ArticlePDF Available

Long-Term Potentiation (LTP) in the Central Amygdala (CeA) Is Enhanced After Prolonged Withdrawal From Chronic Cocaine and Requires CRF1 Receptors

Authors:

Abstract and Figures

The amygdala is part of the brain reward circuitry that plays a role in cocaine-seeking and abstinence in animals and cocaine craving and relapse in humans. Cocaine-seeking is elicited by cocaine-associated cues, and the basolateral amygdala (BLA) and CeA are essential in forming and communicating drug-related associations that are thought to be critical in long-lasting relapse risk associated with drug addiction. Here we simulated a cue stimulus with high-frequency stimulation (HFS) of the BLA-CeA pathway to examine mechanisms that may contribute to drug-related associations. We found enhanced long-term potentiation (LTP) after 14-day but not 1-day withdrawal from 7-day cocaine treatment mediated through N-methyl-d-aspartate (NMDA) receptors (NRs), L-type voltage-gated calcium channels (L-VGCCs), and corticotropin-releasing factor (CRF)(1) receptors; this was accompanied by increased phosphorylated NR1 and CRF(1) protein not associated with changes in NMDA/AMPA ratios in amygdalae from cocaine-treated animals. We suggest that these signaling mechanisms may provide therapeutic targets for the treatment of cocaine cravings.
Content may be subject to copyright.
Report
Long-Term Potentiation (LTP) in the Central Amygdala (CeA) Is Enhanced
After Prolonged Withdrawal From Chronic Cocaine and Requires
CRF
1
Receptors
Yu Fu, Sebastian Pollandt, Jie Liu, Balaji Krishnan, Kathy Genzer, Luis Orozco-Cabal, Joel P. Gallagher,
and Patricia Shinnick-Gallagher
Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, Texas
Submitted 5 April 2006; accepted in final form 26 October 2006
Fu Y, Pollandt S, Liu J, Krishnan B, Genzer K, Orozco-Cabal
L, Gallagher JP, Shinnick-Gallagher P. Long-term potentiation
(LTP) in the central amygdala (CeA) is enhanced after prolonged
withdrawal from chronic cocaine and requires CRF
1
receptors. J
Neurophysiol 97: 937–941, 2007. First published November 1,
2006; doi:10.1152/jn.00349.2006. The amygdala is part of the brain
reward circuitry that plays a role in cocaine-seeking and abstinence in
animals and cocaine craving and relapse in humans. Cocaine-seeking
is elicited by cocaine-associated cues, and the basolateral amygdala
(BLA) and CeA are essential in forming and communicating drug-
related associations that are thought to be critical in long-lasting
relapse risk associated with drug addiction. Here we simulated a cue
stimulus with high-frequency stimulation (HFS) of the BLA–CeA
pathway to examine mechanisms that may contribute to drug-related
associations. We found enhanced long-term potentiation (LTP) after
14-day but not 1-day withdrawal from 7-day cocaine treatment me-
diated through N-methyl-D-aspartate (NMDA) receptors (NRs), L-
type voltage-gated calcium channels (L-VGCCs), and corticotropin-
releasing factor (CRF)
1
receptors; this was accompanied by increased
phosphorylated NR1 and CRF
1
protein not associated with changes in
NMDA/AMPA ratios in amygdalae from cocaine-treated animals. We
suggest that these signaling mechanisms may provide therapeutic
targets for the treatment of cocaine cravings.
INTRODUCTION
The amygdala is essential in forming stimulus–reward asso-
ciations and associational processing of conditioned cues
(Aggleton 1992; Shinnick-Gallagher et al. 2003). Drug-asso-
ciated cues can induce craving in cocaine users and alter neural
activity in the amygdala (Childress et al. 1999), and electrical
stimulation of the basolateral amygdala (BLA) can reinstate
drug-seeking in animals (Hayes et al. 2003). Drug-cue associ-
ations are not well understood, but the mechanisms may be
similar to forms of synaptic plasticity and the induction and
expression of cocaine sensitization, a model for long-term
neuroadaptations important in addiction (De Vries et al. 1999;
Kalivas and Alesdatter 1993; Robinson and Berridge 1993).
Antagonizing N-methyl-D-aspartate (NMDA) receptors (NRs)
in the amygdala can prevent and block locomotor sensitizing
effects of chronic cocaine (Kalivas and Alesdatter 1993), and
amygdala NR1 protein levels are increased after acute and
chronic cocaine (Turchan et al. 2003). Likewise, activation of
L-type calcium channels mimics the induction (Lin et al. 2001)
and antagonists block expression of cocaine sensitization
(Pierce et al. 1998). Furthermore, corticotropin-releasing factor
(CRF) systems in the amygdala play a significant role in
cocaine addiction (Sarnyai et al. 2001). In cocaine-treated
animals, CRF release in the amygdala is enhanced during acute
withdrawal (Richter and Weiss 1999) and in response to a
cocaine challenge (Richter et al. 1995). Amygdala CRF im-
munolabeling decreases after short-term, but increases after
long-term withdrawal from chronic cocaine (Zorrilla et al.
2001). Furthermore, cocaine-induced locomotor activity is
blocked by intracerebroventricular injection of a CRF antago-
nist (Sarnyai et al. 1992). These data provide strong rationale
for testing the role of CRF receptors in synaptic plasticity in the
central amygdala (CeA). This study shows that specific cocaine
treatment and withdrawal paradigms resulted in enhanced syn-
aptic plasticity, that NR, L-type calcium channels, and CRF
1
were required for long-term potentiation (LTP) in the BLA–
CeA pathway, and that phosphorylated-NR1 (P-NR1) and
CRF
1
protein but not synaptic potentials were increased after
withdrawal from chronic cocaine.
METHODS
Cocaine HCl was a gift from the National Institute of Drug Abuse.
Male Sprague-Dawley albino rats (Harlan, 4 6 wk) were injected
daily with cocaine (15 mg/kg, ip) or saline (0.1 ml/kg, ip), once or
twice per day for 1 or 2 wk to assess how duration and frequency of
cocaine treatment and withdrawal time influence synaptic plasticity.
Behavioral sensitization measuring the progressive locomotor stimu-
lant properties resulting from chronic cocaine treatment was analyzed
with a photocell apparatus before and on the first and last days of
cocaine or saline treatment (Fig. 1E) as a measure of cocaine’s
effectiveness. After 7 days, locomotor activity was increased signif-
icantly over 1-day activity in the cocaine-treated (F
1,120
260.07,
P0.001) but not in the saline-treated (P0.05) group. No
significant differences were observed between animals injected once
per day for 7 days or twice daily for 14 days. Coronal brain slices (500
m) were prepared and incubated at room temperature for 1 h with
oxygenated, modified artificial cerebrospinal fluid (ACSF) solution (in
mM): 119 NaCl, 3.0 KCl, 1.2 NaH
2
PO
4
, 1.2 MgSO
4
, 2.5 CaCl
2
,25
NaHCO
3
, and 11.5 glucose. They were then submerged in a chamber
(1.0 ml, 2.5 ml/min) at 30 1°C for another hour before recording.
BLA fibers were stimulated with concentric electrodes (50 k) using
150-
s pulses of varying intensity (3–15 V) at 0.05 Hz, and field
excitatory postsynaptic potentials (fEPSPs) were recorded in the
capsula/medial CeA with tungsten electrodes (2–5 M). fEPSP mag-
Address for reprint requests and other correspondence: P. Shinnick-Gal-
lagher, 301 University Blvd., Galveston, TX 77555-1031 (E-mail:
psgallag@utmb.edu).
The costs of publication of this article were defrayed in part by the payment
of page charges. The article must therefore be hereby marked advertisement
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
J Neurophysiol 97: 937–941, 2007.
First published November 1, 2006; doi:10.1152/jn.00349.2006.
9370022-3077/07 $8.00 Copyright © 2007 The American Physiological Societywww.jn.org
by 10.220.33.2 on May 25, 2017http://jn.physiology.org/Downloaded from
nitude was adjusted to 30% of maximum response and baseline
recorded, and LTP was induced using high-frequency stimulation
(HFS) consisting of five trains of stimuli (100 Hz for 1 s, 3-min
intervals). fEPSPs were recorded at 0.05 Hz for another hour, and
their slopes were normalized to baseline values. A one- or two-
tailed unpaired t-test or one-way ANOVA with appropriate post
hoc tests were used for statistical analysis; nequals the number of
slices. Methodologies for patch recording (Liu et al. 2004) and
Western blot analysis (Zinebi et al. 2003) were similar to that reported
previously.
RESULTS
Amygdala slices were prepared, and LTP was assessed in
the BLA–CeA pathway (Fig. 1, A–D). We first tested the
influence of varying cocaine administration paradigms and
withdrawal time on LTP. Animals received cocaine or saline
either once per day for 7 days or twice per day for 14 days,
followed by either 1 or 14 days of withdrawal. Input– output
relationships were similar in all treatment groups (Fig. 1D). In
slices from animals treated with cocaine twice per day for 14
days and a 14-day withdrawal, fEPSP slopes after HFS
(203.5 11.7%, n10) were significantly enhanced com-
pared with fEPSPs obtained from control animals (144.2
4.7%, n10, P0.001). When treatment duration and
frequency were reduced to 7 days of cocaine once per day,
HFS-LTP (202.1 12.1%, n12) remained significantly
enhanced compared with saline-treated animals (160.1
9.0%, n12; P0.05) after 14 days of withdrawal. LTP was
not significantly different between 7- and 14-day cocaine
treatment groups. However, when withdrawal time was re-
duced to 1 day in animals receiving cocaine twice per day for
14 days, the resulting LTP (168.3 13.7%, n7) was not
significantly different from saline controls (140.8 4.3%, n9,
P0.05), indicating that withdrawal time was crucial in
enhancing LTP, whereas treatment duration or frequency had
no significant impact. Subsequent experiments used the 7-day
treatment (once per day) and 14-day withdrawal paradigm.
Previously, we showed that HFS-LTP in the BLA–CeA
pathway depends on NRs and L-VGCCs (Fu and Shinnick-
Gallagher 2005). To examine whether induction mechanisms
were altered in cocaine-enhanced LTP (Fig. 2), slices were
superfused with the NMDA antagonist APV (50
M) in ACSF
or the L-VGCC antagonist nimodipine (NIM, 10
M) 15 min
before HFS. APV blocked LTP both in cocaine (control:
202.1 12.1%, n12; APV: 110.4 2.4%, n5, P
0.001) and saline (control: 160.1 9.0%, n12; APV:
107.7 7.4%, n5, P0.005) groups. Similarly, NIM
blocked LTP in cocaine-treated (control: 202.1 12.1%, n
12; NIM: 104.0 11.8%, n5, P0.001) and saline-treated
(control: 160.1 9.0%, n12; NIM: 109.0 7.7%, n5,
P0.005) groups. These data indicated that NMDA receptors
and L-VGCCs are necessary for LTP induction by HFS in
cocaine and saline treatment groups.
FIG. 1. Long-term potentiation (LTP) at the basolateral
amygdala (BLA)– central amygdala (CeA) pathway was en-
hanced after 2-wk (B) but not 24-h (A) withdrawal from chronic
cocaine without changes in single field excitatory postsynaptic
potential (fEPSP) responsiveness (D). Aand B: traces above
indicated fEPSPs before and after high-frequency stimulation
(HFS)-LTP at the times indicated in the bottom graphs, showing
summary data of LTP time-course. C: plot of last 10 fEPSPs
(mean SE) 1 h after LTP induction showed enhanced LTP
after 7- and 14-day cocaine tratment and 14-day withdawal but
not after 14-day treatment and 24-h withdrawal. Time-course of
7-day treatment and 14-day withdrawal is shown in Figs. 2 and
3. D: input– output relationships are not altered in any treatment
paradigm. E: horizontal locomotor activity is enhanced after 7
days of cocaine treatment, suggesting behavior sensitization.
Calibrations are the same in Aand B.
Report
938 FU ET AL.
J Neurophysiol VOL 97 JANUARY 2007 www.jn.org
by 10.220.33.2 on May 25, 2017http://jn.physiology.org/Downloaded from
Because CRF has been implicated in the pathophysiology of
drug addiction (Sarnyai et al. 2001), we also examined whether
CRF receptors modulate HFS-LTP (Fig. 3). The selective
CRF
1
antagonist NBI27914 (250 nM) blocked HFS-LTP in
cocaine (control: 202.1 12.1%, n12; NBI: 114.0 7.4%,
n5, P0.001) and saline (control: 160.1 9.0%, n12;
NBI: 119.9 2.8%, n5, P0.05) groups; astressin2-B, a
selective CRF
2
antagonist, did not significantly affect HFS-
LTP in either group (saline: 164 25.1%, n6, P0.05;
cocaine: 191 26.3%, n6, P0.05). These data indicated
an obligatory role for CRF
1
in synaptic plasticity in the
BLA–CeA pathway.
To further study the mechanisms contributing to cocaine-
enhanced LTP, we analyzed amygdala protein levels using
Western blots (7 animals/group; Fig. 3C). NR1 protein was not
quite significantly elevated (P0.07) and P-NR1 protein was
significantly increased (P0.03) in cocaine withdrawn ani-
mals, whereas the L-VGCC
1C subunit (Ca
V
1.2, P0.2)
was not altered, suggesting that signaling through NMDA may
be altered after cocaine treatment. However, NMDA/AMPA
ratios measured at 20mV were not changed in cocaine-
treated animals (saline: 0.097 ⫾⫺0.015, cocaine: 0.110
0.019, n6/group), indicating that increased phosphorylated
NR protein was not reflected in the ratio at this synapse. In
agreement with our electrophysiological results, CRF
1
protein
levels were significantly increased after cocaine treatment (P
0.04), whereas CRF
2
remained unchanged (P0.35).
DISCUSSION
Our studies show for the first time that LTP in the BLA–CeA
pathway was enhanced after long- but not short-term with-
drawal from chronic cocaine, that HFS-induced LTP was
modulated endogenously by CRF
1
, and that elevated P-NR1
and CRF
1
protein may contribute to cocaine-enhanced LTP.
The withdrawal-induced enhancement of LTP may be related
to the neuroadaptive effects associated with behavioral sensi-
tization that can persist for 2 wk (Kalivas et al. 1988) and that
FIG. 2. LTP in the BLA pathway is dependent on N-methyl-
D-aspartate (NMDA) receptors (Aand B) and L-type voltage-
gated calcium channels (L-VGCCs) (Cand D) in slices 14 days
after 7-day treatment with either saline (left) or cocaine (right).
In each panel, numbered traces show responses before and after
HFS at times indicated in graph in slices from saline (Aand C)-
and cocaine (Band D)-treated animals in presence and absence
of APV (Aand B) or nimodipine (NIM; Cand D). Graphs below
show summary date of LTP time-course in slices from saline (A
and C)- and cocaine (Band D)-treated groups in the presence
and absence of APV (Aand B) or NIM (Cand D). The same
control data are plotted for saline (A,C, and Fig. 3C) and
cocaine (B,D, and Fig. 3D). Calibrations in B–D are the same
as in A.
FIG. 3. LTP in BLA–CeA pathway is dependent on corti-
cotropin-releasing factor (CRF)
1
receptors in saline- and co-
caine-treated populations and P-NR1, and CRF
1
protein is
increased in cocaine-treated populations. Aand B: numbered
top traces recorded in slices from saline (A)- and cocaine
(B)-treated groups show fEPSPs before and after HFS-induced
LTP at times indicated in the bottom graphs, which show
summary data for LTP time-course. C: Western blots for NR1
(Santa Cruz), P-NR1 (Upstate), and CRF1 (Santa Cruz) pro-
tein (top blots) and corresponding actin (Santa Cruz) protein
(bottom blots) are expressed as optical density ratios in the
summary graphs below. The same control data are plotted for
saline (Cand Fig. 2, Aand C) and cocaine (Dand Fig. 2, Band
D).
Report
939COCAINE WITHDRAWAL-ENHANCED LTP REQUIRES CRF
1
J Neurophysiol VOL 97 JANUARY 2007 www.jn.org
by 10.220.33.2 on May 25, 2017http://jn.physiology.org/Downloaded from
may have a component of associative contextual conditioning
(Carey and Gui 1998). Markers for BLA neuronal activity
increase in animals exposed to a cocaine environment (Brown
et al. 1992; Neisewander et al. 2000; Thomas et al. 2003) even
after 4-mo withdrawal (Ciccocioppo et al. 2001). Thus stimu-
lating the BLA may simulate neuronal activity during cue
exposure after the animal has been sensitized.
Cocaine-enhanced LTP in the BLA–CeA pathway was crit-
ically dependent on withdrawal, whereas baseline fEPSP re-
sponsiveness was unchanged. After 4- to 6-day withdrawal
after 5-day cocaine treatment, hippocampal LTP was enhanced
(Thompson et al. 2002), but after 100-day withdrawal and at
higher self-administered cocaine doses, LTP was reduced
(Thompson et al. 2004); these effects were also not correlated
with an altered fEPSP amplitude. Conversely, at an intralateral
amygdala (LA) synapse, an increased baseline response and
reduced LTP were observed with cocaine treatment (15 mg/kg,
3 times per day, 1-h intervals for 7 days) and 1- to 3-day
withdrawal, but the effect dissipated within 9 days; this reduc-
tion in LTP was interpreted as occlusion caused by the facili-
tated baseline EPSP response (Goussakov et al. 2006). Func-
tionally, these data indicated that long-lasting effects of co-
caine were consistently revealed with HFS. Although
disparities in findings suggest that changes in synaptic facili-
tation and plasticity are dependent on brain area, synapse, and
treatment paradigm, the studies provide insight into the relative
persistence of the effects of cocaine treatment.
HFS-LTP at the BLA–CeA synapse is dependent on NMDA
receptors and L-VGCCs. After 2-wk withdrawal from chronic
cocaine, NR2B and NR1 subunits are upregulated in other
brain areas (Loftis and Janowsky 2000). Here we report a
similar increase in P-NR1 protein in the amygdala, which
could contribute to the enhanced HFS-LTP after chronic co-
caine. However, changes in NMDA/AMPA ratios were not
detected, suggesting that the increased P-NR1 proteins may not
be accessible to transmitter evoked with single stimuli at this
synapse.Cocaine withdrawal also increases calcium entry
through L-VGCCs (Nasif et al. 2005), and L-VGCC antago-
nists block establishment of conditioned locomotion by co-
caine (Reimer and Martin-Iverson 1994), suggesting that
greater L-VGCC activity in cocaine withdrawn animals could
contribute to the cocaine-enhanced HFS-LTP. However, it is
unlikely that increased L-VGCC activity contributed to the
cocaine-enhanced LTP because
1C subunit protein was un-
changed in the cocaine group. CRF
2
receptor activation poten-
tiated NMDA responses in ventral tegmental area neurons
(Ungless et al. 2003) but CRF
2
was not involved in HFS-LTP
in the BLA–CeA pathway, and amygdala CRF
2
protein was
not increased with chronic cocaine. These data suggest that
L-type VGCCs or CRF
2
receptors may not play a role in the
cocaine-enhanced LTP, whereas increased P-NR1 protein may
contribute to enhanced HFS-LTP but not to singly evoked
EPSPs at the BLA–CeA synapse.
Although the CRF
1
antagonist did not affect baseline fEPSPs,
subsequent HFS failed to induce LTP in slices from cocaine
and saline groups, indicating that CRF
1
receptors are required
for LTP induction. We previously showed that exogenous CRF
directly enhanced mEPSC frequency in the CeA, suggesting a
presynaptic increase in glutamate release (Liu et al. 2004).
Repetitively stimulating cerebellar afferents is known to re-
lease CRF (Tian and Bishop 2003), and afferent stimulation
(foot-shock) can increase endogenous CRF release in the CeA
and BLA (Roozendaal et al. 2002). These results suggested that
HFS could enhance endogenous CRF release in the CeA. CRF
priming enhances HFS-induced LTP (Blank et al. 2002), CRF
itself can induce LTP in the hippocampus (Wang et al. 1998),
and at LA–CeA (Pollandt et al. 2006) and BLA–CeA (Fu et al.
2004) synapses, and CRF-induced LTP is enhanced after
chronic cocaine. Furthermore, both CRF
1
protein (Radulovic et
al. 1998) and mRNA (Chalmers et al. 1995) are found in the
BLA, and CRF
1
receptors are located on excitatory type
terminals in the CeA (Chalmers et al. 1995), suggesting an
anatomical basis for a CRF
1
-mediated effect on glutamate
release. The block of HFS-LTP by the CRF
1
antagonist,
increase in CRF
1
protein, and enhanced responsiveness to CRF
in the BLA–CeA pathway (Fu et al. 2004) after cocaine
withdrawal suggests that endogenously released CRF acting
through CRF
1
receptors contributes to the enhanced LTP in
cocaine. CRF is known to enhance locally evoked GABA
inhibition in the CeA through CRF
1
receptors (Nie et al. 2004).
With GABA inhibition intact, we previously found that low
CRF concentrations inhibited evoked excitatory postsynaptic
currents (EPSCs) 40%, whereas in the presence of GABA
antagonists, CRF inhibited miniature EPSCs by only 20% (Liu
et al. 2004), indicating that one half of CRF-induced inhibition
of evoked EPSCs may be caused by CRF-induced GABA
release. However, HFS-LTP in this pathway is not significantly
altered by GABA antagonists (Fu and Shinnick-Gallagher
2005), and GABA antagonists did not affect NBI inhibition of
HFS-LTP (data not shown). Altogether the results suggest that
an HFS-induced increase in CRF release in the presence of
GABA antagonists resulted in facilitated glutamate release,
which prevailed over an inhibitory effect and induced LTP.
Furthermore, our data suggest that increases in P-NR1 and
CRF
1
protein and/or their downstream signaling mechanisms
may contribute to the cocaine-enhanced LTP at the BLA–CeA
synapse.
ACKNOWLEDGMENTS
The authors thank K. Schmidt for helpful comments on the manuscript.
GRANTS
This work was supported by National Institute of Drug Abuse Grants
DA-017727 to P. Shinnick-Gallagher and DA-011991 to J. P. Gallagher.
REFERENCES
Aggleton JP. The Amygdala: Neurobiological Aspects of Emotion, Memory,
and Mental Dysfunction. New York: WileyLiss, 1992.
Blank T, Nijholt I, Eckart K, Spiess J. Priming of long-term potentiation in
mouse hippocampus by corticotropin-releasing factor and acute stress:
implications for hippocampus-dependent learning. J Neurosci 22: 3788
3794, 2002.
Brown EE, Robertson GS, Fibiger HC. Evidence for conditional neuronal
activation following exposure to a cocaine-paired environment: role of
forebrain limbic structures. J Neurosci 12: 4112– 4121, 1992.
Carey RJ, Gui J. Cocaine conditioning and cocaine sensitization: what is the
relationship. Behav Brain Res 92: 67–76, 1998.
Chalmers DT, Lovenberg TW, De Souza EB. Localization of novel corti-
cotropin-releasing factor receptor (CRF2) mRNA expression to specific
subcortical nuclei in rat brain: comparison with CRF1 receptor mRNA
expression. J Neurosci 15: 6340 6350, 1995.
Childress AR, Mozley PD, McElgin W, Fitzgerald J, Reivich M, O’Brien
CP. Limbic activation during cue-induced cocaine craving. Am J Psychiatry
156: 11–18, 1999.
Ciccocioppo R, Sanna PP, Weiss F. Cocaine-predictive stimulus induces
drug-seeking behavior and neural activation in limbic brain regions after
Report
940 FU ET AL.
J Neurophysiol VOL 97 JANUARY 2007 www.jn.org
by 10.220.33.2 on May 25, 2017http://jn.physiology.org/Downloaded from
multiple months of abstinence: reversal by D(1) antagonists. Proc Natl Acad
Sci USA 98: 1976 –1981, 2001.
De Vries TJ, Schoffelmeer AN, Binnekade R, Vanderschuren LJ. Dopa-
minergic mechanisms mediating the incentive to seek cocaine and heroin
following long-term withdrawal of IV drug self-administration. Psycho-
pharmacology (Berl) 143: 254 –260, 1999.
Fu Y, Liu J, Gallagher JP, Shinnick-Gallagher P. Cocaine withdrawal
enhances corticotropin releasing factor (CRF) dependent long-term poten-
tiation (LTP) in the central amygdala. Soc Neurosci Abstr 855: 14, 2004.
Fu Y, Shinnick-Gallagher P. Two intra-amygdaloid pathways to the central
amygdala exhibit different mechanisms of long-term potentiation. J Neuro-
physiol 93: 3012–3015, 2005.
Goussakov I, Chartoff EH, Tsvetkov E, Gerety LP, Meloni EG, Carlezon
WA, Bolshakov VY. LTP in the lateral amygdala during cocaine with-
drawal. Eur J Neurosci 23: 239 –250, 2006.
Hayes RJ, Vorel SR, Spector J, Liu X, Gardner EL. Electrical and chemical
stimulation of the basolateral complex of the amygdala reinstates cocaine-
seeking behavior in the rat. Psychopharmacology (Berl) 168: 75– 83, 2003.
Kalivas PW, Alesdatter JE. Involvement of N-methyl-D-aspartate receptor
stimulation in the ventral tegmental area and amygdala in behavioral
sensitization to cocaine. J Pharmacol Exp Ther 267: 486 495, 1993.
Kalivas PW, Duffy P, DuMars LA, Skinner C. Behavioral and neurochem-
ical effects of acute and daily cocaine administration in rats. J Pharmacol
Exp Ther 245: 485– 492, 1988.
Lin CH, Yeh SH, Lin CH, Lu KT, Leu TH, Chang WC, Gean PW. A role
for the PI-3 kinase signaling pathway in fear conditioning and synaptic
plasticity in the amygdala. Neuron 31: 841– 851, 2001.
Liu J, Yu B, Neugebauer V, Grigoriadis DE, Rivier J, Vale WW, Shin-
nick-Gallagher P, Gallagher JP. Corticotropin-releasing factor and Uro-
cortin I modulate excitatory glutamatergic synaptic transmission. J Neurosci
24: 4020 4029, 2004.
Liu J, Yu B, Orozco-Cabal L, Grigoriadis DE, Rivier J, Vale WW,
Shinnick-Gallagher P, Gallagher JP. Chronic cocaine administration
switches corticotropin-releasing factor2 receptor-mediated depression to
facilitation of glutamatergic transmission in the lateral septum. J Neurosci
25: 577–583, 2005.
Loftis JM, Janowsky A. Regulation of NMDA receptor subunits and nitric
oxide synthase expression during cocaine withdrawal. J Neurochem 75:
2040 –2050, 2000.
Nasif FJ, Hu XT, White FJ. Repeated cocaine administration increases
voltage-sensitive calcium currents in response to membrane depolarization
in medial prefrontal cortex pyramidal neurons. J Neurosci 25: 3674 –3679,
2005.
Neisewander JL, Baker DA, Fuchs RA, Tran-Nguyen LT, Palmer A,
Marshall JF. Fos protein expression and cocaine-seeking behavior in rats
after exposure to a cocaine self-administration environment. J Neurosci 20:
798 805, 2000.
Nie Z, Schweitzer P, Roberts AJ, Madamba SG, Moore SD, Siggins GR.
Ethanol augments GABAergic transmission in the central amygdala via
CRF1 receptors. Science 303: 1512–1514, 2004.
Pierce RC, Quick EA, Reeder DC, Morgan ZR, Kalivas PW. Calcium-
mediated second messengers modulate the expression of behavioral sensi-
tization to cocaine. J Pharmacol Exp Ther 286: 1171–1176, 1998.
Pollandt S, Liu J, Orozco-Cabal L, Grigoriadis DE, Vale WW, Gallagher
JP, Shinnick-Gallagher P. Cocaine withdrawal enhances long-term poten-
tiation induced by corticotropin releasing factor (CRF) at central amygdala
glutamatergic synapses via CRF
1
and PKA. Eur J Neurosci 24: 1733–1743,
2006.
Radulovic J, Sydow S, Spiess J. Characterization of native corticotropin-
releasing factor receptor type 1 (CRFR1) in the rat and mouse central
nervous system. J Neurosci Res 54: 507–521, 1998.
Reimer AR, Martin-Iverson MT. Nimodipine and haloperidol attenuate
behavioural sensitization to cocaine but only nimodipine blocks the estab-
lishment of conditioned locomotion induced by cocaine. Psychopharmacol-
ogy (Berl) 113: 404 410, 1994.
Richter RM, Pich EM, Koob GF, Weiss F. Sensitization of cocaine-
stimulated increase in extracellular levels of corticotropin-releasing factor
from the rat amygdala after repeated administration as determined by
intracranial microdialysis. Neurosci Lett 187: 169 –172, 1995.
Richter RM, Weiss F. In vivo CRF release in rat amygdala is increased during
cocaine withdrawal in self-administering rats. Synapse 32: 254 –261, 1999.
Robinson TE, Berridge KC. The neural basis of drug craving: an incentive-
sensitization theory of addiction. Brain Res Brain Res Rev 18: 247–291,
1993.
Roozendaal B, Brunson KL, Holloway BL, McGaugh JL, Baram TZ.
Involvement of stress-released corticotropin-releasing hormone in the baso-
lateral amygdala in regulating memory consolidation. Proc Natl Acad Sci
USA 99: 13908 –13913, 2002.
Sarnyai Z, Hohn J, Szabo G, Penke B. Critical role of endogenous cortico-
tropin-releasing factor (CRF) in the mediation of the behavioral action of
cocaine in rats. Life Sci 51: 2019 –2024, 1992.
Sarnyai Z, Shaham Y, Heinrichs SC. The role of corticotropin-releasing
factor in drug addiction. Pharmacol Rev 53: 209 –243, 2001.
Shinnick-Gallagher P, Pitkanen A, Shekhar A, Cahill L. The Amygdala in
Brain Function: Basic and Clinical Implications. New York: New York
Academy of Sciences, 2003.
Thomas KL, Arroyo M, Everitt BJ. Induction of the learning and plasticity-
associated gene Zif268 following exposure to a discrete cocaine-associated
stimulus. Eur J Neurosci 17: 1964 –1972, 2003.
Thompson AM, Gosnell BA, Wagner JJ. Enhancement of long-term poten-
tiation in the rat hippocampus following cocaine exposure. Neuropharma-
cology 42: 1039 –1042, 2002.
Thompson AM, Swant J, Gosnell BA, Wagner JJ. Modulation of long-term
potentiation in the rat hippocampus following cocaine self-administration.
Neuroscience 127: 177–185, 2004.
Tian JB, Bishop GA. Frequency-dependent expression of corticotropin re-
leasing factor in the rat’s cerebellum. Neuroscience 121: 363–377, 2003.
Turchan J, Maj M, Przewlocka B. The effect of drugs of abuse on NMDAR1
receptor expression in the rat limbic system. Drug Alcohol Depend 72:
193–196, 2003.
Ungless MA, Singh V, Crowder TL, Yaka R, Ron D, Bonci A. Cortico-
tropin-releasing factor requires CRF binding protein to potentiate NMDA
receptors via CRF receptor 2 in dopamine neurons. Neuron 39: 401–407,
2003.
Wang HL, Wayner MJ, Chai CY, Lee EH. Corticotrophin-releasing factor
produces a long-lasting enhancement of synaptic efficacy in the hippocam-
pus. Eur J Neurosci 10: 3428 –3437, 1998.
Zinebi F, Xie J, Liu J, Russell RT, Gallagher JP, McKernan MG,
Shinnick-Gallagher P. NMDA currents and receptor protein are downregu-
lated in the amygdala during maintenance of fear memory. J Neurosci 23:
10283–10291, 2003.
Zorrilla EP, Valdez GR, Weiss F. Changes in levels of regional CRF-like-
immunoreactivity and plasma corticosterone during protracted drug with-
drawal in dependent rats. Psychopharmacology (Berl) 158: 374 –381, 2001.
Report
941COCAINE WITHDRAWAL-ENHANCED LTP REQUIRES CRF
1
J Neurophysiol VOL 97 JANUARY 2007 www.jn.org
by 10.220.33.2 on May 25, 2017http://jn.physiology.org/Downloaded from
... To further establish that fear conditioning evokes plasticity in CRF neurons, we quantified LTP induction in fluorescently identified neurons from shocked and non-shocked control mice ( Figure 2F and 2G). Control mice that received only cue presentations displayed robust LTP in CRF neurons of the CeA L following high frequency stimulation of the LA (Fu et al., 2007). The same stimulation pattern failed to induce LTP in shock conditioned mice, consistent with the occlusion of LTP induction. ...
... EPSCs were evoked as described above at 0.5 Hz for a 10 min baseline period. LTP was induced with a high frequency stimulation protocol consisting of 1 ms stimuli at 100 Hz for 1 second, repeated 5 times at 3 minute intervals (Fu et al., 2007) . ...
Article
Full-text available
Fear is a graded central motive state ranging from mild to intense. As threat intensity increases, fear transitions from discriminative to generalized. The circuit mechanisms that process threats of different intensity are not well resolved. Here, we isolate a unique population of locally projecting neurons in the central nucleus of the amygdala (CeA) that produce the neuropeptide corticotropin-releasing factor (CRF). CRF-producing neurons and CRF in the CeA are required for discriminative fear, but both are dispensable for generalized fear at high US intensities. Consistent with a role in discriminative fear, CRF neurons undergo plasticity following threat conditioning and selectively respond to threat-predictive cues. We further show that excitability of genetically isolated CRF-receptive (CRFR1) neurons in the CeA is potently enhanced by CRF and that CRFR1 signaling in the CeA is critical for discriminative fear. These findings demonstrate a novel CRF gain-control circuit and show separable pathways for graded fear processing.
... However, cocaine is known to be associated with molecular and functional changes in synapses; for example, cocaine-facilitated neuroplasticity has been observed in several brain structures, including amygdala (e.g. Goussakov et al. (2006), Fu et al. (2007)) and hippocampus (e.g. Thompson et al. (2002), Thompson et al. (2004), Thompson et al. (2005), Therefore, we suggest the increased hippocampal SV2A density to be linked with cocaine-induced changes in synapses which may occur after a single drug exposure. ...
Article
Full-text available
Objective: Cocaine is a highly addictive psychostimulant that affects synaptic activity with structural and functional adaptations of neurons. The transmembrane synaptic vesicle glycoprotein 2A (SV2A) of pre-synaptic vesicles is commonly used to measure synaptic density, as a novel approach to the detection of synaptic changes. We do not know if a single dose of cocaine suffices to affect pre-synaptic SV2A density, especially during adolescence when synapses undergo intense maturation. Here, we explored potential changes of pre-synaptic SV2A density in target brain areas associated with the cocaine-induced boost of dopaminergic neurotransmission, specifically testing if the effects would last after the return of dopamine levels to baseline. Methods: We administered cocaine (20 mg/kg i.p.) or saline to rats in early adolescence, tested their activity levels, and removed the brains 1 hour and 7 days after injection. To evaluate immediate and lasting effects, we did autoradiography with [3H]UCB-J, a specific tracer for SV2A, in medial prefrontal cortex, striatum, nucleus accumbens, amygdala, and dorsal and ventral areas of hippocampus. We also measured the striatal binding of [3H]GBR-12935 to test cocaine's occupancy of the dopamine transporter at both times of study. Results: We found a significant increase of [3H]UCB-J binding in the dorsal and ventral sections of hippocampus 7 days after the cocaine administration compared to saline-injected rats, but no differences 1 hour after the injection. The [3H]GBR-12935 binding remained unchanged at both times. Conclusion: Cocaine provoked lasting changes of hippocampal synaptic density after a single exposure during adolescence.
... A detailed overview of alterations in CRF signaling that emerge with chronic drug exposure is beyond the scope of this review. However, changes in CRF signaling in a variety of brain regions including the amygdala (see e.g., [97,[224][225][226][227][228][229][230][231][232]), hippocampus (see e.g., [233]), BNST (see e.g., [85,[234][235][236]), septum (see e.g., [237]), prefrontal cortex (see e.g., [238]), nucleus accumbens (see e.g., [239,240]), serotonergic raphe nuclei (see e.g., [241][242][243]), noradrenergic cell groups (see e.g., [244,245]), and VTA [88,119,120,[246][247][248] have been demonstrated. The functional consequences of altered CRF signaling likely extend beyond dysphoric effects and may include CRF-dependent alterations in decision making [249], reward conditioning [250], drug memory reconsolidation [156] and cognition [251]. ...
Article
Full-text available
The neuropeptide, corticotropin releasing factor (CRF), has been an enigmatic target for the development of medications aimed at treating stress-related disorders. Despite a large body of evidence from preclinical studies in rodents demonstrating that CRF receptor antagonists prevent stressor-induced drug seeking, medications targeting the CRF-R1 have failed in clinical trials. Here, we provide an overview of the abundant findings from preclinical rodent studies suggesting that CRF signaling is involved in stressor-induced relapse. The scientific literature that has defined the receptors, mechanisms and neurocircuits through which CRF contributes to stressor-induced reinstatement of drug seeking following self-administration and conditioned place preference in rodents is reviewed. Evidence that CRF signaling is recruited with repeated drug use in a manner that heightens susceptibility to stressor-induced drug seeking in rodents is presented. Factors that may determine the influence of CRF signaling in substance use disorders, including developmental windows, biological sex, and genetics are examined. Finally, we discuss the translational failure of medications targeting CRF signaling as interventions for substance use disorders and other stress-related conditions. We conclude that new perspectives and research directions are needed to unravel the mysterious role of CRF in substance use disorders.
... Cocaine leads to higher spine density [86][87][88] and alteration of spine morphology [89,90]. Chronic cocaine use also enhances long-term potentiation in different brain regions [91][92][93]. Thus, spine morphology is reasonably linked to the learning and memory. ...
Preprint
HIV infection of the central nervous system causes HIV-associated neurocognitive disease (HAND) in up to 50% HIV-infected individuals. Cocaine use is prevalent in the HIV-infected population and has been shown to facilitate the HAND progression. However, the cellular and molecular mechanism of the cocaine-facilitated HAND progression remains largely unknown. In this study, we took advantage of the doxycycline inducible and brain-specific HIV Tat transgenic mouse model (iTat) of HAND and characterized effects of chronic cocaine exposure and long-term Tat expression on HAND-associated neurology and neuropathology. We found that cocaine exposure worsened the learning and memory of iTat mice, coupled with dendritic spine swelling, increased synaptophysin expression, and diminished microglia and astrocyte activation. We then employed the single-base resolution whole genome bisulfate sequencing and RNA sequencing and identified 14,838 hypermethylated CpG-related differentially methylated regions (DMR) and 15,800 hypomethylated CpG-related DMR that were linked to 52 down- and 127 up-regulated genes by cocaine and Tat. We further uncovered these genes to be mostly enriched at neuronal function- and cell morphology- and synapse formation-related ECM-receptor interaction pathway, and to be linked to behavioral and pathological changes altered by cocaine and Tat. Eight mostly affected genes included four in microglia Ift172, Eif2ak4, Pik3c2a, and Phf8, two in astrocytes Garem1 and Adgrb3, and two in neurons Dcun1d4 and Adgrb3. These findings demonstrated for the first time that cocaine and Tat interactively contributed to HAND neurology and neuropathology through genome-wide changes of DNA methylation and gene expression and suggest that targeting epigenetic changes serves as a potentially new therapeutic strategy to treat cocaine use disorder in people living with HAND.
... After repeated drug exposure, the limbic regions are highly vulnerable to undergo neuroplastic and/or neurodegenerative changes (Figure 2). A reduced gray matter volume is often found in the prefrontal cortex, hippocampus, and amygdala of chronic drug users [7, 10, 12, 59,118], together with a dysregulated expression of genes including those involved in GABA and glutamate neurotransmission [119,120] and alteration in LTP or LTD processes [121][122][123][124]. Particularly, alcohol is associated with severe brain damage and neurotoxicity in the limbic system [12], and sufficient exposure may precipitate severe neurocognitive syndromes such as lasting dementia [125]. ...
... In addition to the well-studied role of CRF in the hippocampus, a number of independent studies have investigated the effects of CRF and related ligands, as well as their receptors, by means of electrophysiological methods in various amygdalar nuclei and in the LS, especially in the context of cocaine effects (187,191,335,372,373,449,450,481). In addition, there are extensive studies on the role of CRF in NA (LC) and dopaminergic nuclei (VTA). ...
Article
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1-3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
Article
Full-text available
Cocaine use is a major comorbidity of HIV-associated neurocognitive disorder (HAND). In this study, we show that cocaine exposure worsens the learning and memory of doxycycline-inducible and brain-specific HIV Tat transgenic mice (iTat) and results in 14,838 hypermethylated CpG-related differentially methylated regions (DMRs) and 15,800 hypomethylated CpG-related DMRs, which are linked to 52 down- and 127 upregulated genes, respectively, in the hippocampus of iTat mice. These genes are mostly enriched at the neuronal function-, cell morphology-, and synapse formation-related extracellular matrix (ECM) receptor-ligand interaction pathway and mostly impacted in microglia. The accompanying neuropathological changes include swollen dendritic spines, increased synaptophysin expression, and diminished glial activation. We also find that sex (female) and age additively worsen the behavioral and pathological changes. These findings together indicate that chronic cocaine and long-term Tat expression interactively contribute to HAND, likely involving changes of DNA methylation and ECM receptor-ligand interactions.
Article
Perfluorooctanoic acid (PFOA), a hazardous environmental pollutant, has been found to enhance hepatic synthesis of fibroblast growth factor 21 (FGF21). FGF21 can enter the brain and increase the expression of corticotropin-releasing factor (CRF) in the paraventricular nucleus (PVN). In this study, adult male mice were orally administered PFOA to evaluate how it regulates emotion. Exposure of mice to PFOA (1 mg kg-1 bw) for 10 consecutive days (PFOA-mice) caused anxiety-like behaviors and a peroxisome proliferator-activated receptor α (PPARα)-dependent increase in hepatic FGF21 synthesis. The levels of CRF expression in not only PVN but also basolateral amygdala complex (BLA) neurons of PFOA-mice were increased via FGF receptor 1 (FGF-R1) activation. However, the microinjection of FGF-R1 or CRF 1 receptor (CRF-R1) antagonist in the BLA rather than the PVN of PFOA-mice could relieve their anxiety-like behaviors. In addition, external capsule-BLA synaptic transmission in PFOA-mice was enhanced by increasing CRF-R1-mediated presynaptic glutamate release, which was corrected by the blockade of PPARα, FGF-R1 and CRF-R1 or the inhibition of PKA. Furthermore, the threshold of frequency-dependent long-term potentiation (LTP) induction was decreased in the BLA of PFOA-mice, which depended on the activation of PPARα, FGF-R1, CRF-R1, PKA and NMDA receptor (NMDAR), whereas long-term depression (LTD) induction was unchanged. Thus, the results indicate that the exposure of male mice to PFOA (1 mg kg-1 bw) enhances CRF expression in BLA neurons by increasing hepatic FGF21 synthesis, which then enhances CRF-R1-mediated presynaptic glutamate release to facilitate NMDAR-dependent BLA-LTP induction, leading to the production of anxiety-like behaviors.
Article
Full-text available
Introduction In addition to their role in regulation of the hypothalamic-pituitary-adrenal-axis, corticotropin-releasing factor (CRF) and its related peptides, the urocortins, are important mediators of physiological and pathophysiological processes of the central nervous, cardiovascular, gastrointestinal, immune, endocrine, reproductive, and skin systems. Altered regulation of CRF-mediated adaptive responses to various stressful stimuli disrupts healthy function and might confer vulnerability to several disorders, including depression and anxiety. Methodology This narrative review was conducted through search and analysis of studies retrieved from online databases using a snowball method. Results This review covers aspects beginning with the discovery of CRF, CRF binding protein and their actions via interaction with CRF receptors type 1 and type 2. These are surface plasma membrane receptors, activation of which is associated with conformational changes and interaction with a variety of G-proteins and signaling pathways. We also reviewed the pharmacology and mechanisms of the receptor signaling modulatory activity of these receptors. Conclusion This review compiles and presents knowledge regarding the CRFergic system, including CRF related peptides, CRF binding protein, and CRF receptors, as well as some evidence that is potentially indicative of the biological roles of these entities in several physiological and pathophysiological processes.
Article
Full-text available
To examine neuronal activation associated with incentive motivation for cocaine, cocaine-seeking behavior (operant responding without cocaine reinforcement) and Fos expression were examined in rats exposed to saline and cocaine priming injections and/or a self-administration environment. Rats were first trained to self-administer cocaine or received yoked saline administration ("control"). They then received 21 daily exposures to either the self-administration environment ("extinction") or a different environment ("no extinction") without cocaine available. Extinction training, used to decrease incentive motivation for cocaine elicited by the self-administration environment, decreased cocaine-seeking behavior elicited by both the environment and the cocaine priming injection. Exposure to the self-administration environment enhanced Fos expression in the no extinction group relative to control and extinction groups in the anterior cingulate, basolateral amygdala, hippocampal CA1 region, dentate gyrus, nucleus accumbens shell and core, and central gray area, regardless of whether or not priming injections were given. The priming injections enhanced Fos expression in the ventral tegmental area, caudate putamen, substantia nigra pars reticulata, entorhinal cortex, central amygdala, lateral amygdala, arcuate nucleus, and central gray area, regardless of group. Thus, these changes likely reflect an unconditioned effect from either cocaine or injection stress. The priming injections also enhanced Fos expression in the anterior cingulate, but only in cocaine-experienced groups, suggesting that this enhancement reflects an experience-dependent motivational effect of the priming injections. The results suggest that different neural circuits may be involved in the incentive motivational effects of cocaine-paired environmental stimuli versus priming injections and that the anterior cingulate may be part of a common pathway for both.
Article
Full-text available
Corticotropin-releasing factor (CRF) is the primary factor involved in controlling the release of ACTH from the anterior pituitary and also acts as a neurotransmitter in a variety of brain systems. The actions of CRF are mediated by G-protein coupled membrane bound receptors and a high affinity CRF receptor, CRF1, has been previously cloned and functionally characterized. We have recently isolated a cDNA encoding a second member of the CRF receptor family, designated CRF2, which displays approximately 70% homology at the nucleotide level to the CRF1 receptor and exhibits a distinctive pharmacological profile. The present study utilized in situ hybridization histochemistry to localize the distribution of CRF2 receptor mRNA in rat brain and pituitary gland and compared this with the distribution of CRF1, receptor expression. While CRF1 receptor expression was very high in neocortical, cerebellar, and sensory relay structures, CRF2 receptor expression was generally confined to subcortical structures. The highest levels of CRF2 receptor mRNA in brain were evident within the lateral septal nucleus, the ventromedial hypothalamic nucleus and the choroid plexus. Moderate levels of CRF2 receptor expression were evident in the olfactory bulb, amygdaloid nuclei, the paraventricular and suraoptic nuclei of the hypothalamus, the inferior colliculus and 5-HT-associated raphe nuclei of the midbrain. CRF2-expressing cells were also evident in the bed nucleus of the stria terminalis, the hippocampal formation and anterior and lateral hypothalmic areas. In addition, CRF2 receptor mRNA was also found in cerebral arterioles throughout the brain. Within the pituitary gland, CRF2 receptor mRNA was detectable only at very low levels in scattered cells while CRF1 receptor mRNA was readily detectable in anterior and intermediate lobes. This heterogeneous distribution of CRF1 and CRF2 receptor mRNA suggests distinctive functional roles for each receptor in CRF-related systems. The CRF1 receptor may be regarded as the primary neuroendocrine pituitary CRF receptor and important in cortical, cerebellar and sensory roles of CRF. The anatomical distribution of CRF2 receptor mRNA indicates a role for this novel receptor in hypothalamic neuroendocrine, autonomic and general behavioral actions of central CRF.
Article
Full-text available
The reinforcing properties of cocaine can readily become associated with salient environmental stimuli that acquire secondary reinforcing properties. This form of classical conditioning is of considerable clinical relevance as intense craving can be evoked by the presentation of stimuli previously associated with the effects of cocaine. To understand better the neurobiology of cocaine-induced environment-specific conditioning, Fos expression was examined in the forebrain of rats exposed to an environment in which they had previously received cocaine. These results were compared to those observed following an acute injection of cocaine. Consistent with its stimulant actions, cocaine produced an increase in locomotion that was accompanied by an increase in Fos expression within specific limbic regions (cingulate cortex, claustrum, piriform cortex, lateral septal nucleus, paraventricular nucleus of the thalamus, lateral habenula, and amygdala) as well as the basal ganglia (dorsomedial striatum and nucleus accumbens). Exposure of rats to the cocaine-paired environment also produced an increase in locomotion, as compared to various control groups. In addition to this behavioral effect, conditioned subjects exhibited a significant increase in Fos expression within the cingulate cortex, claustrum, lateral septal nucleus, paraventricular nucleus of the thalamus, lateral habenula, and the amygdala, suggesting increased neuronal activity within these regions. In contrast to the dramatic effects observed within these structures, no conditional activation was observed within the piriform cortex, nucleus accumbens, or dorsal striatum, suggesting that these brain areas are not involved in the conditioned response. The present findings indicate that specific limbic regions exhibit increased neuronal activation during the presentation of cocaine-paired cues and may be involved in the formation of associations between cocaine's stimulant actions and the environment in which the drug administration occurred. Although the nucleus accumbens is necessary for the reinforcing and locomotor effects of cocaine, it does not exhibit a conditional Fos response, suggesting that different neural circuits are involved in the unconditioned and conditioned effects of cocaine.
Article
Previous studies have suggested a role for corticotropin-releasing factor (CRF) in the central nucleus of the amygdala (CeA) in the aversive and anxiogenic effects of withdrawal from opiates and ethanol. To test whether this role of CRF extends to cocaine withdrawal as well, the release of CRF in rat amygdala was monitored by intracranial microdialysis during a 12-hour session of intravenous cocaine self-administration and subsequent 12-hour cocaine withdrawal period. Cocaine self-administration tended to lower dialysate CRF concentrations to approximately 75% of CRF levels in controls. In contrast, subsequent cocaine withdrawal produced a profound increase in CRF release, which reached peak levels of approximately 400% of baseline between 11 and 12 hours post-cocaine. These results provide evidence that cocaine withdrawal activates CRF neurons in the amygdala, a site that has been implicated in emotional and anxiogenic effects of stress and drug withdrawal syndromes. Synapse 32:254–261, 1999. © 1999 Wiley-Liss, Inc.
Article
This paper presents a biopsychological theory of drug addiction, the 'Incentive-Sensitization Theory'. The theory addresses three fundamental questions. The first is: why do addicts crave drugs? That is, what is the psychological and neurobiological basis of drug craving? The second is: why does drug craving persist even after long periods of abstinence? The third is whether 'wanting' drugs (drug craving) is attributable to 'liking' drugs (to the subjective pleasurable effects of drugs)? The theory posits the following. (1) Addictive drugs share the ability to enhance mesotelencephalic dopamine neurotransmission. (2) One psychological function of this neural system is to attribute 'incentive salience' to the perception and mental representation of events associated with activation of the system. Incentive salience is a psychological process that transforms the perception of stimuli, imbuing them with salience, making them attractive, 'wanted', incentive stimuli. (3) In some individuals the repeated use of addictive drugs produces incremental neuroadaptations in this neural system, rendering it increasingly and perhaps permanently, hypersensitive ('sensitized') to drugs and drug-associated stimuli. The sensitization of dopamine systems is gated by associative learning, which causes excessive incentive salience to be attributed to the act of drug taking and to stimuli associated with drug taking. It is specifically the sensitization of incentive salience, therefore, that transforms ordinary 'wanting' into excessive drug craving. (4) It is further proposed that sensitization of the neural systems responsible for incentive salience ('for wanting') can occur independently of changes in neural systems that mediate the subjective pleasurable effects of drugs (drug 'liking') and of neural systems that mediate withdrawal. Thus, sensitization of incentive salience can produce addictive behavior (compulsive drug seeking and drug taking) even if the expectation of drug pleasure or the aversive properties of withdrawal are diminished and even in the face of strong disincentives, including the loss of reputation, job, home and family. We review evidence for this view of addiction and discuss its implications for understanding the psychology and neurobiology of addiction.
Article
This paper presents a biopsychological theory of drug addiction, the ‘Incentive-Sensitization Theory’. The theory addresses three fundamental questions. The first is: why do addicts crave drugs? That is, what is the psychological and neurobiological basis of drug craving? The second is: why does drug craving persist even after long periods of abstinence? The third is whether ‘wanting’ drugs (drug craving) is attributable to ‘liking’ drugs (to the subjective pleasurable effects of drugs)? The theory posits the following.
Article
The role of endogenous CRF in the locomotor hyperactivity induced by cocaine was investigated by using the immunoneutralization of endogenous CRF and an antagonist of CRF-receptors (alpha-helical CRF9-41: alpha h-CRF) in rats. Different dilutions of anti-CRF antibody (1:5, 1:20, but not 1:100) injected intracerebroventricularly (i.c.v.) 24 hours before the cocaine treatment blocked the expression of locomotor hyperactivity. Pretreatment with different doses (0.001, 0.01, 0.1, 1.0 micrograms i.c.v.) of alpha h-CRF inhibited the locomotor hyperactivity induced by cocaine dose-dependently. Neither the immunoneutralization nor the receptor blockade for CRF changed the hyperactivity induced by another locomotor stimulant caffeine. These results serve as indirect in vivo evidence of the selective role of endogenous CRF in the cocaine-induced behavioral alterations. The findings have implications as concerns the possible role of CRF in human psychopathological changes induced by cocaine.
Article
Daily cocaine injection into rodents produces a progressive increase in the motor stimulant effect of acute cocaine administration. In this study it was found that daily cocaine injection (15 mg/kg i.p. x 3 days) produced an enhanced motor stimulant response to acute cocaine injection. The behavioral augmentation was linear with regards to dose in horizontal activity and behavioral intensity rating, but was biphasic in vertical activity. Augmented vertical, but not horizontal, activity in response to acute cocaine was found to persist for 2 weeks after the last daily injection of cocaine. Acute injection of cocaine was found to significantly decrease the level of dopamine (DA) metabolites in the nucleus accumbens, striatum and A10 DA region. In rats pretreated with daily injections of cocaine (15 mg/kg i.p. x 3 days), an acute challenge of cocaine 14 days after the last daily injection produced a more consistent decrease in DA metabolites in the nucleus accumbens, striatum and prefrontal cortex compared to daily saline-pretreated rats. In contrast, daily cocaine treatment abolished the decrease in DA metabolites produced in the A10 region by an acute cocaine challenge. Acute injection with cocaine was found to significantly depress dopa accumulation in the A10 region, nucleus accumbens and striatum. This effect was abolished in the A10 region in rats pretreated 14 days previously with daily injections of cocaine (7.5, 15.0 or 30 mg/kg i.p. x 3 days), but remained intact in the nucleus accumbens and striatum, except after daily pretreatment with the highest dose of cocaine.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Using intracranial microdialysis, the effect of repeated cocaine (30 mg/kg i.p.) versus saline administration for 10 consecutive days upon basal and stimulated release of corticotropin-releasing factor (CRF) was examined in the central amygdaloid nucleus (CeA) of anesthetized rats. No significant differences in basal CRF levels between daily cocaine and saline treated groups were found. However, after cocaine challenge (10 mg/kg i.p.) the increase in CRF overflow was significantly greater in cocaine- as opposed to saline-pretreated rats (266 +/- 55.4% versus 149 +/- 8.5% of basal levels). Local administration of 4-aminopyridine produced a significant increase in CRF efflux (195 +/- 58.5%) in daily cocaine-treated rats with only a weak response in the control group (127 +/- 30.9%). These data demonstrate that repeated administration of cocaine enhances cocaine-induced release of CRF in the rat CeA. The sensitization of CRF release may play a significant role in psychostimulant-induced sensitization phenomena.