ArticlePDF Available

Inactivation of the bed nucleus of the stria terminalis in an animal model of relapse: Effects on conditioned cue-induced reinstatement and its enhancement by yohimbine

Authors:

Abstract and Figures

Drug-associated cues and stress increase craving and lead to greater risk of relapse in abstinent drug users. Animal models of reinstatement of drug seeking have been utilized to study the neural circuitry by which either drug-associated cues or stress exposure elicit drug seeking. Recent evidence has shown a strong enhancing effect of yohimbine stress on subsequent cue-elicited reinstatement; however, there has been no examination of the neural substrates of this interactive effect. The current study examined whether inactivation of the bed nucleus of the stria terminalis (BNST), an area previously implicated in stress activation of drug seeking, would affect reinstatement of cocaine seeking caused by conditioned cues, yohimbine stress, or the combination of these factors. Male rats experienced daily IV cocaine self-administration, followed by extinction of lever responding in the absence of cocaine-paired cues. Reinstatement of responding was measured during presentation of cocaine-paired cues, following pretreatment with the pharmacological stressor, yohimbine (2.5 mg/kg, IP), or the combination of cues and yohimbine. All three conditions led to reinstatement of cocaine seeking, with the highest responding seen after the combination of cues and yohimbine. Reversible inactivation of the BNST using the gamma-aminobutyric acid receptor agonists, baclofen + muscimol, significantly reduced all three forms of reinstatement. These results demonstrate a role for the BNST in cocaine seeking elicited by cocaine-paired cues, and suggest the BNST as a key mediator for the interaction of stress and cues for the reinstatement of cocaine seeking.
Content may be subject to copyright.
Inactivation of the bed nucleus of the stria terminalis in an
animal model of relapse: effects on conditioned cue-induced
reinstatement and its enhancement by yohimbine
Deanne M. Buffalari and
Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
Ronald E. See
Department of Neurosciences, Medical University of South Carolina, BSB416B, 173 Ashley
Avenue, Charleston, SC 29425, USA
Ronald E. See: seere@musc.edu
Abstract
Rationale—Drug-associated cues and stress increase craving and lead to greater risk of relapse in
abstinent drug users. Animal models of reinstatement of drug seeking have been utilized to study
the neural circuitry by which either drug-associated cues or stress exposure elicit drug seeking.
Recent evidence has shown a strong enhancing effect of yohimbine stress on subsequent cue-
elicited reinstatement; however, there has been no examination of the neural substrates of this
interactive effect.
Objectives—The current study examined whether inactivation of the bed nucleus of the stria
terminalis (BNST), an area previously implicated in stress activation of drug seeking, would affect
reinstatement of cocaine seeking caused by conditioned cues, yohimbine stress, or the combination
of these factors.
Methods—Male rats experienced daily IV cocaine self-administration, followed by extinction of
lever responding in the absence of cocaine-paired cues. Reinstatement of responding was
measured during presentation of cocaine-paired cues, following pretreatment with the
pharmacological stressor, yohimbine (2.5 mg/kg, IP), or the combination of cues and yohimbine.
Results—All three conditions led to reinstatement of cocaine seeking, with the highest
responding seen after the combination of cues and yohimbine. Reversible inactivation of the
BNST using the gamma-aminobutyric acid receptor agonists, baclofen+muscimol, significantly
reduced all three forms of reinstatement.
Conclusion—These results demonstrate a role for the BNST in cocaine seeking elicited by
cocaine-paired cues, and suggest the BNST as a key mediator for the interaction of stress and cues
for the reinstatement of cocaine seeking.
Keywords
Cocaine; Yohimbine; Reinstatement; Relapse; Bed nucleus stria terminalis; Norepinephrine;
Neurocircuitry; Stress; Conditioned; Self-administration
© Springer-Verlag 2010
Correspondence to: Ronald E. See, seere@musc.edu.
NIH Public Access
Author Manuscript
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
Published in final edited form as:
Psychopharmacology (Berl)
. 2011 January ; 213(1): 19–27. doi:10.1007/s00213-010-2008-3.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Introduction
Treatment of cocaine addiction is impeded by high rates of relapse to drug seeking and drug
taking in chronic users. Relapse can be triggered by many factors, including exposure to
drug-associated cues and contexts (such as drug paraphernalia or drug-associated
environments) or stressful life events that occur during periods of abstinence. Clinical
laboratory studies have shown that cocaine-associated stimuli increase craving in abstinent
users (Childress et al. 1993), as does exposure to stress-provoking stimuli (Sinha et al.
1999). The triggering of relapse in abstinent users has been modeled in animals using the
self-administration and reinstatement paradigm. Animals trained to self-administer cocaine
will reinstate responding on the previously drug-paired lever after exposure to cocaine-
associated cues (See 2002), stress (Shalev et al. 2000), or a priming dose of cocaine (de Wit
and Stewart 1981). The reinstatement model of relapse (Katz and Higgins 2003) has allowed
for the investigation of the neural circuitry underlying cocaine-seeking behavior triggered by
each of these factors. Evidence to date suggests that cues and stress share some of the same
neural pathways in mediating reinstatement of cocaine seeking, most notably the nucleus
accumbens core and the dorsomedial prefrontal cortex (Capriles et al. 2003; Fuchs et al.
2004a; McFarland et al. 2004; McLaughlin and See 2003). However, some structures (e.g.,
basolateral amygdala) have been found to be important for cue- but not footshock stress-
induced reinstatement of cocaine seeking (McFarland et al. 2004; Meil and See 1997).
While almost all previous studies of the triggering events that initiate reinstatement of drug
seeking have limited their focus to separate factors, we have recently demonstrated that
footshock or yohimbine stress activation potentiates conditioned cue-induced reinstatement
of cocaine seeking (Buffalari and See 2009a; Feltenstein and See 2006). The mechanisms by
which this interaction occurs likely involve a convergence of activity in the neural pathways
that reinitiate drug seeking. Among the structures that mediate drug seeking, the bed nucleus
of the stria terminalis (BNST) is a key component of the extended amygdala that may be a
critical point of convergence for cue and stress interactions. Studies of the BNST have
indicated that it plays a key role in addictive drug actions, including cocaine (Dumont et al.
2005; Kash et al. 2008). Inactivation of the BNST with sodium channel blockers (Erb and
Stewart 1999) or gamma-aminobutyric acid (GABA) receptor agonists (McFarland et al.
2004), as well as beta norepinephrine (NE) receptor antagonism (Leri et al. 2002), have been
shown to block reinstatement of cocaine seeking caused by acute footshock stress. However,
it is not yet known if the BNST plays a role in conditioned cue-induced reinstatement of
cocaine seeking, or reinstatement caused by other stressors.
Several recent studies have successfully used systemic injections of yohimbine to trigger
stress-induced reinstatement of drug seeking in rats (Banna et al. 2010; Bongiovanni and
See 2008; Feltenstein and See 2006; Shepard et al. 2004) and nonhuman primates (Lee et al.
2004), and elicit drug craving in human addicts (Stine et al. 2002). Yohimbine increases NE
in terminal regions via antagonism of α-2 NE receptors (Galvez et al. 1996; Tjurmina et al.
1999). However, nothing is yet known about the neural circuitry underlying reinstatement
caused by systemic yohimbine, or any other drugs that may act as stressors. Both
intermittent footshock (Galvez et al. 1996) and yohimbine (Forray et al. 1997) increase NE
in the amygdala, including the extended amygdala and BNST, and NE receptor blockade in
the BNST disrupts cocaine seeking caused by footshock stress (Leri et al. 2002), supporting
the possibility that stress activation via yohimbine may rely on intact BNST function.
Therefore, in the current study, we examined the role of the BNST in reinstatement of
cocaine seeking caused by yohimbine-induced stress alone, conditioned cues alone, or a
combination of yohimbine and cues in rats with a history of chronic cocaine self-
administration and extinction. Based on the reported role of the BNST in both stress
Buffalari and See Page 2
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
activation and drug seeking, we predicted that the BNST would be critical for mediating
stress-induced reinstatement via yohimbine, as well as the enhancing effects of yohimbine
on conditioned cue-induced reinstatement of cocaine seeking.
Materials and methods
Subjects
Male Sprague–Dawley rats (initial weight 275–300 g; Charles River, Wilmington, MA,
USA) were individually housed in a temperature- and humidity-controlled vivarium on a
reverse 12-h light–dark cycle (lights on 6 PM–6 AM). Animals received water and standard
rat chow (Harlan, Indianapolis, IN, USA) ad libitum, with the exception of 2–3 days of food
restriction during initial cocaine self-administration (animals never received <10 g food/
day). Housing and care of the rats were carried out in accordance with the National Institute
of Health Guide for the Care and Use of Laboratory Animals (NIH Publications No. 80–23)
revised 1996.
Surgery
Rats received chronically indwelling catheters into the right jugular vein as previously
described (Fuchs et al. 2004b). Briefly, male rats were anesthetized using a mixture of
ketamine hydrochloride and xylazine (66 and 1.33 mg/kg, respectively, IP) followed by
equithesin (0.5 ml/kg, IP). Catheters (constructed using previously described methods;
Fuchs et al. 2004b) were inserted into the right jugular vein. To maintain patency, catheters
were flushed with heparin and cefazolin solutions daily. Immediately following catheter
surgery, animals were placed into a stereotaxic frame (Stoelting, Wood Dale, IL, USA).
Bilateral stainless steel guide cannulae (26 gauge; Plastics One, Inc.) were inserted dorsal to
the BNST (±3.5 M/L, 0.4 A/P, 4.6 D/V, 15° angle). Three small screws and cranioplastic
cement secured the guide cannulae to the skull. Stylets (Plastics One, Inc.) were placed into
the guide cannulae and catheter to prevent occlusions. To verify catheter patency, rats
occasionally received a 0.12-ml infusion of methohexital sodium (10.0 mg/ml IV; Eli Lilly
and Co., Indianapolis, IN, USA), a short-acting barbiturate that produces a rapid loss of
muscle tone when administered intravenously.
Cocaine self-administration
Rats self-administered cocaine (cocaine hydrochloride dissolved in 0.9% sterile saline;
cocaine provided by the National Institute on Drug Abuse, Research Triangle Park, NC,
USA) during daily 2-h sessions according to a fixed ratio-1 (FR 1) schedule of
reinforcement as previously described (Feltenstein et al. 2007b; Fuchs et al. 2004b). Briefly,
lever presses on the active lever resulted in cocaine infusions along with presentation of a
light+tone complex and were followed by a 20-s timeout. Inactive lever presses had no
consequences, but were recorded. Daily cocaine self-administration continued until each rat
had obtained the self-administration criterion of 10 sessions with at least 10 infusions per
session.
Extinction and reinstatement of cocaine seeking
Following chronic self-administration and before the first reinstatement test, rats underwent
daily 2-h extinction sessions as previously described (Feltenstein et al. 2007a; Fuchs et al.
2004b). Once extinction criterion was reached (defined here as a minimum of seven
extinction sessions with 15 active lever responses per session for the last two consecutive
days before testing), each rat underwent six separate reinstatement tests. Prior studies have
successfully utilized similar repeated reinstatement testing designs (Feltenstein and See
2006; Kippin et al. 2006). Rats experienced six total reinstatement tests examining three
Buffalari and See Page 3
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
reinstatement factors before and after intracranial vehicle and B/M infusions. The following
three reinstatement triggers were used: conditioned cues, yohimbine administration (IP), and
conditioned cues+yohimbine. Each test was given twice with either vehicle or baclofen–
muscimol (B/M) infusions into the BNST immediately prior to the test. Both drug treatment
(vehicle or B/M) and reinstatement test type were counterbalanced. Animals that did not
finish all six reinstatement tests (e.g., due to cannulae blockade) were not utilized for data
analysis. Animals were extinguished to criterion between reinstatement tests (15 active
lever responses per session for two consecutive days). Yohimbine injections (2.5 mg/kg, IP)
were given 30 min prior to testing, and saline vehicle was given prior to conditioned cue
reinstatement tests. The yohimbine dose was based on previous reinstatement studies in rats
(Feltenstein and See 2006; Shepard et al. 2004). Conditioned cue-induced reinstatement tests
involved contingent presentation of the light+tone stimulus previously associated with the
active lever press during self-administration. Cue presentation was followed by a 20-s time-
out, during which lever presses were recorded, but had no programmed consequences.
Intracranial infusions
For intracranial infusions, stainless steel injection cannulae (33 gauge, Plastics One) were
inserted to a depth of 2 mm below the tip of the guide cannulae immediately prior to
placement into the chamber. The injection cannulae were connected to 10-ml Hamilton
syringes (Hamilton Co., Reno, NV, USA) mounted on an infusion pump (Harvard
Apparatus, South Natick, MA, USA). A combination of B/M (1.0/0.1 mM, 0.2 ul volume) or
phosphate-buffered saline vehicle (pH=7.0 for both solutions) was infused bilaterally over a
2-min time period. Dose–response analyses have shown that this concentration of B/M site-
selectively attenuates cocaine-primed (McFarland and Kalivas 2001) or conditioned cue-
induced reinstatement (Fuchs et al. 2004b) of cocaine seeking, and this dose of B/M has
previously been used to selectively inactivate the BNST (Rogers et al. 2008). The injection
cannulae were left in place for 1 min prior to and after the infusion.
To assess the effects of B/M inactivation on general motor activity, subsequent to
completion of reinstatement testing, a subset of animals were tested for locomotor activity
after vehicle or B/M infusions (n=10/group) into the BNST. Infusions occurred immediately
before placement into clear Plexiglas chambers (22×43×33 cm). Each chamber was
equipped with a Digiscan monitor (Omnitech Electronics, Columbus, OH, USA) containing
a series of 16 photobeams (eight on each horizontal axis) that tabulated total distance (cm)
traveled by each animal. Beam breaks were detected by a Digiscan analyzer and recorded by
DigiPro software (Verson 1.4). Immediately following a 1-h testing period, animals were
returned to their home cages.
Histology and data analysis
After completion of reinstatement testing, the rats were deeply anesthetized with equithesin
and transcardially perfused with PBS and 10% formaldehyde solution and processed as
previously described (Fuchs et al. 2004b). The most ventral point of the microinjector tips
were mapped onto schematics from a rat brain atlas (Paxinos and Watson 1997).
Reinstatement of responding from extinction levels and the effects of B/M inhibition of the
BNST on reinstatement were analyzed using separate one-way analysis of variance
(ANOVA) for each reinstatement condition, followed by pairwise comparisons with the
Student–Newman–Keuls test. Locomotor activity was measured in 12 5 min time bins and
analyzed with a two-way ANOVA (group×time). Data points were not included if they were
three standard deviations beyond the group mean. Analyses were considered statistically
significant at p<0.05. All data are presented as mean±SEM.
Buffalari and See Page 4
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Results
Histology
Inspection of injection cannulae tip locations showed that all correct BNST placements were
within the dorsomedial portions of the more posterior BNST, including the anterolateral and
anteromedial nuclei (Fig. 1a). A representative photomicrograph is shown in Fig. 1b. Note
that due to overlap, some points indicate injector locations for multiple animals. A total of
n=18 animals had correct injector placements in the BNST. Animals (n=8) with inaccurate
cannulae placements served as anatomical controls to validate the specificity of BNST
inactivation on reinstatement. These placements targeted the anterior commissure at various
rostral/caudal levels, as well as the internal capsule and the nucleus of the commissural stria
terminalis. It is important to note that the animals with placement outside the BNST did not
show any effect of B/M treatment on reinstatement responding (shown below).
Cocaine self-administration and extinction
Rats readily acquired cocaine self-administration and demonstrated stable patterns of active
lever responding and cocaine intake throughout the maintenance phase of the experiment.
Figure 2a indicates the mean lever responding for the last 2 days of cocaine self-
administration before extinction and reinstatement testing (mean active lever
responses=54.5±14.3 and mean daily cocaine intake= 20.7±2.3 mg/kg). Furthermore, all
animals decreased responding during extinction sessions in the absence of cocaine infusions
and cue presentations (Fig. 2b). Active lever responding reached the established extinction
criterion (<15 active lever responses over 2 days) in a mean of 8.7± 0.4 days before
subsequent reinstatement testing. Mean lever presses before the first reinstatement test was
measured across animals (mean active lever responses= 9.5±1.1). Inactive lever responding
showed uniformly very low levels throughout the study, and no significant differences were
found between treatment conditions during reinstatement trials (data not shown).
Spontaneous locomotor activity measured after intracranial infusions of either vehicle or B/
M (n=10/group) showed no significant effects of BNST inactivation on total locomotor
activity (data not shown).
Reinstatement testing
Reinstatement data for animals with correct placements in the BNST are shown in Fig. 3.
Four out of 162 datapoints were excluded on the basis of outlier criterion (>3 SD).
Presentation of conditioned cues led to significant reinstatement of responding on the
previously cocaine-paired lever during testing (F2,51=18.28, p<0.0001). Post hoc analyses
revealed significant reinstatement over extinction levels (p<0.05) and blockade of
conditioned cue reinstatement by intracranial B/M administration (p<0.05). Yohimbine
administration alone led to modest, but significantly increased reinstatement responding
(F2,48=3.94, p<0.05) above that of extinction levels (p<0.05). Yohimbine-induced
reinstatement was significantly attenuated by BNST inactivation (p<0.05). As previously
reported (Feltenstein and See 2006), pretreatment with yohimbine increased cue-induced
reinstatement of cocaine seeking beyond that seen with either cues or yohimbine alone.
Analyses revealed a significant main effect (F2,50=17.32, p<0.0001), with pronounced
reinstatement over extinction levels (p<0.05) as well as inhibition of the cue+yohimbine-
induced reinstatement by B/M (p<0.05). In animals with cannulae placements outside the
BNST, infusion of B/M had no effect on reinstatement of cocaine seeking under any of the
three conditions (Fig. 4), with significant responding over extinction levels seen for cue-,
yohimbine-, and cue+ yohimbine-induced reinstatement after either vehicle or B/M
infusions (p<0.05).
Buffalari and See Page 5
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Discussion
The current study shows a role for the BNST in both conditioned cue and yohimbine-
induced reinstatement of cocaine seeking in an animal model of relapse. Further, our results
effectively demonstrate blockade of the interactive effects of yohimbine stress and cues on
reinstatement, supporting the role of the BNST as a critical modulator when both types of
reinstatement factors are applied. While the role of the BNST in footshock stress-induced
reinstatement has previously been established (Erb and Stewart 1999; McFarland et al.
2004), examination of the neural substrates of conditioned cue-induced reinstatement has
focused primarily on the basolateral and central regions of the amygdala, rather than the
extended amygdala (See 2005). In addition to a role for the BNST in conditioned cue-
induced reinstatement, our results indicate that the BNST is critical for the interaction of
yohimbine stress and cues in the promotion of cocaine seeking, as BNST inactivation
blocked this potent form of reinstatement. These results suggest that stress-cue interactions
may be the result of the convergent activation of two separate, yet integrated stress and cue
pathways that include the BNST.
Previous studies examining the neural circuitry underlying stress-induced reinstatement have
generally utilized intermittent footshock stress (Ahmed and Koob 1997; Erb et al. 1996; Erb
and Stewart 1999). Several other forms of stress (e.g., restraint stress) have been found to
not produce reinstatement of drug seeking (Shaham et al. 2000). To date, studies have not
directly examined the neural circuitry underlying yohimbine stress-induced reinstatement, or
the degree to which patterns of neural activation may be similar during footshock or
yohimbine stress-induced reinstatement. Interestingly, both of these stressors (but not
several other types of stressors) induced similar increases in c-fos mRNA in the nucleus
accumbens shell, and the basolateral and central amygdalar nuclei (Funk et al. 2006).
However, other data indicate that yohimbine may promote reinstatement via mechanisms
distinct from those underlying footshock stress-induced reinstatement. While footshock
stress-induced reinstatement is thought to rely on interactions of CRF and NE within the
BNST (Leri et al. 2002), recent studies suggest that yohimbine stress-induced reinstatement
is unaffected by CRF receptor antagonists, or by NE α-2 agonists (Brown et al. 2009).
However, we have recently found blockade of yohimbine-induced and conditioned cue-
induced reinstatement by systemic administration of guanfacine, an α-2 receptor agonist
(Buffalari and See 2009b). Furthermore, the current results suggest that yohimbine and
footshock do both rely on intact function of the bed nucleus of the stria terminalis.
While yohimbine has been well characterized as an enhancer of NE activity via NE α-2
antagonist effects, other mechanisms in both the BNST and other brain regions may
contribute to the effects of the drug, in particular the enhancing effect of yohimbine on cues.
Yohimbine does have some affinity at serotonin (Millan et al. 2000) and dopamine (Scatton
et al. 1980) receptor subtypes, any of which may contribute to reinstatement of drug seeking.
Alternatively, NE increases in other terminal regions may contribute to reinstatement by
both yohimbine stress alone and the enhanced reinstatement with simultaneous stress and
cues. Yohimbine-induced increases in NE release in the basolateral amygdala (Buffalari and
Grace 2009) may enhance the motivational salience of conditioned cues, while increased
prefrontal NE tone (Garcia et al. 2004) could alter attentional processes and further
modulate cue salience.
While yohimbine does not induce a stress state fully analogous to an external stress situation
(e.g., social stress), the use of yohimbine as a stressor for an animal model of relapse offers
several experimental and translational advantages. First, yohimbine has well-characterized
stress and anxiety effects in humans (Southwick et al. 1999) including increased anxiety and
activation of the hypothalamic–pituitary adrenal axis (Charney et al. 1987), elicitation of
Buffalari and See Page 6
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
panic attacks, and increases in blood pressure (Vasa et al. 2009). Yohimbine causes stress
and anxiety responses in animals as well, including increased cortisol in rats (Banihashemi
and Rinaman 2006) and monkeys (Lee et al. 2004), and increased anxiety-like behaviors in
several paradigms (File 1986; Johnston and File 1989; Bijlsma et al. 2010). Yohimbine
therefore offers a homologous method of stress activation across species. Second, while not
yet systematically tested for cocaine, yohimbine increases drug craving in opioid-dependent
patients (Stine et al. 2002). Also, yohimbine reliably reinstates drug-seeking behavior in rats
(Banna et al. 2010; Feltenstein and See 2006; Le et al. 2005; Shepard et al. 2004) and
monkeys (Lee et al. 2004). Finally, yohimbine has a relatively long half-life of several hours
(Hubbard et al. 1988), which allows for maintained stress activation across the duration of a
reinstatement test session, with or without conditioned cue exposure. Further exploration of
both the neural circuitry and pharmacological features of yohimbine stress-induced
reinstatement offers a promising direction for future studies.
While previous studies have delineated various aspects of the neural circuitry underlying
conditioned cue-induced reinstatement (Feltenstein and See 2008; See 2005), further studies
will need to establish the links between amygdalar-based circuitry and the monoaminergic
modulation of the corticostriatal circuitry necessary for reinstatement of drug seeking. The
lack of a direct projection from the basolateral amygdala to the ventral tegmental area
(VTA) suggests that the amygdala may modulate dopaminergic output in the prefrontal
cortex via direct projections to the prefrontal cortex or indirect projections via the central
amygdala regions and the BNST (Alheid 2003), which have glutamatergic, GABAergic, and
peptidergic projections to dopamine neurons (Georges and Aston-Jones 2001; Morrell et al.
1984). Cue presentation may activate basolateral amygdala efferents to these regions,
resulting in enhanced dopamine release in the prefrontal cortex. The importance of cortical
dopamine function is evidenced from studies that have shown attenuation of both
conditioned cue- (Ciccocioppo et al. 2001; See 2009) and footshock stress-induced (Capriles
et al. 2003) reinstatement by dopamine receptor blockade. The BNST may modulate cortical
dopamine levels via its projections to the VTA (Georges and Aston-Jones 2001, 2002).
Multiple changes in the BNST have been characterized after acute or repeated
noncontingent cocaine, as well as cocaine self-administration (Koob 2003). Cocaine has
been reported to increase dopamine in the BNST (Carboni et al. 2000) and enhance BNST
excitatory transmission (Dumont et al. 2005). Dopamine has also been shown to enhance
glutamatergic transmission in the BNST and modulate short-term NMDA-dependent
plasticity (Kash et al. 2008). Plasticity within the neural circuitry associated with
reinstatement has been implicated as a crucial component of cocaine-induced behavioral
changes that may underlie relapse (Kauer and Malenka 2007). Further, chronic cocaine
disrupts plasticity within the BNST, and leads to changes in NE transporter binding. NE in
the BNST, which is critical for footshock-induced reinstatement (Leri et al. 2002), causes
complex inhibitory and excitatory effects that are receptor and subregion dependent (Egli et
al. 2005). Finally, the BNST has been heavily implicated in withdrawal from abused drugs
(Koob 2003; Smith and Aston-Jones 2008), and withdrawal also disrupts long-term
potentiation of intrinsic excitability within the BNST (Francesconi et al. 2009). Such studies
complement the current results and suggest that the BNST is a critical region that may
undergo changes relevant to chronic drug use, craving, withdrawal, and relapse.
The BNST is a complex brain region with as many as 12 anatomically identified subregions
(Dumont 2009). However, the functional dissociation of different regions of the BNST,
particularly with regard to reward and addiction-related functions, has yet to be delineated.
Our placements primarily targeted the dorsomedial subdivisions, primarily in the posterior
portions of BNST. However, this placement was not exclusive, and inactivation of other
areas of the BNST was also effective. This is not surprising, in that other studies examining
Buffalari and See Page 7
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the role of the BNST in drug withdrawal, reinstatement, and reward have identified a role
for multiple subregions of the BNST (Aston-Jones et al. 1999; Delfs et al. 2000; Harris and
Aston-Jones 2003; Jalabert et al. 2009; Leri et al. 2002). Most recently, strong interactions
between the prefrontal cortex, multiple BNST subregions, and dopamine neurons of the
VTA have been identified (Jalabert et al. 2009). These interactions are relevant to the
current results, as the VTA and prefrontal cortical regions have been shown to play a role in
other forms of reinstatement of drug seeking (McFarland et al. 2004; See 2005).
We chose B/M infusions as a means of inactivation for four primary reasons: (1) sparing of
effects on fibers of passage, (2) targeting of both GABA A and B receptors, (3) extensive
previous research with this approach in determining the neural circuitry of reinstatement
(McFarland et al. 2004; McFarland and Kalivas 2001), and (4) previous experience in our
laboratory using B/M to specifically inactivate the BNST. However, as with any
pharmacological approach, the use of B/M necessitates careful interpretation of the resultant
behavioral effects. Often characterized as consisting of GABAergic interneurons and
projection neurons, recent investigations (Jalabert et al. 2009) have suggested that this may
be an oversimplification of the BNST. While combined GABA A and B agonists generally
result in an overall inhibition, in a structure with many interconnected GABAergic
interneurons (such as the BNST), more complex interactions could arise. Follow-up studies
will need to target specific neurotransmitter receptors, with a primary focus on NE receptors
due to yohimbine’s actions on the NE system and prior evidence of a role for BNST NE in
footshock-induced reinstatement of drug seeking (Leri et al. 2002).
The current results have implications for the phenomena whereby heightened stress
increases the likelihood of relapse in drug-dependent individuals in isolation, or when they
experience stimuli or environments associated with prior drug use (Sinha et al. 2006).
Whereas previous clinical and preclinical studies have almost exclusively focused on the
isolated effects of stress or cues, users usually encounter multiple triggers for relapse during
abstinence, and are more at risk during periods of greater stress or other maladaptive states,
such as anxiety or depression (Brady et al. 2007). Thus, the use of animal models aimed at
understanding the neurobiology of relapse need to incorporate the interactions that occur
between factors that promote relapse. The discovery of novel neural substrates underlying
the interaction of stress and cues in reinstatement will ultimately facilitate the development
of more effective interventions that can successfully target both domains that contribute to
relapse.
Acknowledgments
This research was supported by the National Institute on Drug Abuse grants DA16511 and DA21690 (RES), 1F32
DA025411-01 (DMB), and NIH grant C06 RR015455. The authors thank Anthony Carnell, Alisha Henderson, and
Bernard Smalls for technical assistance and data collection.
References
Ahmed SH, Koob GF. Cocaine- but not food-seeking behavior is reinstated by stress after extinction.
Psychopharmacology. 1997; 132:289–295. [PubMed: 9292629]
Alheid GF. Extended amygdala and basal forebrain. Ann NY Acad Sci. 2003; 985:185–205. [PubMed:
12724159]
Aston-Jones G, Delfs JM, Druhan J, Zhu Y. The bed nucleus of the stria terminalis. A target site for
noradrenergic actions in opiate withdrawal. Ann N Y Acad Sci. 1999; 877:486–498. [PubMed:
10415666]
Banihashemi L, Rinaman L. Noradrenergic inputs to the bed nucleus of the stria terminalis and
paraventricular nucleus of the hypothalamus underlie the hypothalamic–pituitary–adrenal axis but
Buffalari and See Page 8
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
not hypophagic or conditioned avoidance responses to system yohimbine. J Neurosci. 2006;
26:11442–11453. [PubMed: 17079674]
Banna KM, Back SE, Do P, See RE. Yohimbine stress potentiates conditioned cue-induced
reinstatement of heroin-seeking in rats. Behav Brain Res. 2010; 208:144–148. [PubMed: 19931573]
Bijlsma EY, de Jongh R, Olivier B, Groenink L. Fear-potentiated startle, but not light-enhanced startle,
is enhanced by anxiogenic drugs. Pharmacol Biochem Behav. 2010; 96:24–31. [PubMed:
20394767]
Bongiovanni M, See RE. A comparison of the effects of different operant training experiences and
dietary restriction on the reinstatement of cocaine-seeking in rats. Pharmacol Biochem Behav. 2008;
89:227–233. [PubMed: 18230406]
Brady KT, Verduin ML, Tolliver BK. Treatment of patients comorbid for addiction and other
psychiatric disorders. Curr Psychiatry Rep. 2007; 9:374–380. [PubMed: 17915076]
Brown ZJ, Tribe E, D’Souza NA, Erb S. Interaction between noradrenaline and corticotrophin-
releasing factor in the reinstatement of cocaine seeking in the rat. Psychopharmacology. 2009;
203:121–130. [PubMed: 18985323]
Buffalari DM, Grace AA. Anxiogenic modulation of spontaneous and evoked neuronal activity in the
basolateral amygdala. Neuroscience. 2009; 163:1069–1077. [PubMed: 19589368]
Buffalari DM, See RE. Footshock stress potentiates cue-induced cocaine-seeking in an animal model
of relapse. Physiol Behav. 2009a; 98:614–617. [PubMed: 19800355]
Buffalari DM, See RE. Guanfacine blockade of stress-induced and conditioend cue-induced cocaine-
seeking in an animal model of relapse. Soc Neurosci Abstr. 2009b:387.5.
Capriles N, Rodaros D, Sorge RE, Stewart J. A role for the prefrontal cortex in stress- and cocaine-
induced reinstatement of cocaine seeking in rats. Psychopharmacology. 2003; 168:66–74.
[PubMed: 12442201]
Carboni E, Silvagni A, Rolando MT, Di Chiara G. Stimulation of in vivo dopamine transmission in the
bed nucleus of stria terminalis by reinforcing drugs. J Neurosci. 2000; 20:RC102. [PubMed:
11027253]
Charney DS, Woods SW, Goodman WK, Heninger GR. Neurobiological mechanisms of panic
anxiety: biochemical and behavioral correlates of yohimbine-induced panic attacks. Am J
Psychiatr. 1987; 144:1030–1036. [PubMed: 3037926]
Childress AR, Hole AV, Ehrman RN, Robbins SJ, McLellan AT, O’Brien CP. Cue reactivity and cue
reactivity interventions in drug dependence. NIDA Res Monogr. 1993; 137:73–95. [PubMed:
8289929]
Ciccocioppo R, Sanna PP, Weiss F. Cocaine-predictive stimulus induces drug-seeking behavior and
neural activation in limbic brain regions after multiple months of abstinence: Reversal by D1
antagonists. Proc Natl Acad Sci U S A. 2001; 98:1976–1981. [PubMed: 11172061]
Delfs JM, Zhu Y, Druhan JP, Aston-Jones G. Noradrenaline in the ventral forebrain is critical for
opiate withdrawal-induced aversion. Nature. 2000; 403:430–434. [PubMed: 10667795]
de Wit H, Stewart J. Reinstatement of cocaine-reinforced responding in the rat. Psychopharmacology.
1981; 75:134–143. [PubMed: 6798603]
Dumont EC. What is the bed nucleus of the stria terminalis? Prog Neuro-Psychopharmacol Biol
Psychiatry. 2009; 33:1289–1290.
Dumont EC, Mark GP, Mader S, Williams JT. Self-administration enhances excitatory synaptic
transmission in the bed nucleus of the stria terminalis. Nat Neurosci. 2005; 8:413–414. [PubMed:
15735642]
Egli RE, Kash TL, Choo K, Savchenko V, Matthews RT, Blakely RD, Winder DG. Norepinephrine
modulates glutamatergic transmission in the bed nucleus of the stria terminalis. Neuro-
psychopharmacology. 2005; 30:657–668.
Erb S, Stewart J. A role for the bed nucleus of the stria terminalis, but not the amygdala, in the effects
of corticotropin-releasing factor on stress-induced reinstatement of cocaine seeking. J Neurosci.
1999; 19:RC35. [PubMed: 10516337]
Erb S, Shaham Y, Stewart J. Stress reinstates cocaine-seeking behavior after prolonged extinction and
a drug-free period. Psychopharmacology. 1996; 128:408–412. [PubMed: 8986011]
Buffalari and See Page 9
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feltenstein MW, See RE. Potentiation of cue-induced reinstatement of cocaine-seeking in rats by the
anxiogenic drug yohimbine. Behav Brain Res. 2006; 174:1–8. [PubMed: 16920204]
Feltenstein MW, See RE. The neurocircuitry of addiction: an overview. Br J Pharmacol. 2008;
154:261–274. [PubMed: 18311189]
Feltenstein MW, Altar CA, See RE. Aripiprazole blocks reinstatement of cocaine seeking in an animal
model of relapse. Biol Psychiatry. 2007; 61:582–590. [PubMed: 16806092]
File SE. Aversive and appetitive properties of anxiogenic and anxiolytic agents. Behav Brain Res.
1986; 21:189–194. [PubMed: 2876714]
Forray MI, Bustos G, Gysling K. Regulation of norepinephrine release from the rat bed nucleus of the
stria terminalis: in vivo microdialysis studies. J Neurosci Res. 1997; 50:1040–1046. [PubMed:
9452019]
Francesconi W, Berton F, Repunte-Canonigo V, Hagihara K, Thurbon D, Lekic D, Specio SE,
Greenwell TN, Chen SA, Rice KC, Richardson HN, O’Dell LE, Zorrilla EP, Morales M, Koob
GF, Sanna PP. Protracted withdrawal from alcohol and drugs of abuse impairs long-term
potentiation of intrinsic excitability in the juxtacapsular bed nucleus of the stria terminalis. J
Neurosci. 2009; 29:5389–5401. [PubMed: 19403807]
Fuchs RA, Evans KA, Parker MC, See RE. Differential involvement of the core and shell subregions
of the nucleus accumbens in conditioned cue-induced reinstatement of cocaine seeking in rats.
Psychopharmacology. 2004a; 176:459–465. [PubMed: 15138757]
Fuchs RA, Evans KA, Parker MP, See RE. Differential involvement of orbitofrontal cortex subregions
in conditioned cue-induced and cocaine-primed reinstatement of cocaine seeking in rats. J
Neurosci. 2004b; 24:6600–6610. [PubMed: 15269272]
Funk D, Li Z, Le AD. Effects of environmental and pharmacological stressors on c-fos and
corticotropin-releasing factor mRNA in rat brain: relationship to the reinstatement of alcohol
seeking. Neuroscience. 2006; 138:235–243. [PubMed: 16359808]
Galvez R, Mesches MH, McGaugh JL. Norepinephrine release in the amygdala in response to
footshock stimulation. Neurobiol Learn Mem. 1996; 66:253–257. [PubMed: 8946419]
Garcia AS, Barrera G, Burke TF, Ma S, Hensler JG, Morilak DA. Autoreceptor-mediated inhibition of
norepinephrine release in rat medial prefrontal cortex is maintained after chronic desipramine
treatment. J Neurochem. 2004; 91:683–693. [PubMed: 15485498]
Georges F, Aston-Jones G. Potent regulation of midbrain dopamine neurons by the bed nucleus of the
stria terminalis. J Neurosci. 2001; 21:RC160. [PubMed: 11473131]
Georges F, Aston-Jones G. Activation of ventral tegmental area cells by the bed nucleus of the stria
terminalis: a novel excitatory amino acid input to midbrain dopamine neurons. J Neurosci. 2002;
22:5173–5187. [PubMed: 12077212]
Harris GC, Aston-Jones G. Critical role for ventral tegmental glutamate in preference for a cocaine-
conditioned environment. Neuropsychopharmacology. 2003; 28:73–76. [PubMed: 12496942]
Hubbard JW, Pfister SL, Biediger AM, Herzig TC, Keeton TK. The pharmacokinetic properties of
yohimbine in the conscious rat. Naunyn Schmiedebergs Arch Pharmacol. 1988; 337:583–587.
[PubMed: 3412496]
Jalabert M, Aston-Jones G, Herzog E, Manzoni O, Georges F. Role of the bed nucleus of the stria
terminalis in the control of ventral tegmental area dopamine neurons. Prog Neuro-
Psychopharmacol Biol Psychiatry. 2009; 33:1336–1346.
Johnston AL, File SE. Yohimbine’s anxiogenic action: evidence for noradrenergic and dopaminergic
sites. Pharmacol Biochem Behav. 1989; 32:151–156. [PubMed: 2567522]
Kash TL, Nobis WP, Matthews RT, Winder DG. Dopamine enhances fast excitatory synaptic
transmission in the extended amygdala by a CRF-R1-dependent process. J Neurosci. 2008;
28:13856–13865. [PubMed: 19091975]
Katz JL, Higgins ST. The validity of the reinstatement model of craving and relapse to drug use.
Psychopharmacology. 2003; 168:21–30. [PubMed: 12695875]
Kauer JA, Malenka RC. Synaptic plasticity and addiction. Nat Rev Neurosci. 2007; 8:844–858.
[PubMed: 17948030]
Buffalari and See Page 10
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Kippin TE, Fuchs RA, See RE. Contributions of prolonged contingent and noncontingent cocaine
exposure to enhanced reinstatement of cocaine seeking in rats. Psychopharmacology. 2006;
187:60–67. [PubMed: 16598453]
Koob GF. Neuroadaptive mechanisms of addiction: studies on the extended amygdala. Eur
Neuropsychopharmacol. 2003; 13:442–452. [PubMed: 14636960]
Le AD, Harding S, Juzytsch W, Funk D, Shaham Y. Role of alpha-2 adrenoceptors in stress-induced
reinstatement of alcohol seeking and alcohol self-administration in rats. Psychopharmacology.
2005; 179:366–373. [PubMed: 15551068]
Lee B, Tiefenbacher S, Platt DM, Spealman RD. Pharmacological blockade of alpha2-adrenoceptors
induces reinstatement of cocaine-seeking behavior in squirrel monkeys.
Neuropsychopharmacology. 2004; 29:686–693. [PubMed: 14872205]
Leri F, Flores J, Rodaros D, Stewart J. Blockade of stress-induced but not cocaine-induced
reinstatement by infusion of noradrenergic antagonists into the bed nucleus of the stria terminalis
or the central nucleus of the amygdala. J Neurosci. 2002; 22:5713–5718. [PubMed: 12097523]
McFarland K, Kalivas PW. The circuitry mediating cocaine-induced reinstatement of drug-seeking
behavior. J Neurosci. 2001; 21:8655–8663. [PubMed: 11606653]
McFarland K, Davidge SB, Lapish CC, Kalivas PW. Limbic and motor circuitry underlying footshock-
induced reinstatement of cocaine-seeking behavior. J Neurosci. 2004; 24:1551–1560. [PubMed:
14973230]
McLaughlin J, See RE. Selective inactivation of the dorsomedial prefrontal cortex and the basolateral
amygdala attenuates conditioned-cued reinstatement of extinguished cocaine-seeking behavior in
rats. Psychopharmacology. 2003; 168:57–65. [PubMed: 12845418]
Meil WM, See RE. Lesions of the basolateral amygdala abolish the ability of drug associated cues to
reinstate responding during withdrawal from self-administered cocaine. Behav Brain Res. 1997;
87:139–148. [PubMed: 9331482]
Millan MJ, Newman-Tancredi A, Audinot V, Cussac D, Lejeune F, Nicolas JP, Coge F, Galizzi JP,
Boutin JA, Rivet JM, Dekeyne A, Gobert A. Agonist and antagonist actions of yohimbine as
compared to fluparoxan at alpha(2)-adrenergic receptors (AR)s, serotonin (5-HT)(1A), 5-HT(1B),
5-HT(1D) and dopamine D(2) and D(3) receptors. Significance for the modulation of fronto-
cortical monoaminergic transmission and depressive states. Synapse. 2000; 35:79–95. [PubMed:
10611634]
Morrell JI, Schwanzel-Fukuda M, Fahrbach SE, Pfaff DW. Axonal projections and peptide content of
steroid hormone concentrating neurons. Peptides. 1984; 5(Suppl 1):227–239. [PubMed: 6384952]
Paxinos, G.; Watson, C. The rat brain in stereotaxic coordinates. 3. Academic; New York: 1997.
Rogers JL, Ghee S, See RE. The neural circuitry underlying reinstatement of heroin-seeking behavior
in an animal model of relapse. Neuroscience. 2008; 151:579–588. [PubMed: 18061358]
Scatton B, Zivkovic B, Dedek J. Antidopaminergic properties of yohimbine. J Pharmacol Exp Ther.
1980; 215:494–499. [PubMed: 7192314]
See RE. Neural substrates of conditioned-cued relapse to drug-seeking behavior. Pharmacol Biochem
Behav. 2002; 71:517–529. [PubMed: 11830186]
See RE. Neural substrates of cocaine-cue associations that trigger relapse. Eur J Pharmacol. 2005;
526:140–146. [PubMed: 16253228]
See RE. Dopamine D1 receptor antagonism in the prelimbic cortex blocks the reinstatement of heroin-
seeking in an animal model of relapse. Int J Neuropsychopharmacol. 2009; 12:431–436. [PubMed:
19236732]
Shaham Y, Erb S, Stewart J. Stress-induced relapse to heroin and cocaine seeking in rats: a review.
Brain Res Brain Res Rev. 2000; 33:13–33. [PubMed: 10967352]
Shalev U, Highfield D, Yap J, Shaham Y. Stress and relapse to drug seeking in rats: studies on the
generality of the effect. Psychopharmacology. 2000; 150:337–346. [PubMed: 10923762]
Shepard JD, Bossert JM, Liu SY, Shaham Y. The anxiogenic drug yohimbine reinstates
methamphetamine seeking in a rat model of drug relapse. Biol Psychiatry. 2004; 55:1082–1089.
[PubMed: 15158427]
Sinha R, Catapano D, O’Malley S. Stress-induced craving and stress response in cocaine dependent
individuals. Psychopharmacology. 1999; 142:343–351. [PubMed: 10229058]
Buffalari and See Page 11
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Sinha R, Garcia M, Paliwal P, Kreek MJ, Rounsaville BJ. Stress-induced cocaine craving and
hypothalamic–pituitary–adrenal responses are predictive of cocaine relapse outcomes. Arch Gen
Psychiatry. 2006; 63:324–331. [PubMed: 16520439]
Smith RJ, Aston-Jones G. Noradrenergic transmission in the extended amygdala: role in increased
drug-seeking and relapse during protracted drug abstinence. Brain Struct Funct. 2008; 213:43–61.
[PubMed: 18651175]
Southwick SM, Bremner JD, Rasmusson A, Morgan CA 3rd, Arnsten A, Charney DS. Role of
norepinephrine in the pathophysiology and treatment of posttraumatic stress disorder. Biol
Psychiatry. 1999; 46:1192–1204. [PubMed: 10560025]
Stine SM, Southwick SM, Petrakis IL, Kosten TR, Charney DS, Krystal JH. Yohimbine-induced
withdrawal and anxiety symptoms in opioid-dependent patients. Biol Psychiatry. 2002; 51:642–
651. [PubMed: 11955464]
Tjurmina OA, Goldstein DS, Palkovits M, Kopin IJ. Alpha2-adrenoceptor-mediated restraint of
norepinephrine synthesis, release, and turnover during immobilization in rats. Brain Research.
1999; 826:243–252. [PubMed: 10224302]
Vasa RA, Pine DS, Masten CL, Vythilingam M, Collin C, Charney DS, Neumeister A, Mogg K,
Bradley BP, Bruck M, Monk CS. Effects of yohimbine and hydrocortisone on panic symptoms,
autonomic responses, and attention to threat in healthy adults. Psychopharmacology. 2009;
204:445–455. [PubMed: 19266185]
Buffalari and See Page 12
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
a Schematic diagram illustrating placements of injection cannulae as confirmed through
histology (modified from Paxinos and Watson 1997). Coronal sections depicted are 0.2 to
0.6 mm from bregma along the A/P coordinates. Placements are shown within (circles) or
outside of (triangles) the BNST. Please note some overlap in placements. The majority of
BNST placements were in the dorsomedial portions of the BNST, with a few placed in more
ventral locations. b A representative photomicrograph of BNST placements
Buffalari and See Page 13
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Responses (mean±SEM) on the active (ACT) and inactive (INACT) levers during cocaine
self-administration (left panel) and the last 7 days of extinction responding before
reinstatement (right panel)
Buffalari and See Page 14
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
Responses (mean±SEM) on the active (previously cocaine-paired) lever for the last day of
extinction responding before testing (EXT), and following saline vehicle or baclofen–
muscimol (B/M) infusions into the BNST for conditioned cue-induced reinstatement tests
(CUE), yohimbine-induced reinstatement tests (YOH), and yohimbine+cue-induced
reinstatement tests (CUE+YOH). Significant differences (Student–Newman–Keuls test) are
indicated for reinstatement compared with extinction level responding (*p<0.05) or vehicle
vs B/M (†p<0.05)
Buffalari and See Page 15
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 4.
Responses (mean±SEM) on the active (previously cocaine-paired) lever for the last day of
extinction responding before testing (EXT), and following saline vehicle or baclofen-
muscimol (B/M) infusions directed at the BNST for conditioned cue-induced reinstatement
tests (CUE), yohimbine-induced reinstatement tests (YOH), and yohimbine+cue-induced
reinstatement tests (CUE+YOH) in animals with cannulae located outside of the BNST.
Significant differences (Student–Newman–Keuls test) are indicated for reinstatement
compared with extinction level responding (*p<0.05)
Buffalari and See Page 16
Psychopharmacology (Berl). Author manuscript; available in PMC 2011 July 8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... Importantly, both dBNST and vBNST project to the VTA (Silberman and Winder, 2013). Studies investigating regional or whole BNST's role in emotional or motivational processes have demonstrated that its functional profile spreads over a wide-range of physiological or pathological behaviours from food intake, mating, arousal, fear, to anxiety (Kalin et al., 2005;Waddell et al., 2006;Davis et al., 2009;Fox et al., 2015), depression-like behaviours (Stout et al., 2000;Hammack et al., 2004), substance abuse disorders (Erb and Stewart, 1999;Aston-Jones and Harris, 2004;Koob, 2008Koob, , 2015Buffalari and See, 2011;Pleil et al., 2015), obsessive-compulsive disorder (van Kuyck et al., 2008;Kohl et al., 2016;Wu et al., 2016;Raymaekers et al., 2017), anorexia (Roman et al., 2012), and pain (Tran et al., 2014). The growing body of evidence on BNST's functions highlights its potential as a therapeutic target for various maladaptive reward-seeking behaviours and has attracted considerable interest in the mechanism of its regulation over affective or motivational states. ...
... In addition, the BNST→VTA pathway plays an important role in cue-or stress-induced drug seeking behaviour. Inactivation of BNST attenuates cue-or stress-induced relapse of cocaineseeking behaviours (Buffalari and See, 2011). VTA-projecting BNST cells show enhanced c-Fos immunoreactivity during cue-induced reinstatement of cocaine seeking (Mahler and Aston-Jones, 2012) whereas disconnection of BNST and VTA reduces stress-induced cocaine seeking (Vranjkovic et al., 2014). ...
Article
Full-text available
We review recent studies assessing the role of the bed nucleus of the stria terminalis (BNST) in the motivational control of instrumental conditioning. This evidence suggests that the BNST and central nucleus of the amygdala (CeA) form a circuit that modulates the ventral tegmental area (VTA) input to the nucleus accumbens core (NAc core) to control the influence of Pavlovian cues on instrumental performance. In support of these claims, we found that activity in the oval region of BNST was increased by instrumental conditioning, as indexed by phosphorylated ERK activity (Experiment 1), but that this increase was not due to exposure to the instrumental contingency or to the instrumental outcome per se (Experiment 2). Instead, BNST activity was most significantly incremented in a test conducted when the instrumental outcome was anticipated but not delivered, suggesting a role for BNST in the motivational effects of anticipated outcomes on instrumental performance. To test this claim, we examined the effect of NMDA-induced cell body lesions of the BNST on general Pavlovian-to-instrumental transfer (Experiment 3). These lesions had no effect on instrumental performance or on conditioned responding during Pavlovian conditioning to either an excitory conditioned stimulus (CS) or a neutral CS (CS0) but significantly attenuated the excitatory effect of the Pavlovian CS on instrumental performance. These data are consistent with the claim that the BNST mediates the general excitatory influence of Pavlovian cues on instrumental performance and suggest BNST activity may be central to CeA-BNST modulation of a VTA-NAc core circuit in incentive motivation.
... During cocaine CPP, ventral BNST (vBNST) neurons projecting to the LH OX cells displayed unique activation and Fos expression [99]. BNST involves in stress and anxiety and hence shows a prominent role in drug-seeking behaviors [100][101][102]. Selective orexin/hypocretin receptor antagonists have significant therapeutic potential for reducing ...
Article
Full-text available
Orexin (OX, hypocretin: HCRT) as a neuropeptide is produced in a distinct population of neurons in the posterior lateral hypothalamus (LH). OX neurons implicate in reward function. OX makes a main input from the hypothalamus to the ventral tegmental area (VTA) of the midbrain. OX, through OX receptors (OXR1, OXR2) activates VTA dopamine (DA) neurons. VTA neurons are involved in reward processing and motivation. In this review, we will discuss the OX effect on addiction through VTA activation and related areas of the brain.
... However, stress exposure does not predict the severity of cocaine cravings (19). In cocaine-experienced rats, stressors (i.e., footshock, restraint, yohimbine) are highly effective at promoting reinstatement of cocaine seeking after extinction (20)(21)(22)(23)(24)(25), and there are sex differences in this phenomenon. For example, female rats are more susceptible to cocaine reinstatement after yohimbine administration than males (26); and relative to male rats, female rats exhibit greater reinstatement of cocaine seeking induced by the combination of a priming dose of cocaine and restraint stress (27). ...
Article
Full-text available
Traumatic stress disorders are defined in part by persistent avoidance of trauma-related contexts. Our lab uses a preclinical model of traumatic stress using predator odor (i.e., bobcat urine) in which some but not all rats exhibit persistent avoidance of odor-paired stimuli, similar to what is seen in humans. Bobcat urine exposure increases alcohol consumption in male Avoider rats, but it has not been tested for its effects on intake of other drugs. Here, we tested the effect of bobcat urine exposure on cocaine self-administration in adult male and female Wistar rats. We did not observe any effect of bobcat urine exposure on cocaine self-administration in male or female rats. We observed that (1) female rats with long access (6 h) to cocaine self-administer more cocaine than long-access males, (2) long-access males and females exhibit escalation of cocaine intake over time, (3) stressed rats gain less weight than unstressed rats following acute predator odor exposure, (4) baseline cocaine self-administration is predictive of subsequent cocaine self-administration. The results of this study may inform future work on predator odor effects on cocaine self-administration.
... However, stress exposure does not predict the severity of cocaine cravings (Preston and Epstein, 2011). In cocaine-experienced rats, stressors (i.e., footshock, restraint, yohimbine) are highly effective at promoting reinstatement of cocaine seeking after extinction (Buffalari and See, 2009;Buffalari and See, 2011;Graf et al., 2013;McReynolds et al., 2018;Ahmed and Koob, 1997;& Erb, Shaham, and Stewart, 1996), and there are sex differences in this phenomenon. For example, female rats are more susceptible to cocaine reinstatement after yohimbine administration than males (Feltenstein et al., 2011); and relative to male rats, female rats exhibit greater reinstatement of cocaine seeking induced by the combination of a priming dose of cocaine and restraint stress (Doncheck et al., 2020). ...
Preprint
Traumatic stress disorders are defined in part by persistent avoidance of trauma-related contexts. Our lab uses a preclinical model of traumatic stress using predator odor (i.e., bobcat urine) in which some but not all rats exhibit persistent avoidance of odor-paired stimuli, similar to what is seen in humans. Bobcat urine exposure increases alcohol consumption in male Avoider rats, but it has not been tested for its effects on intake of other drugs. Here, we tested the effect of bobcat urine exposure on cocaine self-administration in adult male and female Wistar rats. We did not observe any effect of bobcat urine exposure on cocaine self-administration in male or female rats. We observed that (1) female rats with long access (6 hours) to cocaine self-administer more cocaine than long-access males, (2) long-access males and females exhibit escalation of cocaine intake over time, (3) stressed rats gain less weight than unstressed rats following acute predator odor exposure, (4) baseline cocaine self-administration is predictive of subsequent cocaine self-administration. The results of this study may inform future work on predator odor effects on cocaine self-administration.
... The PAG provides inputs to the BNST [138,139] which is a critical structure for stress, anxiety [140][141][142], and fear responses [143]; and it is enriched mainly in NPY and CRF neurons [144]. Release of the neurotransmitter NPY has anti-drinking effects mediated through the Y 1 receptor, which inhibits BNST-CRF neurons [145], and is a key structure in the withdrawal/negative affect and anticipation/drug craving [82,146,147]. The BNST receives 50% of its DA contribution from the D-and V-PAG [139,148] (Fig. 1), and this could be involved in pain regulation and anxiety [149]. ...
Article
The periaqueductal gray (PAG) is a complex mesencephalic structure involved in the integration and execution of active and passive self-protective behaviors against imminent threats, such as immobility or flight from a predator. PAG activity is also associated with the integration of responses against physical discomfort (e.g., anxiety, fear, pain, and disgust) which occurs prior an imminent attack, but also during withdrawal from drugs such as morphine and cocaine. The PAG sends and receives projections to and from other well-documented nuclei linked to the phenomenon of drug addiction including: (i) the ventral tegmental area; (ii) extended amygdala; (iii) medial prefrontal cortex; (iv) pontine nucleus; (v) bed nucleus of the stria terminalis; and (vi) hypothalamus. Preclinical models have suggested that the PAG contributes to the modulation of anxiety, fear, and nociception (all of which may produce physical discomfort) linked with chronic exposure to drugs of abuse. Withdrawal produced by the major pharmacological classes of drugs of abuse is mediated through actions that include participation of the PAG. In support of this, there is evidence of functional, pharmacological, molecular. And/or genetic alterations in the PAG during the impulsive/compulsive intake or withdrawal from a drug. Due to its small size, it is difficult to assess the anatomical participation of the PAG when using classical neuroimaging techniques, so its physiopathology in drug addiction has been underestimated and poorly documented. In this theoretical review, we discuss the involvement of the PAG in drug addiction mainly via its role as an integrator of responses to the physical discomfort associated with drug withdrawal.
... Inactivation of the BNST reduces drug-seeking behavior (Buffalari and See, 2011). Several distinct neurotransmitters and neuromodulators are implicated in this role of the BNST in drug-related behavior (Vranjkovic et al., 2017). ...
Chapter
The bed nucleus of the stria terminalis (BNST) is a medial basal forebrain structure that modulates the hypothalamo-pituitary-adrenal (HPA) axis. The heterogeneous subnuclei of the BNST integrate inputs from mood and reward-related areas and send direct inhibitory projections to the hypothalamus. The connections between the BNST and hypothalamus are conserved across species, promote activation of the HPA axis, and can increase avoidance of aversive environments, which is historically associated with anxiety behaviors. However, BNST-hypothalamus circuitry is also implicated in motivated behaviors, drug seeking, feeding, and sexual behavior. These complex and diverse roles, as well its sexual dimorphism, indicate that the BNST-hypothalamus circuitry is an essential component of the neural circuitry that may underlie various psychiatric diseases, ranging from anorexia to anxiety to addiction. The following review is a cross-species exploration of BNST-hypothalamus circuitry. First, we describe the BNST subnuclei, microcircuitry and complex reciprocal connections with the hypothalamus. We will then discuss the behavioral functions of BNST-hypothalamus circuitry, including valence surveillance, addiction, feeding, and social behavior. Finally, we will address sex differences in morphology and function of the BNST and hypothalamus.
... Much like in people, we and others have found that, under conditions in which stress alone does not reliably reinstate extinguished cocaine seeking in rats, it can augment reinstatement in response to drug-associated cues (Buffalari et al., 2009) or administration of a low dose of cocaine that by itself is insufficient to prime cocaine seeking (Graf et al., 2013). We have found that both footshock and restraint can potentiate cocaine-primed reinstatement (Doncheck et al., 2020;Graf et al., 2013;McReynolds, Doncheck, et al., 2018;McReynolds et al., 2016), while See and colleagues have found that footshock (Buffalari & See, 2009) and yohimbine administration (Buffalari & See, 2011;Feltenstein & See, 2006) can potentiate reinstatement in response to cocaine-associated cues. ...
Article
Full-text available
In individuals with substance use disorders, stress is a critical determinant of relapse susceptibility. In some cases, stressors directly trigger cocaine use. In others, stressors interact with other stimuli to promote drug seeking, thereby setting the stage for relapse. Here, we review the mechanisms and neurocircuitry that mediate stress‐triggered and stress‐potentiated cocaine seeking. Stressors trigger cocaine seeking by activating noradrenergic projections originating in the lateral tegmentum that innervate the bed nucleus of the stria terminalis to produce beta adrenergic receptor‐dependent regulation of neurons that release corticotropin releasing factor (CRF) into the ventral tegmental area (VTA). CRF promotes the activation of VTA dopamine neurons that innervate the prelimbic prefrontal cortex resulting in D1 receptor‐dependent excitation of a pathway to the nucleus accumbens core that mediates cocaine seeking. The stage‐setting effects of stress require glucocorticoids, which exert rapid non‐canonical effects at several sites within the mesocorticolimbic system. In the nucleus accumbens, corticosterone attenuates dopamine clearance via the organic cation transporter 3 to promote dopamine signaling. In the prelimbic cortex, corticosterone mobilizes the endocannabinoid, 2‐arachidonoylglycerol (2‐AG), which produces CB1 receptor‐dependent reductions in inhibitory transmission, thereby increasing excitability of neurons which comprise output pathways responsible for cocaine seeking. Factors that influence the role of stress in cocaine seeking, including prior history of drug use, biological sex, chronic stress/co‐morbid stress‐related disorders, adolescence, social variables, and genetics are discussed. Better understanding when and how stress contributes to drug seeking should guide the development of more effective interventions, particularly for those whose drug use is stress related. image
... The role of BNST as well as central amygdala inputs to VTA in relapse also remains underexplored. Given recruitment of these inputs by drug-related stimuli and stressors, the well-documented role of BNST in stress-and cue-induced reinstatement, as well as the role for corticotropin-releasing hormone in the VTA during stressinduced reinstatement, these inputs are likely to play an important role in both cue-and stress-induced reinstatement of drug seeking (Briand et al., 2010;Buffalari & See, 2011;Erb et al., 2001;Erb & Stewart, 1999;Leri et al., 2002;Mahler & Aston-Jones, 2012;Perez et al., 2020;Wang et al., 2007;Wise & Morales, 2010). ...
Article
Full-text available
The actions of dopamine are essential to relapse to drug seeking but we still lack a precise understanding of how dopamine achieves these effects. Here we review recent advances from animal models in understanding how dopamine controls relapse to drug seeking. These advances have been enabled by important developments in understanding about the basic neurochemical, molecular, anatomical, physiological and functional properties of the major dopamine pathways in the mammalian brain. The literature shows that although different forms of relapse to seeking different drugs of abuse each depend on dopamine, there are distinct dopamine mechanisms for relapse. Different circuit level mechanisms, different populations of dopamine neurons, and different activity profiles within these dopamine neurons, are important for driving different forms of relapse. This diversity highlights the need to better understand when, where and how dopamine contributes to relapse behaviours. image
... Baclofen/muscimol injection into the NAc core or shell suppressed context-induced reinstatement of cocaine seeking (Fuchs et al. 2008). Baclofen/muscimol injection into the bed nucleus of the stria terminalis (BNST), a key convergence point of cues and stress, reduced cue and/or yohimbine stressinduced reinstatement of cocaine seeking (Buffalari and See 2011). The LHb encodes aversive and anxiogenic states, and when inactivated by baclofen/muscimol resulted in reduced cue-induced cocaine reinstatement in the presence of yohimbine stress (Gill et al. 2013). ...
Chapter
Full-text available
Metabotropic GABAB receptors (GABABRs) mediate slow inhibition and modulate synaptic plasticity throughout the brain. Dysfunction of GABABRs has been associated with psychiatric illnesses and addiction. Drugs of abuse alter GABAB receptor (GABABR) signaling in multiple brain regions, which partly contributes to the development of drug addiction. Recently, GABABR ligands and positive allosteric modulators (PAMs) have been shown to attenuate the initial rewarding effect of addictive substances, inhibit seeking and taking of these drugs, and in some cases, ameliorate drug withdrawal symptoms. The majority of the anti-addiction effects seen with GABABR modulation can be localized to ventral tegmental area (VTA) dopamine neurons, which receive complex inhibitory and excitatory inputs that are modified by drugs of abuse. Preclinical research suggests that GABABR PAMs are emerging as promising candidates for the treatment of drug addiction. Clinical studies on drug dependence have shown positive results with GABABR ligands but more are needed, and compounds with better pharmacokinetics and fewer side effects are critically needed.
Article
The microdialysis technique was used to simultaneously study the in vivo extracellular levels of noradrenaline, glutamate, and gamma aminobutyric acid (GABA) in the bed nucleus of the stria terminalis in order to assess the regulation that noradrenaline may exert upon the release of amino acid neurotransmitters. Perfusion through the probe with UK14304, a selective alpha(2)-adrenergic agonist, produced a significant decrease of noradrenaline and glutamate extracellular levels. Perfusion through the probe with RX821002, a selective alpha(2)-adrenergic antagonist, produced a significant increase of noradrenaline and glutamate basal extracellular levels. Perfusion with prazosine, a selective alpha(1)-adrenergic antagonist, produced a significant decrease of noradrenaline basal extracellular levels without affecting glutamate levels. Under the same conditions, GABA basal extracellular levels were not changed in the presence of any of the alpha-adrenergic ligands studied. The perfusion of high potassium through the probe induced a significant Ca++- dependent release of the three neurotransmitters; however, extracellular noradrenaline returned to normal levels even though potassium was still present. In addition, it was observed that alpha-adrenergic receptor ligands exerted differential effects upon K+-induced release of noradrenaline and glutamate. Perfusion with the nonselective alpha-adrenergic antagonist, phenoxybenzamine, presented a biphasic effect upon K+-induced release of noradrenaline; a significant decrease during the first 5 min of stimulation followed by a significant increase in the next 5 min of stimulation. Perfusion with RX821002 produced a significant increase in K+-induced release of noradrenaline that returned to normal basal values before the end of the stimulation period. In contrast, local perfusion with prazosine caused a significant decrease of K+-induced noradrenaline release. In the case of glutamate, perfusion through the probe with phenoxybenzamine produced a significant increase in K+-induced release of glutamate. In addition, RX821002 and prazosine produced a significant increase in K+-induced release of glutamate. Perfusion through the probe with UK14304 produced a significant decrease of both noradrenaline and glutamate K+-induced release. The present results show that noradrenaline in the bed nucleus of stria terminalis exerts a significant inhibition over its own release through alpha(2)-adrenergic receptors and over glutamate release mainly through alpha(2)-adrenergic receptors. Thus, the results suggest that noradrenaline in the bed nucleus of the stria terminalis maintains an inhibitory tone over the information flow mediated by glutamate. J. Neurosci. Res. 55:311-320, 1999. (C) 1999 Wiley-Liss, Inc.
Article
Brain noradrenaline is involved in footshock stress-induced reinstatement of drug seeking in a rat relapse model. We studied whether yohimbine, an α-2 adrenoceptor antagonist that increases noradrenaline release and induces anxiety-like responses in human and nonhuman subjects, would reinstate methamphetamine seeking in rats.Methods In experiment 1, the effect of yohimbine (1.25–2.5 mg/kg) on reinstatement was compared with that of intermittent footshock (5 min; .2–.6 mA) in rats that were trained to lever press for intravenous methamphetamine (9–11 days) and subsequently underwent 7 days of extinction training. In experiment 2, the effect of yohimbine on reinstatement of drug seeking was determined during early (1 day) and late (21 or 51 days) withdrawal periods. On the test days, rats were first given 3-hour extinction sessions and were then tested for reinstatement induced by yohimbine.ResultsIn experiment 1, both yohimbine and footshock stress reinstated methamphetamine seeking after extinction. In experiment 2, extinction responding was higher after 21 or 51 withdrawal days than after 1 withdrawal day. In contrast, no significant time-dependent changes in yohimbine-induced reinstatement were observed.Conclusions Results indicate that yohimbine is a potent stimulus for reinstatement of methamphetamine seeking in a rat relapse model.
Article
Herein, we evaluate the interaction of the 2-AR antagonist, yohimbine, as compared to fluparoxan, at multiple monoaminergic receptors and examine their roles in the modulation of adrenergic, dopaminergic and serotonergic transmission in freely-moving rats. Yohimbine displays marked affinity at human (h)2A-, h2B- and h2C-ARs, significant affinity for h5-HT1A, h5-HT1B, h5-HT1D, and hD2 receptors and weak affinity for hD3 receptors. In [35S]GTPS binding protocols, yohimbine exerts antagonist actions at h2A-AR, h5-HT1B, h5-HT1D, and hD2 sites, yet partial agonist actions at h5-HT1A sites. In vivo, agonist actions of yohimbine at 5-HT1A sites are revealed by WAY100,635-reversible induction of hypothermia in the rat. In guinea pigs, antagonist actions of yohimbine at 5-HT1B receptors are revealed by blockade of hypothermia evoked by the 5-HT1B agonist, GR46,611. In distinction to yohimbine, fluparoxan shows only modest partial agonist actions at h5-HT1A sites versus marked antagonist actions at h2-ARs. While fluparoxan selectively enhances hippocampal noradrenaline (NAD) turnover, yohimbine also enhances striatal dopamine (DA) turnover and suppresses striatal turnover of 5-HT. Further, yohimbine decreases firing of serotonergic neurones in raphe nuclei, an action reversed by WAY100,635. Fluparoxan increases extracellular levels of DA and NAD, but not 5-HT, in frontal cortex. In analogy, yohimbine enhances FCX levels of DA and NAD, yet suppresses those of 5-HT, the latter effect being antagonized by WAY100,635. The induction by fluoxetine of FCX levels of 5-HT, DA, and NAD is potentiated by fluparoxan. Yohimbine likewise facilitates the influence of fluoxetine upon DA and NAD levels, but not those of 5-HT. In conclusion, the 2-AR antagonist properties of yohimbine increase DA and NAD levels both alone and in association with fluoxetine. However, in contrast to the selective 2-AR antagonist, fluparoxan, the 5-HT1A agonist actions of yohimbine suppress 5-HT levels alone and underlie its inability to augment the influence of fluoxetine upon 5-HT levels. Synapse 35:79–95, 2000. © 2000 Wiley-Liss, Inc.
Article
Hyperactivity of brain norepinephrine (NE) systems has long been implicated in mechanisms of opiate withdrawal (OW). However, little is known about where elevated NE may act to promote OW. Here we report that the bed nucleus of the stria terminalis (BNST), the densest NE target in the brain, is critical for NE actions in OW. (1) Many BNST neurons become Fos+ after OW. Pretreatment with the β antagonist, propranolol, markedly reduces OW symptoms and the number of Fos+ cells in the BNST. (2) Numerous neurons in the nucleus tractus solitarius (A2 neurons) and the A1 cell group are triple labeled for tyrosine hydroxylase, a retrograde tracer from the BNST, and Fos after OW, revealing numerous NE neurons that project to the BNST from the medulla that are stimulated by OW. Fewer such triple-labeled neurons were found in the locus caeruleus. (3) Behavioral studies reveal that local microinjections of selective β-adrenergic antagonists into the BNST attenuate OW symptoms. In particular, withdrawal-induced place aversion is abolished by bilateral microinjection of a cocktail of selective beta 1 (betaxolol) plus the beta 2 (ICI 181,555) antagonists (1.0 nmol each/0.5 μL per side) into the BNST. Similar results were obtained with neurochemically selective lesions of the ventral ascending NE bundle, the pathway for A1 and A2 projections to the BNST. Similar lesions of the dorsal NE bundle of projections from the locus caeruleus had no effect on either aversive or somatic withdrawal symptoms. Together, these results indicate that β-receptor activation in the BNST is critical for aversive withdrawal symptoms, and that A1 and A2 neurons in the medulla are the source of this critical NE.
Article
Stress-related release of norepinephrine (NE) in the brain and periphery probably underlies several neuroendocrine and neurocirculatory responses. NE might influence its own synthesis, release, and turnover, by negative feedback regulation via α2-adrenoceptors. We examined central and peripheral noradrenergic function by measuring concentrations of NE, dihydroxyphenylglycol (DHPG), and dihydroxyphenylacetic acid (DOPAC) in hypothalamic paraventricular nucleus (PVN) microdialysate and arterial plasma simultaneously during immobilization (IMMO) in conscious rats. The α2-adrenoceptor antagonist yohimbine (YOH) was injected i.p. or perfused locally into the PVN via the microdialysis probe. The i.p. YOH increased plasma NE, epinephrine (EPI), DHPG, dihydroxyphenylalanine, and DOPAC levels by 4.3, 7.3, 2.5, 0.6 and 1.8-fold and PVN microdialysate NE, DHPG, and DOPAC by 1.2, 0.6 and 0.5-fold. The i.p. YOH also enhanced effects of IMMO on plasma and microdialysate NE, DHPG, and DOPAC. YOH delivered via the PVN microdialysis probe did not affect microdialysate or plasma levels of the analytes at baseline and only slightly augmented microdialysate NE responses to IMMO. The results indicate that α2-adrenoceptors tonically restrain NE synthesis, release, and turnover in sympathetic nerves and limit IMMO-induced peripheral noradrenergic activation. In the PVN, α2-adrenoceptors do not appear to contribute to these processes tonically and exert relatively little restraint on IMMO-induced local noradrenergic activation.