchemneu.2013.04.007. [DOI] [PubMed] [Google Scholar]
- Hoge EA, Worthington JJ, Nagurney JT, Chang Y, Kay EB, Feterowski CM, et al. Effect of acute posttrauma propranolol on PTSD outcome and physiological responses during script-driven imagery. CNS Neurosci Ther. 2012;18:21–27. doi: 10.1111/j.1755-5949.2010.00227.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes A. Genetic variation in cortico-amygdala serotonin function and risk for stress-related disease. Neurosci Biobehav Rev. 2008;32:1293–1314. doi: 10.1016/j.neubiorev.2008.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes A, Quirk GJ. Pharmacological facilitation of fear extinction and the search for adjunct treatments for anxiety disorders – the case of yohimbine. Trends Pharmacol Sci. 2010;31:2–7. doi: 10.1016/j.tips.2009.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes A, Singewald N. Individual differences in recovery from traumatic fear. Trends Neurosci. 2013;36:23–31. doi: 10.1016/j.tins.2012.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes A, Heilig M, Rupniak NM, Steckler T, Griebel G. Neuropeptide systems as novel therapeutic targets for depression and anxiety disorders. Trends Pharmacol Sci. 2003;24:580–588. doi: 10.1016/j.tips.2003.09.011. [DOI] [PubMed] [Google Scholar]
- Holmes A, Lachowicz JE, Sibley DR. Phenotypic analysis of dopamine receptor knockout mice; recent insights into the functional specificity of dopamine receptor subtypes. Neuropharmacology. 2004;47:1117–1134. doi: 10.1016/j.neuropharm.2004.07.034. [DOI] [PubMed] [Google Scholar]
- Holmes A, Fitzgerald PJ, Macpherson KP, Debrouse L, Colacicco G, Flynn SM, et al. Chronic alcohol remodels prefrontal neurons and disrupts NMDAR-mediated fear extinction encoding. Nat Neurosci. 2012;15:1359–1361. doi: 10.1038/nn.3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes NM, Parkes SL, Killcross AS, Westbrook RF. The basolateral amygdala is critical for learning about neutral stimuli in the presence of danger, and the perirhinal cortex is critical in the absence of danger. J Neurosci. 2013;33:13112–13125. doi: 10.1523/JNEUROSCI.1998-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holtzman-Assif O, Laurent V, Westbrook RF. Blockade of dopamine activity in the nucleus accumbens impairs learning extinction of conditioned fear. Learn Mem. 2010;17:71–75. doi: 10.1101/lm.1668310. [DOI] [PubMed] [Google Scholar]
- Hoover WB, Vertes RP. Anatomical analysis of afferent projections to the medial prefrontal cortex in the rat. Brain Struct Funct. 2007;212:149–179. doi: 10.1007/s00429-007-0150-4. [DOI] [PubMed] [Google Scholar]
- Huber D, Veinante P, Stoop R. Vasopressin and oxytocin excite distinct neuronal populations in the central amygdala. Science. 2005;308:245–248. doi: 10.1126/science.1105636. [DOI] [PubMed] [Google Scholar]
- Hugues S, Garcia R, Lena I. Time course of extracellular catecholamine and glutamate levels in the rat medial prefrontal cortex during and after extinction of conditioned fear. Synapse. 2007;61:933–937. doi: 10.1002/syn.20448. [DOI] [PubMed] [Google Scholar]
- Hurley KM, Herbert H, Moga MM, Saper CB. Efferent projections of the infralimbic cortex of the rat. J Comp Neurol. 1991;308:249–276. doi: 10.1002/cne.903080210. [DOI] [PubMed] [Google Scholar]
- Iafrati J, Orejarena MJ, Lassalle O, Bouamrane L, Chavis P. Reelin, an extracellular matrix protein linked to early onset psychiatric diseases, drives postnatal development of the prefrontal cortex via GluN2B-NMDARs and the mTOR pathway. Mol Psychiatry. 2013 doi: 10.1038/mp.2013.66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikawa A, Nakamura S. Convergence and interaction of hippocampal and amygdalar projections within the prefrontal cortex in the rat. J Neurosci. 2003;23:9987–9995. doi: 10.1523/JNEUROSCI.23-31-09987.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikawa S, Saito Y, Yanagawa Y, Otani S, Hiraide S, Shimamura K, et al. Early postnatal stress alters extracellular signal-regulated kinase signaling in the corticolimbic system modulating emotional circuitry in adult rats. Eur J Neurosci. 2012;35:135–145. doi: 10.1111/j.1460-9568.2011.07921.x. [DOI] [PubMed] [Google Scholar]
- Isiegas C, Park A, Kandel ER, Abel T, Lattal KM. Transgenic inhibition of neuronal protein kinase A activity facilitates fear extinction. J Neurosci. 2006;26:12700–12707. doi: 10.1523/JNEUROSCI.2743-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Izquierdo A, Wellman CL, Holmes A. Brief uncontrollable stress causes dendritic retraction in infralimbic cortex and resistance to fear extinction in mice. J Neurosci. 2006;26:5733–5738. doi: 10.1523/JNEUROSCI.0474-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobsen KX, Hoistad M, Staines WA, Fuxe K. The distribution of dopamine D1 receptor and mu-opioid receptor 1 receptor immunoreactivities in the amygdala and interstitial nucleus of the posterior limb of the anterior commissure: relationships to tyrosine hydroxylase and opioid peptide terminal systems. Neuroscience. 2006;141:2007–2018. doi: 10.1016/j.neuroscience.2006.05.054. [DOI] [PubMed] [Google Scholar]
- Jacobson LH, Kelly PH, Bettler B, Kaupmann K, Cryan JF. GABA(B(1) ) receptor isoforms differentially mediate the acquisition and extinction of aversive taste memories. J Neurosci. 2006;26:8800–8803. doi: 10.1523/JNEUROSCI.2076-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jasnow AM, Ehrlich DE, Choi DC, Dabrowska J, Bowers ME, McCullough KM, et al. Thy1-expressing neurons in the basolateral amygdala may mediate fear inhibition. J Neurosci. 2013;33:10396–10404. doi: 10.1523/JNEUROSCI.5539-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johansen JP, Hamanaka H, Monfils MH, Behnia R, Deisseroth K, Blair HT, et al. Optical activation of lateral amygdala pyramidal cells instructs associative fear learning. Proc Natl Acad Sci U S A. 2010;107:12692–12697. doi: 10.1073/pnas.1002418107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johansen JP, Wolff SB, Luthi A, LeDoux JE. Controlling the elements: an optogenetic approach to understanding the neural circuits of fear. Biol Psychiatry. 2012;71:1053–1060. doi: 10.1016/j.biopsych.2011.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Judo C, Matsumoto M, Yamazaki D, Hiraide S, Yanagawa Y, Kimura S, et al. Early stress exposure impairs synaptic potentiation in the rat medial prefrontal cortex underlying contextual fear extinction. Neuroscience. 2010;169:1705–1714. doi: 10.1016/j.neuroscience.2010.06.035. [DOI] [PubMed] [Google Scholar]
- Jungling K, Seidenbecher T, Sosulina L, Lesting J, Sangha S, Clark SD, et al. Neuropeptide S-mediated control of fear expression and extinction: role of intercalated GABAergic neurons in the amygdala. Neuron. 2008;59:298–310. doi: 10.1016/j.neuron.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamano DK. Using drugs to modify the effect of response prevention on avoidance extinction. Behav Res Ther. 1972;10:367–370. doi: 10.1016/0005-7967(72)90059-9. [DOI] [PubMed] [Google Scholar]
- Kamprath K, Plendl W, Marsicano G, Deussing JM, Wurst W, Lutz B, et al. Endocannabinoids mediate acute fear adaptation via glutamatergic neurons independently of corticotropin-releasing hormone signaling. Genes Brain Behav. 2009;8:203–211. doi: 10.1111/j.1601-183X.2008.00463.x. [DOI] [PubMed] [Google Scholar]
- Kaoru T, Liu FC, Ishida M, Oishi T, Hayashi M, Kitagawa M, et al. Molecular characterization of the intercalated cell masses of the amygdala: implications for the relationship with the striatum. Neuroscience. 2010;166:220–230. doi: 10.1016/j.neuroscience.2009.12.004. [DOI] [PubMed] [Google Scholar]
- Karpova NN, Pickenhagen A, Lindholm J, Tiraboschi E, Kulesskaya N, Agustsdottir A, et al. Fear erasure in mice requires synergy between antidepressant drugs and extinction training. Science. 2012;334:1731–1734. doi: 10.1126/science.1214592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med. 2003;9:76–81. doi: 10.1038/nm803. [DOI] [PubMed] [Google Scholar]
- Kessler RC, Avenevoli S, McLaughlin KA, Green JG, Lakoma MD, Petukhova M, et al. Lifetime co-morbidity of DSM-IV disorders in the US National Comorbidity Survey Replication Adolescent Supplement (NCS-A) Psychol Med. 2011;42:1997–2010. doi: 10.1017/S0033291712000025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, Lee S, Park H, Song B, Hong I, Geum D, et al. Blockade of amygdala metabotropic glutamate receptor subtype 1 impairs fear extinction. Biochem Biophys Res Commun. 2007a;355:188–193. doi: 10.1016/j.bbrc.2007.01.125. [DOI] [PubMed] [Google Scholar]
- Kim J, Lee S, Park K, Hong I, Song B, Son G, et al. Amygdala depotentiation and fear extinction. Proc Natl Acad Sci U S A. 2007b;104:20955–20960. doi: 10.1073/pnas.0710548105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JH, Richardson R. A developmental dissociation of context and GABA effects on extinguished fear in rats. Behav Neurosci. 2007;121:131–139. doi: 10.1037/0735-7044.121.1.131. [DOI] [PubMed] [Google Scholar]
- Kim JH, Richardson R. Expression of renewal is dependent on the extinction-test interval rather than the acquisition-extinction interval. Behav Neurosci. 2009;123:641–649. doi: 10.1037/a0015237. [DOI] [PubMed] [Google Scholar]
- Kim JH, Li S, Richardson R. Immunohistochemical analyses of long-term extinction of conditioned fear in adolescent rats. Cereb Cortex. 2011;21:530–538. doi: 10.1093/cercor/bhq116. [DOI] [PubMed] [Google Scholar]
- Kim SC, Jo YS, Kim IH, Kim H, Choi JS. Lack of medial prefrontal cortex activation underlies the immediate extinction deficit. J Neurosci. 2010;30:832–837. doi: 10.1523/JNEUROSCI.4145-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klumpers F, Denys D, Kenemans JL, Grillon C, van der Aart J, Baas JM. Testing the effects of Δ9-THC and D-cycloserine on extinction of conditioned fear in humans. J Psychopharmacol. 2012;26:471–478. doi: 10.1177/0269881111431624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knapska E, Maren S. Reciprocal patterns of c-Fos expression in the medial prefrontal cortex and amygdala after extinction and renewal of conditioned fear. Learn Mem. 2009;16:486–493. doi: 10.1101/lm.1463909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knapska E, Macias M, Mikosz M, Nowak A, Owczarek D, Wawrzyniak M, et al. Functional anatomy of neural circuits regulating fear and extinction. Proc Natl Acad Sci U S A. 2012;109:17093–17098. doi: 10.1073/pnas.1202087109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knobloch HS, Charlet A, Hoffmann LC, Eliava M, Khrulev S, Cetin AH, et al. Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron. 2012;73:553–566. doi: 10.1016/j.neuron.2011.11.030. [DOI] [PubMed] [Google Scholar]
- Knoll AT, Muschamp JW, Sillivan SE, Ferguson D, Dietz DM, Meloni EG, et al. Kappa opioid receptor signaling in the basolateral amygdala regulates conditioned fear and anxiety in rats. Biol Psychiatry. 2011;70:425–433. doi: 10.1016/j.biopsych.2011.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knox D, George SA, Fitzpatrick CJ, Rabinak CA, Maren S, Liberzon I. Single prolonged stress disrupts retention of extinguished fear in rats. Learn Mem. 2012a;19:43–49. doi: 10.1101/lm.024356.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knox D, Nault T, Henderson C, Liberzon I. Glucocorticoid receptors and extinction retention deficits in the single prolonged stress model. Neuroscience. 2012b;223:163–173. doi: 10.1016/j.neuroscience.2012.07.047. [DOI] [PubMed] [Google Scholar]
- Kohara K, Yasuda H, Huang Y, Adachi N, Sohya K, Tsumoto T. A local reduction in cortical GABAergic synapses after a loss of endogenous brain-derived neurotrophic factor, as revealed by single-cell gene knock-out method. J Neurosci. 2007;27:7234–7244. doi: 10.1523/JNEUROSCI.1943-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kondo M, Nakamura Y, Ishida Y, Yamada T, Shimada S. The 5-HT3A receptor is essential for fear extinction. Learn Mem. 2013;21:740–743. doi: 10.1101/lm.032193.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovacs GL, Bohus B, Versteeg DH, de Kloet ER, de Wied D. Effect of oxytocin and vasopressin on memory consolidation: sites of action and catecholaminergic correlates after local microinjection into limbic-midbrain structures. Brain Res. 1979;175:303–314. doi: 10.1016/0006-8993(79)91009-6. [DOI] [PubMed] [Google Scholar]
- Krettek JE, Price JL. Projections from the amygdaloid complex to the cerebral cortex and thalamus in the rat and cat. J Comp Neurol. 1977;172:687–722. doi: 10.1002/cne.901720408. [DOI] [PubMed] [Google Scholar]
- Kroner S, Rosenkranz JA, Grace AA, Barrionuevo G. Dopamine modulates excitability of basolateral amygdala neurons in vitro. J Neurophysiol. 2005;93:1598–1610. doi: 10.1152/jn.00843.2004. [DOI] [PubMed] [Google Scholar]
- Kuhnert S, Meyer C, Koch M. Involvement of cannabinoid receptors in the amygdala and prefrontal cortex of rats in fear learning, consolidation, retrieval and extinction. Behav Brain Res. 2013;250:274–284. doi: 10.1016/j.bbr.2013.05.002. [DOI] [PubMed] [Google Scholar]
- Lach G, de Lima TC. Role of NPY Y1 receptor on acquisition, consolidation and extinction on contextual fear conditioning: dissociation between anxiety, locomotion and non-emotional memory behavior. Neurobiol Learn Mem. 2013;103:26–33. doi: 10.1016/j.nlm.2013.04.005. [DOI] [PubMed] [Google Scholar]
- Lahoud N, Maroun M. Oxytocinergic manipulations in corticolimbic circuit differentially affect fear acquisition and extinction. Psychoneuroendocrinology. 2013;38:2184–2195. doi: 10.1016/j.psyneuen.2013.04.006. [DOI] [PubMed] [Google Scholar]
- Lammel S, Tye KM, Warden MR. Progress in understanding mood disorders: optogenetic dissection of neural circuits. Genes Brain Behav. 2014;13:38–51. doi: 10.1111/gbb.12049. [DOI] [PubMed] [Google Scholar]
- Langton JM, Richardson R. The effect of D-cycloserine on immediate vs. delayed extinction of learned fear. Learn Mem. 2010;17:547–551. doi: 10.1101/lm.1927310. [DOI] [PubMed] [Google Scholar]
- Laricchiuta D, Centonze D, Petrosini L. Effects of endocannabinoid and endovanilloid systems on aversive memory extinction. Behav Brain Res. 2013;256:101–107. doi: 10.1016/j.bbr.2013.08.010. [DOI] [PubMed] [Google Scholar]
- Lattal KM, Barrett RM, Wood MA. Systemic or intrahippocampal delivery of histone deacetylase inhibitors facilitates fear extinction. Behav Neurosci. 2007;121:1125–1131. doi: 10.1037/0735-7044.121.5.1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laurent V, Westbrook RF. Distinct contributions of the basolateral amygdala and the medial prefrontal cortex to learning and relearning extinction of context conditioned fear. Learn Mem. 2008;15:657–666. doi: 10.1101/lm.1080108. [DOI] [PubMed] [Google Scholar]
- Laurent V, Westbrook RF. Inactivation of the infralimbic but not the prelimbic cortex impairs consolidation and retrieval of fear extinction. Learn Mem. 2009a;16:520–529. doi: 10.1101/lm.1474609. [DOI] [PubMed] [Google Scholar]
- Laurent V, Westbrook RF. Infusion of the NMDA receptor antagonist, DL-APV, into the basolateral amygdala disrupts learning to fear a novel and a familiar context as well as relearning to fear an extinguished context. Learn Mem. 2009b;16:96–105. doi: 10.1101/lm.1218709. [DOI] [PubMed] [Google Scholar]
- Laurent V, Westbrook RF. Role of the basolateral amygdala in the reinstatement and extinction of fear responses to a previously extinguished conditioned stimulus. Learn Mem. 2010;17:86–96. doi: 10.1101/lm.1655010. [DOI] [PubMed] [Google Scholar]
- Laurent V, Marchand AR, Westbrook RF. The basolateral amygdala is necessary for learning but not relearning extinction of context conditioned fear. Learn Mem. 2008;15:304–314. doi: 10.1101/lm.928208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laviolette SR, Lipski WJ, Grace AA. A subpopulation of neurons in the medial prefrontal cortex encodes emotional learning with burst and frequency codes through a dopamine D4 receptor-dependent basolateral amygdala input. J Neurosci. 2005;25:6066–6075. doi: 10.1523/JNEUROSCI.1168-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leaderbrand K, Corcoran KA, Radulovic J. Co-activation of NR2A and NR2B subunits induces resistance to fear extinction. Neurobiol Learn Mem. 2014;113:35–40. doi: 10.1016/j.nlm.2013.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lebron-Milad K, Milad MR. Sex differences, gonadal hormones and the fear extinction network: implications for anxiety disorders. Biol Mood Anxiety Disord. 2012;2:3. doi: 10.1186/2045-5380-2-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ledgerwood L, Richardson R, Cranney J. Effects of D-cycloserine on extinction of conditioned freezing. Behav Neurosci. 2003;117:341–349. doi: 10.1037/0735-7044.117.2.341. [DOI] [PubMed] [Google Scholar]
- Ledgerwood L, Richardson R, Cranney J. D-cycloserine facilitates extinction of learned fear: effects on reacquisition and generalized extinction. Biol Psychiatry. 2005;57:841–847. doi: 10.1016/j.biopsych.2005.01.023. [DOI] [PubMed] [Google Scholar]
- Lee H, Kim JJ. Amygdalar NMDA receptors are critical for new fear learning in previously fear-conditioned rats. J Neurosci. 1998;18:8444–8454. doi: 10.1523/JNEUROSCI.18-20-08444.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JL, Milton AL, Everitt BJ. Reconsolidation and extinction of conditioned fear: inhibition and potentiation. J Neurosci. 2006;26:10051–10056. doi: 10.1523/JNEUROSCI.2466-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee S, Song B, Kim J, Park K, Hong I, An B, et al. GluA1 phosphorylation at serine 831 in the lateral amygdala is required for fear renewal. Nat Neurosci. 2013;16:1436–1444. doi: 10.1038/nn.3491. [DOI] [PubMed] [Google Scholar]
- Lesting J, Narayanan RT, Kluge C, Sangha S, Seidenbecher T, Pape HC. Patterns of coupled theta activity in amygdala-hippocampal-prefrontal cortical circuits during fear extinction. PLoS ONE. 2011;6:e21714. doi: 10.1371/journal.pone.0021714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G, Amano T, Pare D, Nair SS. Impact of infralimbic inputs on intercalated amygdala neurons: a biophysical modeling study. Learn Mem. 2011;18:226–240. doi: 10.1101/lm.1938011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H, Penzo MA, Taniguchi H, Kopec CD, Huang ZJ, Li B. Experience-dependent modification of a central amygdala fear circuit. Nat Neurosci. 2013;16:332–339. doi: 10.1038/nn.3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, Kim JH, Richardson R. Differential involvement of the medial prefrontal cortex in the expression of learned fear across development. Behav Neurosci. 2012;126:217–225. doi: 10.1037/a0027151. [DOI] [PubMed] [Google Scholar]
- Likhtik E, Pelletier JG, Paz R, Pare D. Prefrontal control of the amygdala. J Neurosci. 2005;25:7429–7437. doi: 10.1523/JNEUROSCI.2314-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Likhtik E, Popa D, Apergis-Schoute J, Fidacaro GA, Pare D. Amygdala intercalated neurons are required for expression of fear extinction. Nature. 2008;454:642–645. doi: 10.1038/nature07167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin CH, Lee CC, Gean PW. Involvement of a calcineurin cascade in amygdala depotentiation and quenching of fear memory. Mol Pharmacol. 2003a;63:44–52. doi: 10.1124/mol.63.1.44. [DOI] [PubMed] [Google Scholar]
- Lin CH, Yeh SH, Leu TH, Chang WC, Wang ST, Gean PW. Identification of calcineurin as a key signal in the extinction of fear memory. J Neurosci. 2003b;23:1574–1579. doi: 10.1523/JNEUROSCI.23-05-01574.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin CH, Yeh SH, Lu HY, Gean PW. The similarities and diversities of signal pathways leading to consolidation of conditioning and consolidation of extinction of fear memory. J Neurosci. 2003c;23:8310–8317. doi: 10.1523/JNEUROSCI.23-23-08310.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin HC, Mao SC, Gean PW. Effects of intra-amygdala infusion of CB1 receptor agonists on the reconsolidation of fear-potentiated startle. Learn Mem. 2006;13:316–321. doi: 10.1101/lm.217006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin HC, Mao SC, Gean PW. Block of gamma-aminobutyric acid-A receptor insertion in the amygdala impairs extinction of conditioned fear. Biol Psychiatry. 2009a;66:665–673. doi: 10.1016/j.biopsych.2009.04.003. [DOI] [PubMed] [Google Scholar]
- Lin HC, Mao SC, Su CL, Gean PW. The role of prefrontal cortex CB1 receptors in the modulation of fear memory. Cereb Cortex. 2009b;19:165–175. doi: 10.1093/cercor/bhn075. [DOI] [PubMed] [Google Scholar]
- Lin HC, Mao SC, Su CL, Gean PW. Alterations of excitatory transmission in the lateral amygdala during expression and extinction of fear memory. Int J Neuropsychopharmacol. 2010;13:335–345. doi: 10.1017/S1461145709990678. [DOI] [PubMed] [Google Scholar]
- Little JP, Carter AG. Subcellular synaptic connectivity of layer 2 pyramidal neurons in the medial prefrontal cortex. J Neurosci. 2012;32:12808–12819. doi: 10.1523/JNEUROSCI.1616-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Little JP, Carter AG. Synaptic mechanisms underlying strong reciprocal connectivity between the medial prefrontal cortex and basolateral amygdala. J Neurosci. 2013;33:15333–15342. doi: 10.1523/JNEUROSCI.2385-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Ramirez S, Pang PT, Puryear CB, Govindarajan A, Deisseroth K, et al. Optogenetic stimulation of a hippocampal engram activates fear memory recall. Nature. 2012;484:381–385. doi: 10.1038/nature11028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Livneh U, Paz R. Aversive-bias and stage-selectivity in neurons of the primate amygdala during acquisition, extinction, and overnight retention. J Neurosci. 2012;32:8598–8610. doi: 10.1523/JNEUROSCI.0323-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Long VA, Fanselow MS. Stress-enhanced fear learning in rats is resistant to the effects of immediate massed extinction. Stress. 2012;15:627–636. doi: 10.3109/10253890.2011.650251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loretan K, Bissiere S, Luthi A. Dopaminergic modulation of spontaneous inhibitory network activity in the lateral amygdala. Neuropharmacology. 2004;47:631–639. doi: 10.1016/j.neuropharm.2004.07.015. [DOI] [PubMed] [Google Scholar]
- Macpherson K, Whittle N, Camp M, Gunduz-Cinar O, Singewald N, Holmes A. Temporal factors in the extinction of fear in inbred mouse strains differing in extinction efficacy. Biol Mood Anxiety Disord. 2013;3:13. doi: 10.1186/2045-5380-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Makkar SR, Zhang SQ, Cranney J. Behavioral and neural analysis of GABA in the acquisition, consolidation, reconsolidation, and extinction of fear memory. Neuropsychopharmacology. 2010;35:1625–1652. doi: 10.1038/npp.2010.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manko M, Geracitano R, Capogna M. Functional connectivity of the main intercalated nucleus of the mouse amygdala. J Physiol. 2011;589:1911–1925. doi: 10.1113/jphysiol.2010.201475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao SC, Hsiao YH, Gean PW. Extinction training in conjunction with a partial agonist of the glycine site on the NMDA receptor erases memory trace. J Neurosci. 2006;26:8892–8899. doi: 10.1523/JNEUROSCI.0365-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao SC, Lin HC, Gean PW. Augmentation of fear extinction by D-cycloserine is blocked by proteasome inhibitors. Neuropsychopharmacology. 2008;33:3085–3095. doi: 10.1038/npp.2008.30. [DOI] [PubMed] [Google Scholar]
- Mao SC, Lin HC, Gean PW. Augmentation of fear extinction by infusion of glycine transporter blockers into the amygdala. Mol Pharmacol. 2009;76:369–378. doi: 10.1124/mol.108.053728. [DOI] [PubMed] [Google Scholar]
- Mao SC, Chang CH, Wu CC, Orejanera MJ, Manzoni OJ, Gean PW. Inhibition of spontaneous recovery of fear by mGluR5 after prolonged extinction training. PLoS ONE. 2013;8:e59580. doi: 10.1371/journal.pone.0059580. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Marek R, Coelho CM, Sullivan RK, Baker-Andresen D, Li X, Ratnu V, et al. Paradoxical enhancement of fear extinction memory and synaptic plasticity by inhibition of the histone acetyltransferase p300. J Neurosci. 2011;31:7486–7491. doi: 10.1523/JNEUROSCI.0133-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maren S. Nature and causes of the immediate extinction deficit: a brief review. Neurobiol Learn Mem. 2014;113:19–24. doi: 10.1016/j.nlm.2013.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maren S, Chang CH. Recent fear is resistant to extinction. Proc Natl Acad Sci U S A. 2006;103:18020–18025. doi: 10.1073/pnas.0608398103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maroun M. Stress reverses plasticity in the pathway projecting from the ventromedial prefrontal cortex to the basolateral amygdala. Eur J Neurosci. 2006;24:2917–2922. doi: 10.1111/j.1460-9568.2006.05169.x. [DOI] [PubMed] [Google Scholar]
- Maroun M, Richter-Levin G. Exposure to acute stress blocks the induction of long-term potentiation of the amygdala-prefrontal cortex pathway in vivo. J Neurosci. 2003;23:4406–4409. doi: 10.1523/JNEUROSCI.23-11-04406.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maroun M, Ioannides PJ, Bergman KL, Kavushansky A, Holmes A, Wellman CL. Fear extinction deficits following acute stress associate with increased spine density and dendritic retraction in basolateral amygdala neurons. Eur J Neurosci. 2013;38:2611–2620. doi: 10.1111/ejn.12259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marowsky A, Yanagawa Y, Obata K, Vogt KE. A specialized subclass of interneurons mediates dopaminergic facilitation of amygdala function. Neuron. 2005;48:1025–1037. doi: 10.1016/j.neuron.2005.10.029. [DOI] [PubMed] [Google Scholar]
- Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature. 2002;418:530–534. doi: 10.1038/nature00839. [DOI] [PubMed] [Google Scholar]
- Martel G, Hevi C, Wong A, Zushida K, Uchida S, Shumyatsky GP. Murine GRPR and stathmin control in opposite directions both cued fear extinction and neural activities of the amygdala and prefrontal cortex. PLoS ONE. 2012;7:e30942. doi: 10.1371/journal.pone.0030942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marvar PJ, Goodman J, Fuchs S, Choi DC, Banerjee S, Ressler KJ. Angiotensin type 1 receptor inhibition enhances the extinction of fear memory. Biol Psychiatry. 2014;75:864–872. doi: 10.1016/j.biopsych.2013.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mascagni F, McDonald AJ. Immunohistochemical characterization of cholecystokinin containing neurons in the rat basolateral amygdala. Brain Res. 2003;976:171–184. doi: 10.1016/s0006-8993(03)02625-8. [DOI] [PubMed] [Google Scholar]
- Mascagni F, McDonald AJ. A novel subpopulation of 5-HT type 3A receptor subunit immunoreactive interneurons in the rat basolateral amygdala. Neuroscience. 2007;144:1015–1024. doi: 10.1016/j.neuroscience.2006.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuda S, Matsuzawa D, Nakazawa K, Sutoh C, Ohtsuka H, Ishii D, et al. d-serine enhances extinction of auditory cued fear conditioning via ERK1/2 phosphorylation in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:895–902. doi: 10.1016/j.pnpbp.2010.04.013. [DOI] [PubMed] [Google Scholar]
- Matsumoto M, Togashi H, Konno K, Koseki H, Hirata R, Izumi T, et al. Early postnatal stress alters the extinction of context-dependent conditioned fear in adult rats. Pharmacol Biochem Behav. 2008;89:247–252. doi: 10.1016/j.pbb.2007.12.017. [DOI] [PubMed] [Google Scholar]
- Matsumoto Y, Morinobu S, Yamamoto S, Matsumoto T, Takei S, Fujita Y, et al. Vorinostat ameliorates impaired fear extinction possibly via the hippocampal NMDA-CaMKII pathway in an animal model of posttraumatic stress disorder. Psychopharmacology (Berl) 2013;229:51–62. doi: 10.1007/s00213-013-3078-9. [DOI] [PubMed] [Google Scholar]
- McCallum J, Kim JH, Richardson R. Impaired extinction retention in adolescent rats: effects of D-cycloserine. Neuropsychopharmacology. 2010;35:2134–2142. doi: 10.1038/npp.2010.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonald AJ. Organization of amygdaloid projections to the prefrontal cortex and associated striatum in the rat. Neuroscience. 1991;44:1–14. doi: 10.1016/0306-4522(91)90247-l. [DOI] [PubMed] [Google Scholar]
- McDonald AJ. Cortical pathways to the mammalian amygdala. Prog Neurobiol. 1998;55:257–332. doi: 10.1016/s0301-0082(98)00003-3. [DOI] [PubMed] [Google Scholar]
- McDonald AJ, Augustine JR. Localization of GABA-like immunoreactivity in the monkey amygdala. Neuroscience. 1993;52:281–294. doi: 10.1016/0306-4522(93)90156-a. [DOI] [PubMed] [Google Scholar]
- McDonald AJ, Mascagni F. Neuronal localization of 5-HT type 2A receptor immunoreactivity in the rat basolateral amygdala. Neuroscience. 2007;146:306–320. doi: 10.1016/j.neuroscience.2007.01.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonald AJ, Pearson JC. Coexistence of GABA and peptide immunoreactivity in non-pyramidal neurons of the basolateral amygdala. Neurosci Lett. 1989;100:53–58. doi: 10.1016/0304-3940(89)90659-9. [DOI] [PubMed] [Google Scholar]
- McDonald AJ, Mascagni F, Guo L. Projections of the medial and lateral prefrontal cortices to the amygdala: a Phaseolus vulgaris leucoagglutinin study in the rat. Neuroscience. 1996;71:55–75. doi: 10.1016/0306-4522(95)00417-3. [DOI] [PubMed] [Google Scholar]
- McGaugh JL, Castellano C, Brioni J. Picrotoxin enhances latent extinction of conditioned fear. Behav Neurosci. 1990;104:264–267. doi: 10.1037//0735-7044.104.2.264. [DOI] [PubMed] [Google Scholar]
- McNally GP. Facilitation of fear extinction by midbrain periaqueductal gray infusions of RB101(S), an inhibitor of enkephalin-degrading enzymes. Behav Neurosci. 2005;119:1672–1677. doi: 10.1037/0735-7044.119.6.1672. [DOI] [PubMed] [Google Scholar]
- McNally GP, Westbrook RF. Opioid receptors regulate the extinction of Pavlovian fear conditioning. Behav Neurosci. 2003;117:1292–1301. doi: 10.1037/0735-7044.117.6.1292. [DOI] [PubMed] [Google Scholar]
- McNally GP, Pigg M, Weidemann G. Opioid receptors in the midbrain periaqueductal gray regulate extinction of pavlovian fear conditioning. J Neurosci. 2004;24:6912–6919. doi: 10.1523/JNEUROSCI.1828-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miczek KA, Luttinger D. Differential attenuation of two kinds of conditioned suppression by d-amphetamine and pentobarbital. J Pharmacol Exp Ther. 1978;205:282–290. [PubMed] [Google Scholar]
- Milad MR, Quirk GJ. Neurons in medial prefrontal cortex signal memory for fear extinction. Nature. 2002;420:70–74. doi: 10.1038/nature01138. [DOI] [PubMed] [Google Scholar]
- Millhouse OE. The intercalated cells of the amygdala. J Comp Neurol. 1986;247:246–271. doi: 10.1002/cne.902470209. [DOI] [PubMed] [Google Scholar]
- Miracle AD, Brace MF, Huyck KD, Singler SA, Wellman CL. Chronic stress impairs recall of extinction of conditioned fear. Neurobiol Learn Mem. 2006;85:213–218. doi: 10.1016/j.nlm.2005.10.005. [DOI] [PubMed] [Google Scholar]
- Morawska MM, Fendt M. The effects of muscimol and AMN082 injections into the medial prefrontal cortex on the expression and extinction of conditioned fear in mice. J Exp Biol. 2012;215:1394–1398. doi: 10.1242/jeb.068213. [DOI] [PubMed] [Google Scholar]
- Morris MJ, Mahgoub M, Na ES, Pranav H, Monteggia LM. Loss of histone deacetylase 2 improves working memory and accelerates extinction learning. J Neurosci. 2013;33:6401–6411. doi: 10.1523/JNEUROSCI.1001-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris RW, Bouton ME. The effect of yohimbine on the extinction of conditioned fear: a role for context. Behav Neurosci. 2007;121:501–514. doi: 10.1037/0735-7044.121.3.501. [DOI] [PubMed] [Google Scholar]
- Morrow BA, Elsworth JD, Rasmusson AM, Roth RH. The role of mesoprefrontal dopamine neurons in the acquisition and expression of conditioned fear in the rat. Neuroscience. 1999;92:553–564. doi: 10.1016/s0306-4522(99)00014-7. [DOI] [PubMed] [Google Scholar]
- Mueller D, Porter JT, Quirk GJ. Noradrenergic signaling in infralimbic cortex increases cell excitability and strengthens memory for fear extinction. J Neurosci. 2008;28:369–375. doi: 10.1523/JNEUROSCI.3248-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller D, Olivera-Figueroa LA, Pine DS, Quirk GJ. The effects of yohimbine and amphetamine on fear expression and extinction in rats. Psychopharmacology (Berl) 2009;204:599–606. doi: 10.1007/s00213-009-1491-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller D, Bravo-Rivera C, Quirk GJ. Infralimbic D2 receptors are necessary for fear extinction and extinction-related tone responses. Biol Psychiatry. 2010;68:1055–1060. doi: 10.1016/j.biopsych.2010.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muigg P, Hetzenauer A, Hauer G, Hauschild M, Gaburro S, Frank E, et al. Impaired extinction of learned fear in rats selectively bred for high anxiety – evidence of altered neuronal processing in prefrontal-amygdala pathways. Eur J Neurosci. 2008;28:2299–2309. doi: 10.1111/j.1460-9568.2008.06511.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muller JF, Mascagni F, McDonald AJ. Dopaminergic innervation of pyramidal cells in the rat basolateral amygdala. Brain Struct Funct. 2009;213:275–288. doi: 10.1007/s00429-008-0196-y. [DOI] [PubMed] [Google Scholar]
- Muschamp JW, Van’t Veer A, Parsegian A, Gallo MS, Chen M, Neve RL, et al. Activation of CREB in the nucleus accumbens shell produces anhedonia and resistance to extinction of fear in rats. J Neurosci. 2011;31:3095–3103. doi: 10.1523/JNEUROSCI.5973-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Myers KM, Davis M. Mechanisms of fear extinction. Mol Psychiatry. 2007;12:120–150. doi: 10.1038/sj.mp.4001939. [DOI] [PubMed] [Google Scholar]
- Myers KM, Ressler KJ, Davis M. Different mechanisms of fear extinction dependent on length of time since fear acquisition. Learn Mem. 2006;13:216–223. doi: 10.1101/lm.119806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nader K, LeDoux JE. Inhibition of the mesoamygdala dopaminergic pathway impairs the retrieval of conditioned fear associations. Behav Neurosci. 1999;113:891–901. doi: 10.1037//0735-7044.113.5.891. [DOI] [PubMed] [Google Scholar]
- Narayanan V, Heiming RS, Jansen F, Lesting J, Sachser N, Pape HC, et al. Social defeat: impact on fear extinction and amygdala-prefrontal cortical theta synchrony in 5-HTT deficient mice. PLoS ONE. 2011;6:e22600. doi: 10.1371/journal.pone.0022600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nietzer SL, Bonn M, Jansen F, Heiming RS, Lewejohann L, Sachser N, et al. Serotonin transporter knockout and repeated social defeat stress: impact on neuronal morphology and plasticity in limbic brain areas. Behav Brain Res. 2011;220:42–54. doi: 10.1016/j.bbr.2011.01.011. [DOI] [PubMed] [Google Scholar]
- Nijholt IM, Ostroveanu A, Scheper WA, Penke B, Luiten PG, Van der Zee EA, et al. Inhibition of PKA anchoring to A-kinase anchoring proteins impairs consolidation and facilitates extinction of contextual fear memories. Neurobiol Learn Mem. 2008;90:223–229. doi: 10.1016/j.nlm.2008.03.008. [DOI] [PubMed] [Google Scholar]
- Nitecka L, Ben-Ari Y. Distribution of GABA-like immunoreactivity in the rat amygdaloid complex. J Comp Neurol. 1987;266:45–55. doi: 10.1002/cne.902660105. [DOI] [PubMed] [Google Scholar]
- Nonkes LJ, de Pooter M, Homberg JR. Behavioural therapy based on distraction alleviates impaired fear extinction in male serotonin transporter knockout rats. J Psychiatry Neurosci. 2012;37:224–230. doi: 10.1503/jpn.110116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Norcross M, Mathur P, Enoch AJ, Karlsson RM, Brigman JL, Cameron HA, et al. Effects of adolescent fluoxetine treatment on fear-, anxiety- or stress-related behaviors in C57BL/6J or BALB/cJ mice. Psychopharmacology (Berl) 2008;200:413–424. doi: 10.1007/s00213-008-1215-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogden KK, Khatri A, Traynelis SF, Heldt SA. Potentiation of GluN2C/D NMDA receptor subtypes in the amygdala facilitates the retention of fear and extinction learning in mice. Neuropsychopharmacology. 2014;39:625–637. doi: 10.1038/npp.2013.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orr SP, Milad MR, Metzger LJ, Lasko NB, Gilbertson MW, Pitman RK. Effects of beta blockade, PTSD diagnosis, and explicit threat on the extinction and retention of an aversively conditioned response. Biol Psychol. 2006;73:262–271. doi: 10.1016/j.biopsycho.2006.05.001. [DOI] [PubMed] [Google Scholar]
- Orsini CA, Maren S. Neural and cellular mechanisms of fear and extinction memory formation. Neurosci Biobehav Rev. 2012;36:1773–1802. doi: 10.1016/j.neubiorev.2011.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otto MW, McHugh RK, Simon NM, Farach FJ, Worthington JJ, Pollack MH. Efficacy of CBT for benzodiazepine discontinuation in patients with panic disorder: further evaluation. Behav Res Ther. 2010;48:720–727. doi: 10.1016/j.brat.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ouyang M, Thomas SA. A requirement for memory retrieval during and after long-term extinction learning. Proc Natl Acad Sci U S A. 2005;102:9347–9352. doi: 10.1073/pnas.0502315102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pamplona FA, Prediger RD, Pandolfo P, Takahashi RN. The cannabinoid receptor agonist WIN 55,212-2 facilitates the extinction of contextual fear memory and spatial memory in rats. Psychopharmacology (Berl) 2006;188:641–649. doi: 10.1007/s00213-006-0514-0. [DOI] [PubMed] [Google Scholar]
- Pamplona FA, Bitencourt RM, Takahashi RN. Short- and long-term effects of cannabinoids on the extinction of contextual fear memory in rats. Neurobiol Learn Mem. 2008;90:290–293. doi: 10.1016/j.nlm.2008.04.003. [DOI] [PubMed] [Google Scholar]
- Pape HC, Pare D. Plastic synaptic networks of the amygdala for the acquisition, expression, and extinction of conditioned fear. Physiol Rev. 2010;90:419–463. doi: 10.1152/physrev.00037.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pare D, Quirk GJ, Ledoux JE. New vistas on amygdala networks in conditioned fear. J Neurophysiol. 2004;92:1–9. doi: 10.1152/jn.00153.2004. [DOI] [PubMed] [Google Scholar]
- Park SM, Williams CL. Contribution of serotonin type 3 receptors in the successful extinction of cued or contextual fear conditioned responses: interactions with GABAergic signaling. Rev Neurosci. 2012;23:555–569. doi: 10.1515/revneuro-2012-0052. [DOI] [PubMed] [Google Scholar]
- Parkes SL, Westbrook RF. The basolateral amygdala is critical for the acquisition and extinction of associations between a neutral stimulus and a learned danger signal but not between two neutral stimuli. J Neurosci. 2010;30:12608–12618. doi: 10.1523/JNEUROSCI.2949-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parkes SL, Westbrook RF. Role of the basolateral amygdala and NMDA receptors in higher-order conditioned fear. Rev Neurosci. 2011;22:317–333. doi: 10.1515/RNS.2011.025. [DOI] [PubMed] [Google Scholar]
- Parnas AS, Weber M, Richardson R. Effects of multiple exposures to D-cycloserine on extinction of conditioned fear in rats. Neurobiol Learn Mem. 2005;83:224–231. doi: 10.1016/j.nlm.2005.01.001. [DOI] [PubMed] [Google Scholar]
- Parsons RG, Gafford GM, Helmstetter FJ. Regulation of extinction-related plasticity by opioid receptors in the ventrolateral periaqueductal gray matter. Front Behav Neurosci. 2010;4:1–11. doi: 10.3389/fnbeh.2010.00044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pattwell SS, Duhoux S, Hartley CA, Johnson DC, Jing D, Elliott MD, et al. Altered fear learning across development in both mouse and human. Proc Natl Acad Sci U S A. 2012;109:16318–16323. doi: 10.1073/pnas.1206834109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pereira ME, Rosat R, Huang CH, Godoy MG, Izquierdo I. Inhibition by diazepam of the effect of additional training and of extinction on the retention of shuttle avoidance behavior in rats. Behav Neurosci. 1989;103:202–205. doi: 10.1037//0735-7044.103.1.202. [DOI] [PubMed] [Google Scholar]
- Perez-Jaranay JM, Vives F. Electrophysiological study of the response of medial prefrontal cortex neurons to stimulation of the basolateral nucleus of the amygdala in the rat. Brain Res. 1991;564:97–101. doi: 10.1016/0006-8993(91)91357-7. [DOI] [PubMed] [Google Scholar]
- Peters J, Dieppa-Perea LM, Melendez LM, Quirk GJ. Induction of fear extinction with hippocampal-infralimbic BDNF. Science. 2010;328:1288–1290. doi: 10.1126/science.1186909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pezawas L, Meyer-Lindenberg A, Drabant EM, Verchinski BA, Munoz KE, Kolachana BS, et al. 5-HTTLPR polymorphism impacts human cingulate-amygdala interactions: a genetic susceptibility mechanism for depression. Nat Neurosci. 2005;8:828–834. doi: 10.1038/nn1463. [DOI] [PubMed] [Google Scholar]
- Pfeiffer UJ, Fendt M. Prefrontal dopamine D4 receptors are involved in encoding fear extinction. Neuroreport. 2006;17:847–850. doi: 10.1097/01.wnr.0000220142.29413.6f. [DOI] [PubMed] [Google Scholar]
- Pinard CR, Muller JF, Mascagni F, McDonald AJ. Dopaminergic innervation of interneurons in the rat basolateral amygdala. Neuroscience. 2008;157:850–863. doi: 10.1016/j.neuroscience.2008.09.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinard CR, Mascagni F, McDonald AJ. Medial prefrontal cortical innervation of the intercalated nuclear region of the amygdala. Neuroscience. 2012;205:112–124. doi: 10.1016/j.neuroscience.2011.12.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinto A, Sesack SR. Ultrastructural analysis of prefrontal cortical inputs to the rat amygdala: spatial relationships to presumed dopamine axons and D1 and D2 receptors. Brain Struct Funct. 2008;213:159–175. doi: 10.1007/s00429-008-0180-6. [DOI] [PubMed] [Google Scholar]
- Pitts MW, Raman AV, Hashimoto AC, Todorovic C, Nichols RA, Berry MJ. Deletion of selenoprotein P results in impaired function of parvalbumin interneurons and alterations in fear learning and sensorimotor gating. Neuroscience. 2012;208:58–68. doi: 10.1016/j.neuroscience.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plendl W, Wotjak CT. Dissociation of within- and between-session extinction of conditioned fear. J Neurosci. 2010;30:4990–4998. doi: 10.1523/JNEUROSCI.6038-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ponnusamy R, Nissim HA, Barad M. Systemic blockade of D2-like dopamine receptors facilitates extinction of conditioned fear in mice. Learn Mem. 2005;12:399–406. doi: 10.1101/lm.96605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powers MB, Smits JA, Otto MW, Sanders C, Emmelkamp PM. Facilitation of fear extinction in phobic participants with a novel cognitive enhancer: a randomized placebo controlled trial of yohimbine augmentation. J Anxiety Disord. 2009;23:350–356. doi: 10.1016/j.janxdis.2009.01.001. [DOI] [PubMed] [Google Scholar]
- Psotta L, Lessmann V, Endres T. Impaired fear extinction learning in adult heterozygous BDNF knock-out mice. Neurobiol Learn Mem. 2013;103:34–38. doi: 10.1016/j.nlm.2013.03.003. [DOI] [PubMed] [Google Scholar]
- de Quervain DJ, Bentz D, Michael T, Bolt OC, Wiederhold BK, Margraf J, et al. Glucocorticoids enhance extinction-based psychotherapy. Proc Natl Acad Sci U S A. 2011;108:6621–6625. doi: 10.1073/pnas.1018214108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quirk GJ, Likhtik E, Pelletier JG, Pare D. Stimulation of medial prefrontal cortex decreases the responsiveness of central amygdala output neurons. J Neurosci. 2003;23:8800–8807. doi: 10.1523/JNEUROSCI.23-25-08800.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rabinak CA, Angstadt M, Sripada CS, Abelson JL, Liberzon I, Milad MR, et al. Cannabinoid facilitation of fear extinction memory recall in humans. Neuropharmacology. 2013;64:396–402. doi: 10.1016/j.neuropharm.2012.06.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rabinak CA, Angstadt M, Lyons M, Mori S, Milad MR, Liberzon I, et al. Cannabinoid modulation of prefrontal-limbic activation during fear extinction learning and recall in humans. Neurobiol Learn Mem. 2014;113:125–134. doi: 10.1016/j.nlm.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rainnie DG. Serotonergic modulation of neurotransmission in the rat basolateral amygdala. J Neurophysiol. 1999;82:69–85. doi: 10.1152/jn.1999.82.1.69. [DOI] [PubMed] [Google Scholar]
- Rainnie DG, Bergeron R, Sajdyk TJ, Patil M, Gehlert DR, Shekhar A. Corticotrophin releasing factor-induced synaptic plasticity in the amygdala translates stress into emotional disorders. J Neurosci. 2004;24:3471–3479. doi: 10.1523/JNEUROSCI.5740-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rainnie DG, Mania I, Mascagni F, McDonald AJ. Physiological and morphological characterization of parvalbumin-containing interneurons of the rat basolateral amygdala. J Comp Neurol. 2006;498:142–161. doi: 10.1002/cne.21049. [DOI] [PubMed] [Google Scholar]
- Ramirez S, Liu X, Lin PA, Suh J, Pignatelli M, Redondo RL, et al. Creating a false memory in the hippocampus. Science. 2013;341:387–391. doi: 10.1126/science.1239073. [DOI] [PubMed] [Google Scholar]
- Reich CG, Iskander AN, Weiss MS. Cannabinoid modulation of chronic mild stress-induced selective enhancement of trace fear conditioning in adolescent rats. J Psychopharmacol. 2013;27:947–955. doi: 10.1177/0269881113499207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Repa JC, Muller J, Apergis J, Desrochers TM, Zhou Y, LeDoux JE. Two different lateral amygdala cell populations contribute to the initiation and storage of memory. Nat Neurosci. 2001;4:724–731. doi: 10.1038/89512. [DOI] [PubMed] [Google Scholar]
- Ressler KJ, Mayberg HS. Targeting abnormal neural circuits in mood and anxiety disorders: from the laboratory to the clinic. Nat Neurosci. 2007;10:1116–1124. doi: 10.1038/nn1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riddle MC, McKenna MC, Yoon YJ, Pattwell SS, Santos PM, Casey BJ, et al. Caloric restriction enhances fear extinction learning in mice. Neuropsychopharmacology. 2013;38:930–937. doi: 10.1038/npp.2012.268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riebe CJ, Pamplona F, Kamprath K, Wotjak CT. Fear relief-toward a new conceptual frame work and what endocannabinoids gotta do with it. Neuroscience. 2012;204:159–185. doi: 10.1016/j.neuroscience.2011.11.057. [DOI] [PubMed] [Google Scholar]
- Rodrigues H, Figueira I, Goncalves R, Mendlowicz M, Macedo T, Ventura P. CBT for pharmacotherapy non-remitters – a systematic review of a next-step strategy. J Affect Disord. 2011;129:219–228. doi: 10.1016/j.jad.2010.08.025. [DOI] [PubMed] [Google Scholar]
- Rodriguez-Romaguera J, Sotres-Bayon F, Mueller D, Quirk GJ. Systemic propranolol acts centrally to reduce conditioned fear in rats without impairing extinction. Biol Psychiatry. 2009;65:887–892. doi: 10.1016/j.biopsych.2009.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenkranz JA, Grace AA. Modulation of basolateral amygdala neuronal firing and afferent drive by dopamine receptor activation in vivo. J Neurosci. 1999;19:11027–11039. doi: 10.1523/JNEUROSCI.19-24-11027.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenkranz JA, Grace AA. Cellular mechanisms of infralimbic and prelimbic prefrontal cortical inhibition and dopaminergic modulation of basolateral amygdala neurons in vivo. J Neurosci. 2002;22:324–337. doi: 10.1523/JNEUROSCI.22-01-00324.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Royer S, Martina M, Pare D. An inhibitory interface gates impulse traffic between the input and output stations of the amygdala. J Neurosci. 1999;19:10575–10583. doi: 10.1523/JNEUROSCI.19-23-10575.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruehle S, Remmers F, Romo-Parra H, Massa F, Wickert M, Wortge S, et al. Cannabinoid CB1 receptor in dorsal telencephalic glutamatergic neurons: distinctive sufficiency for hippocampus-dependent and amygdala-dependent synaptic and behavioral functions. J Neurosci. 2013;33:10264–10277. doi: 10.1523/JNEUROSCI.4171-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saito Y, Matsumoto M, Otani S, Yanagawa Y, Hiraide S, Ishikawa S, et al. Phase-dependent synaptic changes in the hippocampal CA1 field underlying extinction processes in freely moving rats. Neurobiol Learn Mem. 2012;97:361–369. doi: 10.1016/j.nlm.2012.02.006. [DOI] [PubMed] [Google Scholar]
- Saito Y, Matsumoto M, Yanagawa Y, Hiraide S, Inoue S, Kubo Y, et al. Facilitation of fear extinction by the 5-HT(1A) receptor agonist tandospirone: possible involvement of dopaminergic modulation. Synapse. 2013;67:161–170. doi: 10.1002/syn.21621. [DOI] [PubMed] [Google Scholar]
- Sangha S, Narayanan RT, Bergado-Acosta JR, Stork O, Seidenbecher T, Pape HC. Deficiency of the 65 kDa isoform of glutamic acid decarboxylase impairs extinction of cued but not contextual fear memory. J Neurosci. 2009;29:15713–15720. doi: 10.1523/JNEUROSCI.2620-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sangha S, Ilenseer J, Sosulina L, Lesting J, Pape HC. Differential regulation of glutamic acid decarboxylase gene expression after extinction of a recent memory vs. intermediate memory. Learn Mem. 2012;19:194–200. doi: 10.1101/lm.025874.112. [DOI] [PubMed] [Google Scholar]
- Santini E, Porter JT. M-type potassium channels modulate the intrinsic excitability of infralimbic neurons and regulate fear expression and extinction. J Neurosci. 2010;30:12379–12386. doi: 10.1523/JNEUROSCI.1295-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santini E, Muller RU, Quirk GJ. Consolidation of extinction learning involves transfer from NMDA-independent to NMDA-dependent memory. J Neurosci. 2001;21:9009–9017. doi: 10.1523/JNEUROSCI.21-22-09009.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santini E, Sepulveda-Orengo M, Porter JT. Muscarinic receptors modulate the intrinsic excitability of infralimbic neurons and consolidation of fear extinction. Neuropsychopharmacology. 2012;37:2047–2056. doi: 10.1038/npp.2012.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneier FR, Neria Y, Pavlicova M, Hembree E, Suh EJ, Amsel L, et al. Combined prolonged exposure therapy and paroxetine for PTSD related to the World Trade Center attack: a randomized controlled trial. Am J Psychiatry. 2012;169:80–88. doi: 10.1176/appi.ajp.2011.11020321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Segev A, Rubin AS, Abush H, Richter-Levin G, Akirav I. Cannabinoid receptor activation prevents the effects of chronic mild stress on emotional learning and LTP in a rat model of depression. Neuropsychopharmacology. 2014;39:919–933. doi: 10.1038/npp.2013.292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Senn V, Wolff SB, Herry C, Grenier F, Ehrlich I, Grundemann J, et al. Long-range connectivity defines behavioral specificity of amygdala neurons. Neuron. 2014;81:428–437. doi: 10.1016/j.neuron.2013.11.006. [DOI] [PubMed] [Google Scholar]
- Sepulveda-Orengo MT, Lopez AV, Soler-Cedeno O, Porter JT. Fear extinction induces mGluR5-mediated synaptic and intrinsic plasticity in infralimbic neurons. J Neurosci. 2013;33:7184–7193. doi: 10.1523/JNEUROSCI.5198-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sesack SR, Deutch AY, Roth RH, Bunney BS. Topographical organization of the efferent projections of the medial prefrontal cortex in the rat: an anterograde tract-tracing study with Phaseolus vulgaris leucoagglutinin. J Comp Neurol. 1989;290:213–242. doi: 10.1002/cne.902900205. [DOI] [PubMed] [Google Scholar]
- Sharma SK. Protein acetylation in synaptic plasticity and memory. Neurosci Biobehav Rev. 2010;34:1234–1240. doi: 10.1016/j.neubiorev.2010.02.009. [DOI] [PubMed] [Google Scholar]
- Shinonaga Y, Takada M, Mizuno N. Topographic organization of collateral projections from the basolateral amygdaloid nucleus to both the prefrontal cortex and nucleus accumbens in the rat. Neuroscience. 1994;58:389–397. doi: 10.1016/0306-4522(94)90045-0. [DOI] [PubMed] [Google Scholar]
- Shumyatsky GP, Malleret G, Shin RM, Takizawa S, Tully K, Tsvetkov E, et al. stathmin, a gene enriched in the amygdala, controls both learned and innate fear. Cell. 2005;123:697–709. doi: 10.1016/j.cell.2005.08.038. [DOI] [PubMed] [Google Scholar]
- Si W, Aluisio L, Okamura N, Clark SD, Fraser I, Sutton SW, et al. Neuropeptide S stimulates dopaminergic neurotransmission in the medial prefrontal cortex. J Neurochem. 2010;115:475–482. doi: 10.1111/j.1471-4159.2010.06947.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sierra-Mercado D, Padilla-Coreano N, Quirk GJ. Dissociable roles of prelimbic and infralimbic cortices, ventral hippocampus, and basolateral amygdala in the expression and extinction of conditioned fear. Neuropsychopharmacology. 2011;36:529–538. doi: 10.1038/npp.2010.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sierra-Mercado D, Jr, Corcoran KA, Lebron-Milad K, Quirk GJ. Inactivation of the ventromedial prefrontal cortex reduces expression of conditioned fear and impairs subsequent recall of extinction. Eur J Neurosci. 2006;24:1751–1758. doi: 10.1111/j.1460-9568.2006.05014.x. [DOI] [PubMed] [Google Scholar]
- Silvestri AJ, Root DH. Effects of REM deprivation and an NMDA agonist on the extinction of conditioned fear. Physiol Behav. 2008;93:274–281. doi: 10.1016/j.physbeh.2007.08.020. [DOI] [PubMed] [Google Scholar]
- Soeter M, Kindt M. Stimulation of the noradrenergic system during memory formation impairs extinction learning but not the disruption of reconsolidation. Neuropsychopharmacology. 2012;37:1204–1215. doi: 10.1038/npp.2011.307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soliman F, Glatt CE, Bath KG, Levita L, Jones RM, Pattwell SS, et al. A genetic variant BDNF polymorphism alters extinction learning in both mouse and human. Science. 2010;327:863–866. doi: 10.1126/science.1181886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sotres-Bayon F, Bush DE, LeDoux JE. Acquisition of fear extinction requires activation of NR2B-containing NMDA receptors in the lateral amygdala. Neuropsychopharmacology. 2007;32:1929–1940. doi: 10.1038/sj.npp.1301316. [DOI] [PubMed] [Google Scholar]
- Sotres-Bayon F, Diaz-Mataix L, Bush DE, LeDoux JE. Dissociable roles for the ventromedial prefrontal cortex and amygdala in fear extinction: NR2B contribution. Cereb Cortex. 2009;19:474–482. doi: 10.1093/cercor/bhn099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sotres-Bayon F, Sierra-Mercado D, Pardilla-Delgado E, Quirk GJ. Gating of fear in prelimbic cortex by hippocampal and amygdala inputs. Neuron. 2012;76:804–812. doi: 10.1016/j.neuron.2012.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spennato G, Zerbib C, Mondadori C, Garcia R. Fluoxetine protects hippocampal plasticity during conditioned fear stress and prevents fear learning potentiation. Psychopharmacology (Berl) 2008;196:583–589. doi: 10.1007/s00213-007-0993-7. [DOI] [PubMed] [Google Scholar]
- Stafford JM, Maughan DK, Ilioi EC, Lattal KM. Exposure to a fearful context during periods of memory plasticity impairs extinction via hyperactivation of frontal–amygdalar circuits. Learn Mem. 2013;20:156–163. doi: 10.1101/lm.029801.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steckler T, Risbrough V. Pharmacological treatment of PTSD – established and new approaches. Neuropharmacology. 2012;62:617–627. doi: 10.1016/j.neuropharm.2011.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stewart RE, Chambless DL. Cognitive–behavioral therapy for adult anxiety disorders in clinical practice: a meta-analysis of effectiveness studies. J Consult Clin Psychol. 2009;77:595–606. doi: 10.1037/a0016032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stutzmann GE, LeDoux JE. GABAergic antagonists block the inhibitory effects of serotonin in the lateral amygdala: a mechanism for modulation of sensory inputs related to fear conditioning. J Neurosci. 1999;19:RC8. doi: 10.1523/JNEUROSCI.19-11-j0005.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun N, Laviolette SR. Inactivation of the basolateral amygdala during opiate reward learning disinhibits prelimbic cortical neurons and modulates associative memory extinction. Psychopharmacology (Berl) 2012;222:645–661. doi: 10.1007/s00213-012-2665-5. [DOI] [PubMed] [Google Scholar]
- Surís A, North C, Adinoff B, Powell CM, Greene R. Effects of exogenous glucocorticoid on combat-related PTSD symptoms. Ann Clin Psychiatry. 2010;22:274–279. [PMC free article] [PubMed] [Google Scholar]
- Suzuki A, Josselyn SA, Frankland PW, Masushige S, Silva AJ, Kida S. Memory reconsolidation and extinction have distinct temporal and biochemical signatures. J Neurosci. 2004;24:4787–4795. doi: 10.1523/JNEUROSCI.5491-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sweeney FF, O’Leary OF, Cryan JF. GABAB receptor ligands do not modify conditioned fear responses in BALB/c mice. Behav Brain Res. 2013;256:151–156. doi: 10.1016/j.bbr.2013.07.035. [DOI] [PubMed] [Google Scholar]
- Tang YP, Shimizu E, Dube GR, Rampon C, Kerchner GA, Zhuo M, et al. Genetic enhancement of learning and memory in mice. Nature. 1999;401:63–69. doi: 10.1038/43432. [DOI] [PubMed] [Google Scholar]
- Ter Horst JP, Carobrez AP, van der Mark MH, de Kloet ER, Oitzl MS. Sex differences in fear memory and extinction of mice with forebrain-specific disruption of the mineralocorticoid receptor. Eur J Neurosci. 2012;36:3096–3102. doi: 10.1111/j.1460-9568.2012.08237.x. [DOI] [PubMed] [Google Scholar]
- Terzian AL, Drago F, Wotjak CT, Micale V. The dopamine and cannabinoid interaction in the modulation of emotions and cognition: assessing the role of cannabinoid CB1 receptor in neurons expressing dopamine D1 receptors. Front Behav Neurosci. 2011;5:49. doi: 10.3389/fnbeh.2011.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson BM, Baratta MV, Biedenkapp JC, Rudy JW, Watkins LR, Maier SF. Activation of the infralimbic cortex in a fear context enhances extinction learning. Learn Mem. 2010;17:591–599. doi: 10.1101/lm.1920810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toledo-Rodriguez M, Pitiot A, Paus T, Sandi C. Stress during puberty boosts metabolic activation associated with fear-extinction learning in hippocampus, basal amygdala and cingulate cortex. Neurobiol Learn Mem. 2012;98:93–101. doi: 10.1016/j.nlm.2012.05.006. [DOI] [PubMed] [Google Scholar]
- Tomilenko RA, Dubrovina NI. Effects of activation and blockade of NMDA receptors on the extinction of a conditioned passive avoidance response in mice with different levels of anxiety. Neurosci Behav Physiol. 2007;37:509–515. doi: 10.1007/s11055-007-0044-1. [DOI] [PubMed] [Google Scholar]
- Toth I, Dietz M, Peterlik D, Huber SE, Fendt M, Neumann ID, et al. Pharmacological interference with metabotropic glutamate receptor subtype 7 but not subtype 5 differentially affects within- and between-session extinction of Pavlovian conditioned fear. Neuropharmacology. 2012a;62:1619–1626. doi: 10.1016/j.neuropharm.2011.10.021. [DOI] [PubMed] [Google Scholar]
- Toth I, Neumann ID, Slattery DA. Central administration of oxytocin receptor ligands affects cued fear extinction in rats and mice in a timepoint-dependent manner. Psychopharmacology (Berl) 2012b;223:149–158. doi: 10.1007/s00213-012-2702-4. [DOI] [PubMed] [Google Scholar]
- Trouche S, Sasaki JM, Tu T, Reijmers LG. Fear extinction causes target-specific remodeling of perisomatic inhibitory synapses. Neuron. 2013;80:1054–1065. doi: 10.1016/j.neuron.2013.07.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanelzakker MB, Kathryn Dahlgren M, Caroline Davis F, Dubois S, Shin LM. From Pavlov to PTSD: the extinction of conditioned fear in rodents, humans, and in anxiety disorders. Neurobiol Learn Mem. 2014;113:3–18. doi: 10.1016/j.nlm.2013.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verma D, Tasan RO, Herzog H, Sperk G. NPY controls fear conditioning and fear extinction by combined action on Y(1) and Y(2) receptors. Br J Pharmacol. 2012;166:1461–1473. doi: 10.1111/j.1476-5381.2012.01872.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vertes RP. Differential projections of the infralimbic and prelimbic cortex in the rat. Synapse. 2004;51:32–58. doi: 10.1002/syn.10279. [DOI] [PubMed] [Google Scholar]
- Viviani D, Charlet A, van den Burg E, Robinet C, Hurni N, Abatis M, et al. Oxytocin selectively gates fear responses through distinct outputs from the central amygdala. Science. 2011;333:104–107. doi: 10.1126/science.1201043. [DOI] [PubMed] [Google Scholar]
- Vouimba RM, Maroun M. Learning-induced changes in mPFC-BLA connections after fear conditioning, extinction, and reinstatement of fear. Neuropsychopharmacology. 2011;36:2276–2285. doi: 10.1038/npp.2011.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waddell J, Bouton ME, Falls WA. Central CRF receptor antagonist a-helical CRF9-41 blocks reinstatement of extinguished fear: the role of the bed nucleus of the stria terminalis. Behav Neurosci. 2008;122:1061–1069. doi: 10.1037/a0013136. [DOI] [PubMed] [Google Scholar]
- Walker DL, Ressler KJ, Lu KT, Davis M. Facilitation of conditioned fear extinction by systemic administration or intra-amygdala infusions of D-cycloserine as assessed with fear-potentiated startle in rats. J Neurosci. 2002;22:2343–2351. doi: 10.1523/JNEUROSCI.22-06-02343.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waltereit R, Mannhardt S, Nescholta S, Maser-Gluth C, Bartsch D. Selective and protracted effect of nifedipine on fear memory extinction correlates with induced stress response. Learn Mem. 2008;15:348–356. doi: 10.1101/lm.808608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, Phan T, Storm DR. The type 3 adenylyl cyclase is required for novel object learning and extinction of contextual memory: role of cAMP signaling in primary cilia. J Neurosci. 2011;31:5557–5561. doi: 10.1523/JNEUROSCI.6561-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weber M, Hart J, Richardson R. Effects of D-cycloserine on extinction of learned fear to an olfactory cue. Neurobiol Learn Mem. 2007;87:476–482. doi: 10.1016/j.nlm.2006.12.010. [DOI] [PubMed] [Google Scholar]
- Wei W, Coelho CM, Li X, Marek R, Yan S, Anderson S, et al. p300/CBP-associated factor selectively regulates the extinction of conditioned fear. J Neurosci. 2012;32:11930–11941. doi: 10.1523/JNEUROSCI.0178-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wellman CL, Izquierdo A, Garret JE, Martin KP, Carroll J, Millstein R, et al. Impaired stress-coping and fear extinction and abnormal corticolimbic morphology in serotonin transporter knock-out mice. J Neurosci. 2007;27:684–691. doi: 10.1523/JNEUROSCI.4595-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wellman CL, Camp M, Jones VM, Macpherson KP, Ihne J, Fitzgerald P, et al. Convergent effects of mouse Pet-1 deletion and human PET-1 variation on amygdala fear and threat processing. Exp Neurol. 2013;250C:260–269. doi: 10.1016/j.expneurol.2013.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Werner-Seidler A, Richardson R. Effects of D-cycloserine on extinction: consequences of prior exposure to imipramine. Biol Psychiatry. 2007;62:1195–1197. doi: 10.1016/j.biopsych.2007.04.010. [DOI] [PubMed] [Google Scholar]
- Whittle N, Singewald N. HDAC inhibitors as cognitive enhancers in fear, anxiety and trauma therapy: where do we stand? Biochem Soc Trans. 2014;42:569–581. doi: 10.1042/BST20130233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whittle N, Hauschild M, Lubec G, Holmes A, Singewald N. Rescue of impaired fear extinction and normalization of cortico-amygdala circuit dysfunction in a genetic mouse model by dietary zinc restriction. J Neurosci. 2010;30:13586–13596. doi: 10.1523/JNEUROSCI.0849-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whittle N, Schmuckermair C, Gunduz Cinar O, Hauschild M, Ferraguti F, Holmes A, et al. Deep brain stimulation, histone deacetylase inhibitors and glutamatergic drugs rescue resistance to fear extinction in a genetic mouse model. Neuropharmacology. 2013;64:414–423. doi: 10.1016/j.neuropharm.2012.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilber AA, Southwood CJ, Wellman CL. Brief neonatal maternal separation alters extinction of conditioned fear and corticolimbic glucocorticoid and NMDA receptor expression in adult rats. Dev Neurobiol. 2009;69:73–87. doi: 10.1002/dneu.20691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilber AA, Walker AG, Southwood CJ, Farrell MR, Lin GL, Rebec GV, et al. Chronic stress alters neural activity in medial prefrontal cortex during retrieval of extinction. Neuroscience. 2011;174:115–131. doi: 10.1016/j.neuroscience.2010.10.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson CA, Vazdarjanova A, Terry AV., Jr Exposure to variable prenatal stress in rats: effects on anxiety-related behaviors, innate and contextual fear, and fear extinction. Behav Brain Res. 2013;238:279–288. doi: 10.1016/j.bbr.2012.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wittchen HU, Jacobi F, Rehm J, Gustavsson A, Svensson M, Jonsson B, et al. The size and burden of mental disorders and other disorders of the brain in Europe 2010. Eur Neuropsychopharmacol. 2011;21:655–679. doi: 10.1016/j.euroneuro.2011.07.018. [DOI] [PubMed] [Google Scholar]
- Woods AM, Bouton ME. D-cycloserine facilitates extinction but does not eliminate renewal of the conditioned emotional response. Behav Neurosci. 2006;120:1159–1162. doi: 10.1037/0735-7044.120.5.1159. [DOI] [PubMed] [Google Scholar]
- World Health Organisation. 1994. International classification of diseases (ICD-10)
- Wrubel KM, Barrett D, Shumake J, Johnson SE, Gonzalez-Lima F. Methylene blue facilitates the extinction of fear in an animal model of susceptibility to learned helplessness. Neurobiol Learn Mem. 2007;87:209–217. doi: 10.1016/j.nlm.2006.08.009. [DOI] [PubMed] [Google Scholar]
- Xu J, Zhu Y, Contractor A, Heinemann SF. mGluR5 has a critical role in inhibitory learning. J Neurosci. 2009;29:3676–3684. doi: 10.1523/JNEUROSCI.5716-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu YL, Gall CM, Jackson VR, Civelli O, Reinscheid RK. Distribution of neuropeptide S receptor mRNA and neurochemical characteristics of neuropeptide S-expressing neurons in the rat brain. J Comp Neurol. 2007;500:84–102. doi: 10.1002/cne.21159. [DOI] [PubMed] [Google Scholar]
- Yamada D, Zushida K, Wada K, Sekiguchi M. Pharmacological discrimination of extinction and reconsolidation of contextual fear memory by a potentiator of AMPA receptors. Neuropsychopharmacology. 2009;34:2574–2584. doi: 10.1038/npp.2009.86. [DOI] [PubMed] [Google Scholar]
- Yamada D, Wada K, Sekiguchi M. Facilitating actions of an AMPA receptor potentiator upon extinction of contextually conditioned fear response in stressed mice. Neurosci Lett. 2011;488:242–246. doi: 10.1016/j.neulet.2010.11.038. [DOI] [PubMed] [Google Scholar]
- Yamamoto S, Morinobu S, Fuchikami M, Kurata A, Kozuru T, Yamawaki S. Effects of single prolonged stress and D-cycloserine on contextual fear extinction and hippocampal NMDA receptor expression in a rat model of PTSD. Neuropsychopharmacology. 2008;33:2108–2116. doi: 10.1038/sj.npp.1301605. [DOI] [PubMed] [Google Scholar]
- Yamamoto S, Morinobu S, Takei S, Fuchikami M, Matsuki A, Yamawaki S, et al. Single prolonged stress: toward an animal model of posttraumatic stress disorder. Depress Anxiety. 2009;26:1110–1117. doi: 10.1002/da.20629. [DOI] [PubMed] [Google Scholar]
- Yang CH, Huang CC, Hsu KS. Novelty exploration elicits a reversal of acute stress-induced modulation of hippocampal synaptic plasticity in the rat. J Physiol. 2006;577:601–615. doi: 10.1113/jphysiol.2006.120386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang CH, Shi HS, Zhu WL, Wu P, Sun LL, Si JJ, et al. Venlafaxine facilitates between-session extinction and prevents reinstatement of auditory-cue conditioned fear. Behav Brain Res. 2012;230:268–273. doi: 10.1016/j.bbr.2012.02.023. [DOI] [PubMed] [Google Scholar]
- Yang YL, Lu KT. Facilitation of conditioned fear extinction by d-cycloserine is mediated by mitogen-activated protein kinase and phosphatidylinositol 3-kinase cascades and requires de novo protein synthesis in basolateral nucleus of amygdala. Neuroscience. 2005;134:247–260. doi: 10.1016/j.neuroscience.2005.04.003. [DOI] [PubMed] [Google Scholar]
- Yang YL, Chao PK, Ro LS, Wo YY, Lu KT. Glutamate NMDA receptors within the amygdala participate in the modulatory effect of glucocorticoids on extinction of conditioned fear in rats. Neuropsychopharmacology. 2007;32:1042–1051. doi: 10.1038/sj.npp.1301215. [DOI] [PubMed] [Google Scholar]
- Yang YL, Hsieh CW, Wo YY, Yang YC, Lu KT. Intra-amygdaloid infusion of Ginkgo biloba leaf extract (EGb761) facilitates fear-potentiated startle in rats. Psychopharmacology (Berl) 2009;202:187–196. doi: 10.1007/s00213-008-1138-3. [DOI] [PubMed] [Google Scholar]
- Yehuda R, LeDoux J. Response variation following trauma: a translational neuroscience approach to understanding PTSD. Neuron. 2007;56:19–32. doi: 10.1016/j.neuron.2007.09.006. [DOI] [PubMed] [Google Scholar]
- Yizhar O, Fenno LE, Prigge M, Schneider F, Davidson TJ, O’Shea DJ, et al. Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature. 2011;477:171–178. doi: 10.1038/nature10360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zelikowsky M, Hast TA, Bennett RZ, Merjanian M, Nocera NA, Ponnusamy R, et al. Cholinergic blockade frees fear extinction from its contextual dependency. Biol Psychiatry. 2013;73:345–352. doi: 10.1016/j.biopsych.2012.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang G, Asgeirsdottir HN, Cohen SJ, Munchow AH, Barrera MP, Stackman RW., Jr Stimulation of serotonin 2A receptors facilitates consolidation and extinction of fear memory in C57BL/6J mice. Neuropharmacology. 2013;64:403–413. doi: 10.1016/j.neuropharm.2012.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W, Rosenkranz JA. Repeated restraint stress enhances cue-elicited conditioned freezing and impairs acquisition of extinction in an age-dependent manner. Behav Brain Res. 2013;248:12–24. doi: 10.1016/j.bbr.2013.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng X, Deschaux O, Lavigne J, Nachon O, Cleren C, Moreau JL, et al. Prefrontal high-frequency stimulation prevents sub-conditioning procedure-provoked, but not acute stress-provoked, reemergence of extinguished fear. Neurobiol Learn Mem. 2013;101:33–38. doi: 10.1016/j.nlm.2013.01.003. [DOI] [PubMed] [Google Scholar]
- Zimmerman JM, Maren S. NMDA receptor antagonism in the basolateral but not central amygdala blocks the extinction of Pavlovian fear conditioning in rats. Eur J Neurosci. 2010;31:1664–1670. doi: 10.1111/j.1460-9568.2010.07223.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zushida K, Sakurai M, Wada K, Sekiguchi M. Facilitation of extinction learning for contextual fear memory by PEPA: a potentiator of AMPA receptors. J Neurosci. 2007;27:158–166. doi: 10.1523/JNEUROSCI.3842-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]