ArticlePDF Available

Nucleus accumbens NMDA receptor activation regulates amphetamine cross-sensitization and deltaFosB expression following sexual experience in male rats

Authors:

Figures

Content may be subject to copyright.
Nucleus accumbens NMDA receptor activation regulates
amphetamine cross-sensitization and deltaFosB expression following
sexual experience in male rats
Lauren N. Beloate
a
,
b
, Peyton W. Weems
a
,
b
, Graham R. Casey
a
, Ian C. Webb
a
,
Lique M. Coolen
a
,
c
,
*
a
Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
b
Graduate Program in Neuroscience, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
c
Department of Physiology &Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
article info
Article history:
Received 21 August 2015
Received in revised form
14 September 2015
Accepted 16 September 2015
Available online 21 September 2015
Keywords:
Glutamate
Striatum
Sensitization
Amphetamine
FosB
cFos
abstract
Sexual experience in male rats followed by a period of absti nence causes sensitization to
D
-Amphetamine
(Amph) reward, evidenced by an increased conditioned place preference (CPP) for low doses of Amph.
Moreover, sexual experience induces neural plasticity within the nucleus accumbens (NAc), including
induction of deltaFosB, which plays a key role in Amph reward cross-sensitization. The NMDA receptor
subunit NR1 is also upregulated by mating, but the functional relevance of NMDA receptors in sex
experience-induced effects is unknown. Here, we examined the inuence of intra-NAc MK 801 infusions
on sex experience-induced NAc deltaFosB and cFos expression, as well as mating- and Amph-induced
CPP in adult male rats. In experiment 1, males received MK 801 or saline into the NAc during each of
4 consecutive days of mating or handling and were tested for Amph CPP and experience-induced del-
taFosB 10 days later. Intra-NAc MK 801 during sexual behavior prevented experience-induced increases
in Amph CPP and NAc deltaFosB expression without affecting sexual behavior. In experiment 2, the ef-
fects of intra-NAc MK 801 on mating-induced CPP were examined by intra-NAc infusion of MK 801 or
saline prior to mating on conditioning days. Intra-NAc MK 801 did not affect mating-induced CPP. Next,
effects of intra-NAc MK 801 on mating-induced cFos immunoreactivity were examined. MK 801 pre-
vented mating-induced cFos expression in NAc shell and core. Together, these results provide evidence
that NAc NMDA receptor activation during sexual behavior plays a key role in mating-induced cFos and
deltaFosB expression and subsequent experience-induced cross-sensitization to Amph reward.
©2015 Published by Elsevier Ltd.
1. Introduction
Drugs of abuse cause neural plasticity within the mesolimbic
pathway, which in turn, contributes to the development and
maintenance of addiction (Chen et al., 2010; Koob and Volkow,
2010; Mameli and Luscher, 2011; Feltenstein and See, 2013;
Grueter et al., 2013; Gipson et al., 2014). The mesolimbic pathway
regulates natural reward behaviors (Frohmader et al., 2010; Olsen,
2011; Volkow et al., 2011, 2013), and repeated exposure to natural
rewards causes neural plasticity similar to that induced by drugs of
abuse (Solinas et al., 2008; Pitchers et al., 2010b, 2013, 2014; Olsen,
2011; Nader et al., 2012; Adams et al., 2013; Bardo et al., 2013),
suggesting that drugs of abuse act on the same plasticity mecha-
nisms that regulate natural reward learning (Pitchers et al., 2013,
2014). Indeed, natural reward experience inuences subsequent
drug-seeking behavior in rodent models. For example, pair bonding
and environmental enrichment can serve as a protective compo-
nent to drug-seeking behavior (Aragona et al., 2007; Solinas et al.,
2008; Gipson et al., 2011; Liu et al., 2011; Burkett and Young,
2012; Puhl et al., 2012). Conversely, social isolation or the
Abbreviations: 3V, third ventricle; ac, anterior commissure; Amph,
D
-Amphet-
amine; ANOVA, analysis of variance; BLA, basolateral amygdala; CPP, conditioned
place preference; DAB, 3,3
0
-diaminobenzidine tetrahydrochloride; i.p., intraperito-
neally; MK 801, (5S,10R)-(þ)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-
5,10-imine maleate; mPFC, medial prefrontal cortex; MPN, medial preoptic nucleus;
mPOA, medial preoptic area; NAc, nucleus accumbens; PB, phosphate buffer; PBS,
phosphate buffered saline; s.c., subcutaneous.
*Corresponding author. Department of Physiology &Biophysics, University of
Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
E-mail address: lcoolen@umc.edu (L.M. Coolen).
Contents lists available at ScienceDirect
Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm
http://dx.doi.org/10.1016/j.neuropharm.2015.09.023
0028-3908/©2015 Published by Elsevier Ltd.
Neuropharmacology 101 (2016) 154e164
removal of environmental enrichment can cause an increased
vulnerability to drug seeking (Lu et al., 2003; Raz and Berger, 2010;
Nader et al., 2012; Neisewander et al., 2012).
Repeated experience with sexual behavior causes subsequent
sensitization of drug-induced locomotor activity in female ham-
sters (Bradley and Meisel, 2001) and male rats (Pitchers et al.,
2010a, 2012). Moreover, in male rats sexual experience causes
sensitized conditioned place preference (CPP) for low doses of the
psychostimulant,
D
-Amphetamine (Amph) (Pitchers et al., 2010a),
which is dependent on a period of sexual abstinence and is long-
lasting (Pitchers et al., 2010a, 2013). Sexual experience also cau-
ses neural alterations in the nucleus accumbens (NAc) and ventral
tegmental area (VTA), including increased dendritic arborization
and spine density in NAc medium spiny neurons (Pitchers et al.,
2010a, 2013; Staffend et al., 2014), reduction of VTA dopamine
cell soma size (Pitchers et al., 2014), and upregulation of the tran-
scription factor deltaFosB, in both NAcand VTA (Meisel and Mullins,
2006; Pitchers et al., 2010b, 2013). Hence, our laboratory has pro-
posed that these neural alterations contribute to the effects of
sexual experience on cross-sensitization to Amph reward.
Glutamate is a key mediator of drug-induced neural plasticity in
the NAc (Kalivas and Volkow, 2011; Loweth et al., 2014; Pomierny-
Chamiolo et al., 2014; van Huijstee and Mansvelder, 2014). The NAc
receives multiple glutamatergic inputs, including those from the
medial prefrontal cortex (mPFC), basolateral amygdala (BLA) and
hippocampus (Britt et al., 2012; Papp et al., 2012; Tye, 2012), but a
role for glutamate in the regulation of sexual behavior or for the
effects of sexual experience on neural and behavioral plasticity has
not been established. We have previously demonstrated that sexual
experience inuences the function, expression and distribution of
glutamate receptors in the NAc of male rats. In particular, patch
clamp studies showed reduced AMPA/NMDA ratios in the post-
synaptic response to frontal cortex inputs, with no alterations in
presynaptic inputs (Pitchers et al., 2012). Moreover, biochemical
analysis showed that expression of the NMDA receptor NR1 subunit
was upregulated shortly following sexual behavior, whereas AMPA
receptor GluA1 and 2 subunits were upregulated following pro-
longed periods of reward abstinence (Pitchers et al., 2012). These
results suggest an initial activation and increase in NMDA receptors
during sexual experience, followed by increased synthesis and
trafcking of AMPA receptors during the abstinence period. How-
ever, the functional relevance of NMDA receptor activation during
sexual behavior for experience-induced neuroplasticity and cross-
sensitization to Amph reward remains unknown and will be the
focus of the current set of studies.
As mentioned above, sexual experience causes long-lasting
expression of deltaFosB in the NAc (Meisel and Mullins, 2006;
Pitchers et al., 2010b, 2013). DeltafosB is also persistently
expressed in reward-related brain regions, including the NAc, in
response to drugs of abuse (Perrotti et al., 2008; Robison and
Nestler, 2011) and natural rewards, such as food (Teegarden et al.,
2009) and sucrose consumption (Wallace et al., 2008;
Christiansen et al., 2011), wheel running (Werme et al., 2002;
Greenwood et al., 2011) and environmental enrichment (Solinas
et al., 2008). DeltaFosB expression in the NAc has been shown to
play a causal role in the sensitivity to drugs of abuse, particularly
psychostimulants (Kelz et al., 1999; Nestler, 2008; Grueter et al.,
2012; Robison et al., 2013). In addition, deltaFosB expression in
the NAc is essential for the effects of sex experience on cross-
sensitization of Amph reward (Pitchers et al., 2010b, 2013) and
facilitation of sexual behavior (Pitchers et al., 2012). In the NAc,
dopamine plays a major role in the induction of deltaFosB. It is
induced in D1 receptor neurons in response to drugs of abuse (Lee
et al., 2006; Kim et al., 2009; Lobo et al., 2013), and drug- and sex-
induced deltaFosB induction is prevented by D1 receptor
antagonism (Muller and Unterwald, 2005; Pitchers et al., 2013) and
absent in D1 receptor mutant mice (Zhang et al., 2002). Besides the
role for dopamine in activation of deltaFosB, there is also limited
evidence that NMDA receptor activation may play a role, as treat-
ment with NMDA receptor agonists increases striatal deltaFosB
(Hollen et al., 1997). However, the involvement of NMDA receptor
activation in induction of this transcription factor by natural or drug
rewards is largely unexplored. Therefore, here we examine the role
of NMDA receptor activation in sex-induced deltaFosB in the NAc.
In conclusion, the current study had four objectives. First, we
tested the role of NMDA receptor activation in the NAc for the
initiation, facilitation and reward of sexual behavior in male rats.
Next, we tested the hypothesis that sex experience-induced Amph
reward cross-sensitization is dependent on NMDA receptor acti-
vation in the NAc during mating. And third, the contribution of
NMDA receptor activation for sexual experience-induced deltaFosB
was examined. In particular, the effects of intra-NAc infusions of the
NMDA receptor antagonist, MK 801, on sex behavior, experience-
induced Amph CPP and NAc deltaFosB expression were deter-
mined. Finally, as NMDA receptor activation also contributes to the
induction of cFos in the NAc by drugs of abuse (Liu et al., 1994;
Hussain et al., 2001; Yanahashi et al., 2004) and in the medial
preoptic nucleus (MPN) by sex behavior (Dominguez et al., 2007),
we tested the hypothesis that NAc NMDA receptor activity regu-
lates mating-induced cFos expression in the NAc.
2. Materials and methods
2.1. Animals
Young adult male Sprague Dawley rats (Charles River, Wil-
mington, MA; 225e250 g) were housed in same sex pairs (of
identical treatment groups) in standard Plexiglas cages. Food and
water were provided ad libitum, and animals were maintained in
temperature and humidity-controlled rooms on a 12/12 h darke-
light cycle with lights off at 6e8 am. Female Sprague Dawley rats
(Charles River; 210e225 g) were bilaterally ovariectomized and
implanted with subcutaneous (s.c.) capsules (Dow Corning tubing,
Midland, MI; 1.98 mm internal diameter) containing 5% 17-
b
-
estradiol-benzoate (in cholesterol; SigmaeAldrich, St. Louis, MO;
1cmlled area) and received 500
m
g progesterone in 0.1 ml of
sesame oil (SigmaeAldrich; s.c.) 3e6 h prior to each mating session
to induce sexual receptivity. All experiments were carried out in
accordance with the National Institutes of Health guidelines
involving vertebrate animals in research and were approved by the
Institutional Animal Care and Use Committee at the University of
Mississippi Medical Center. All efforts were made to minimize an-
imal suffering and to reduce the number of animals used. Alter-
natives to in vivo techniques were not available.
2.2. Cannulae implantation surgeries
Each male was implanted with bilateral cannulae aimed to-
wards the NAc as described previously (Pitchers et al., 2013). Briey,
animals were deeply anesthetized with ketamine (87 mg/ml/kg)/
xylazine (13 mg/ml/kg; i.p.) and placed into a stereotaxic apparatus
(Kopf Instruments, Tujunga, CA). Bilateral 21 gauge guide cannulae
(Plastics One, Roanoke, VA; 2.4 mm wide, 6.4 mm below pedestal)
were directed at the NAc at þ1.7 A/P and ±1.2 M/L from Bregma and
6.4 D/V from skull. Cannulae placement and drug injections were
aimed to target both NAc shell and core, as sex-induced deltaFosB
and cFos expression is comparable in both NAc subareas (Pitchers
et al., 2013). Animals received 5 mg/ml/kg carprofen (s.c.) during
surgery and 24 h later for analgesia, and were allowed to recover for
10 days prior to onset of experiments.
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e164 155
2.3. Drugs
Males received local NAc infusions of the NMDA receptor
antagonist, (5S,10R)-(þ)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]
cyclohepten-5,10-imine maleate (MK 801; Tocris, Bristol, UK Cat.
No. 0924; 1
m
g/1
m
lor1
m
g/0.5
m
l in saline), at a ow rate of 1
m
l/
min over a 1 min interval, followed byan additional 1 min with the
injection cannula (1 mm projection below guide cannula) left in
place. This dose was chosen based on previous studies showing an
effect on drug- and natural reward-related behavior (Leonibus
et al., 2002; Reynolds and KC, 2003; Wang et al., 2005; Hunt
et al., 2010; Kawasaki et al., 2011). The higher volume is based on
our previous studies and was shown to target both NAc shell and
core (Pitchers et al., 2013). Amph (
D
-Amphetamine hemisulfate
salt; SigmaeAldrich product no. A5880; 0.5 mg/ml/kg calculated on
basis of the free base; s.c.) was administered during the CPP para-
digm. At this dose, sexually experienced rats develop a CPP while
naïve rats do not (Pitchers et al., 2010a, 2013).
2.4. Sexual behavior testing
All sexual behavior testing took place 3e6 h after lights off,
under dim red lighting. Males were placed in a mating arena
(60 45 50 cm) with clean bedding for 5 min, after which they
mated with a receptive female until one ejaculation. If animals did
not reach ejaculation, tests were terminated one hour after intro-
duction of the female. Males that were assigned to sexually expe-
rienced groups mated during 4 subsequent daily sessions. Males
assigned to the sexually naïve groups were placed in identical
testing cages with clean bedding for 30 min, but did not receive
sexually receptive females. Naïve males were housed and handled
in the same rooms as the sexually experienced males and thus
subjected to distant female odors and identical noise and
disturbances.
2.5.
D
-Amphetamine conditioned place preference
CPP for Amph was conducted as previously described (Pitchers
et al., 2010a, 2013; Frohmader et al., 2011). The apparatus consists
of two main chambers separated by a smaller middle chamber, each
distinguishable by visual and tactile cues (Med Associates, St.
Albans, VT). The apparatus is unbiased, but to determine each an-
imal's initial preference, a 15 min pre-test was conducted, in which
males freely roamed the CPP apparatus. As a group, animals did not
differ in their initial preference for each of the two main chambers.
Amph or saline was paired with either CPP chamber in a counter-
balanced manner (unbiased design) during two conditioning days
(single pairing) for 30 min. The change in preference was deter-
mined during a post-test that was procedurally identical to the pre-
test. The difference in time spent in the drug-paired chamber
during the post-test minus the pre-test were calculated for each
animal and termed the CPP score.
2.6. Mating-induced conditioned place preference
CPP for mating was tested as described previously (Tenk et al.,
2009;Davis et al., 2010; Di Sebastiano et al., 2011; Frohmader
et al., 2011) and the procedure was similar to that for Amph CPP.
A 15 min pre-test determined the initial preference for each animal.
As a group, the animals showed no preference. The sex-paired
chamber was assigned to the initially non-preferred chamber
(biased design). During conditioning, males were placed into the
sex-paired chamber (single pairing) immediately after ejaculation
for 30 min. On the alternate conditioning day, animals were placed
into the non sex-paired chamber for 30 min. The order of the two
conditioning days was counterbalanced. Preference scores (% time
spent in sex-paired chamber) and difference scores (time spent in
paired unpaired chamber) were calculated for each animal.
2.7. Immunohistochemistry
2.7.1. Tissue preparation
Animals received an overdose of sodium pentobarbital (Vortech
Pharmaceutical Ltd., Dearborn, MI; 390 mg/ml/kg; i.p.) and were
transcardially perfused with 10 ml saline (0.9% NaCl (Sigma-
eAldrich) in ddH
2
O) and 500 ml of 4% paraformaldehyde (Electron
Microscopy Sciences, Hateld, PA; in 0.1 M phosphate buffer (PB)).
The brains were removed and post-xed for one hour in the same
xative at room temperature and stored in a sucrose solution
(Fisher Scientic, Fair Lawn, NJ; 20% in 0.1 M PB containing 0.01%
sodium azide (SigmaeAldrich); at 4
C). Brains were sectioned
coronally (35
m
m) into 4 parallel series, using a freezing microtome
(SM 2000R, Leica Biosystems, Lawrenceville, GA) and stored
in 20
C in cryoprotectant solution (30% sucrose in 0.1 m PB
containing 30% ethylene glycol (Fisher Scientic) and 0.01% sodium
azide).
2.7.2. DeltaFosB and cFos immunoprocessing
Free oating sections were thoroughly washed in phosphate-
buffered saline (PBS; pH 7.4) at room temperature under gentile
agitation between each incubation. Tissue was exposed to 1% H
2
O
2
for 10 min and incubation solution consisting of PBS containing
0.1% bovine serum albumin (Fisher Scientic) and 0.4% Triton X-100
(SigmaeAldrich) for 1 h, followed by incubation in either pan-FosB
rabbit polyclonal antibody raised against an internal region shared
by FosB and deltaFosB (Santa Cruz Biotechnology, Santa Cruz, CA;
sc-48; 1:5000 in incubation solution) or rabbit anti-cFos (Santa
Cruz Biotechnology; sc-52; 1:2500 in incubation solution) for 17 h.
Sections were then incubated in biotinylated goat anti-rabbit IgG
(Vector Laboratories, Burlingame, CA; 1:500 in incubation solution;
1 h), avidinebiotin-horseradish peroxidase (ABC Elite; Vector
Laboratories; 1:1000 in PBS; 1 h) and 0.02% 3,3
0
-diaminobenzidine
tetrahydrochloride (DAB; SigmaeAldrich) with 0.02% nickel sulfate
(SigmaeAldrich) and 0.015% H
2
O
2
in 0.1 M PB (10 min). Sections
were mounted onto Superfrost plus glass slides (Fisher Labora-
tories), dehydrated and coverslipped with dibutyl phthalate xylene
(Electron Microscopy Sciences). Both primary antibodies have been
previously validated to specically visualize deltaFosB or cFos un-
der these conditions (Perrotti et al., 2004, 2008; Di Sebastiano et al.,
2010, 2011; Pitchers et al. 2010b, 2013).
2.7.3. Cannulae placement verication
The placement of cannulae was conrmed using sections
stained for cFos or deltaFosB. Only animals with correct placement
were included in analyses. For clarity, nal numbers of animals for
each of the experiments are listed in the appropriate gure legends.
3. Experimental design
3.1. Experiment 1: effects of intra-NAc MK 801 on sexual behavior
and experience-induced Amph CPP and NAc deltaFosB expression
The main goals of this study were to investigate if intra-NAc MK
801 during mating experience would prevent experience-induced
Amph CPP sensitization and NAc deltaFosB expression.
Sexually experienced and naïve groups of males received intra-
NAc MK 801 (1
m
g in 0.5
m
lor1
m
l saline) or saline 15 (1
m
l) min prior
to each of the four daily mating or handling sessions. Latencies to
rst mount, intromission and ejaculation were analyzed between
days 1 and 4 for all groups using a two-way analysis of variance
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e16 4156
(ANOVA; drug day) and StudenteNewmaneKeuls post-hoc test.
Latencies to rst mount, intromission and ejaculation between days
1 and 4 were also analyzed within each group using a one-tailed
ManneWhitney rank sum test. 95% condence levels were used
for all tests. Animals were included in analyses only for the sexual
parameters displayed for each of the sessions.
Ten days following the last mating or handling session, males
were tested for Amph CPP. CPP scores (time spent in the paired
chamber during the post-test minus the pre-test) were compared
between groups and analyzed using a two-way ANOVA (drug sex)
and HolmeSidak post-hoc tests. 95% condence levels were used for
all tests. Animals were excluded from analysis if they displayed an
initial preference of 120 s or more forone of the 3 chambers, had CPP
scores that were more than 2 standard deviations away from the
group mean, did not reach three or more days with one ejaculation
during sexual experience, or had missed cannula placements.
One day after the Amph CPP post-test, brains were collected and
analyzed for deltaFosB. DeltaFosB-immunoreactive cells were
quantied in the NAc shell and core using a standard area of
analysis (400 600
m
m per subregion) on a light microscope with a
drawing tube attachment (Leica DMR; Leica Microsystems, Inc.,
Buffalo Grove, IL) as described previously (Pitchers et al., 2010b,
2013). Briey, at least two NAc sections per subregion, per animal
were counted and averaged, then a group average was calculated.
Dorsal striatum (200 600
m
m area of analysis) immuno-positive
cells were counted to check for drug spread. Group averages were
compared between groups using a two-way ANOVA (drug sex
experience) and HolmeSidak post-hoc tests. 95% condence levels
were used for all tests.
3.2. Experiment 2: effects of intra-NAc MK 801 on mating-induced
CPP and NAc cFos expression
The main goals of this study were to investigate if intra-NAc MK
801 during mating would prevent sexual reward and mating-
induced neuronal activation in the NAc. Males received intra-NAc
MK 801 (1
m
g in 0.5
m
lor1
m
l saline) or saline 15 min prior to
introduction of female on the sex conditioning day for CPP. La-
tencies to rst mount, intromission and ejaculation were recorded
and compared between groups using a one-way ANOVA on ranks.
CPP preference and difference scores were compared between pre-
and post-tests, within groups, using unpaired t-tests. 95% con-
dence levels were used for all tests. Animals were excluded if they
spent more than 120 s in one of the three chambers.
One day following the sex CPP post-test, the same males
received intra-NAc MK 801 (1
m
g in 0.5
m
lor1
m
l saline) or saline
and either mated or were placed in the cages without females. One
hour after introduction of females, brains were collected and
analyzed for cFos expression. cFos immunoreactive cells were
quantied in the NAc shell and core with a standard area of analysis
(400 600
m
m per subregion) on a light microscope with a drawing
tube attachment (Leica DMR) as described in Experiment 1. At least
two NAc sections per subregion per animal were counted and
averaged, then a group average was calculated. medial preoptic
nucleus (MPN) (600 800
m
m area of analysis) immuno-positive
cells were counted to check for drug spread. The no mating
groups did not differ between drug treatments on any measures
and were combined for analysis. Groups were compared using a
one-way ANOVA and Fisher LSD post hoc test, with 95% condence
levels.
4. Results
4.1. Intra-NAc MK 801 did not disrupt sexual behavior
In the rst experiment, intra-NAc infusions of MK 801 did not
affect sexual behavior on any of the four daily mating tests, and no
differences between groups were detected. Moreover, animals in all
groups showed facilitation of sexual behavior with experience
(mount latencies: F
(2,92)
¼5.413, p ¼0.022; intromission latencies:
F
(2,91)
¼6.742, p ¼0.011; ejaculation latencies: F
(2,91)
¼8.565,
p¼0.004), evidenced by signicantly lower latencies or trends in
the fourth mating session compared to the rst mating session to
mount, intromit, and ejaculate in all groups (Fig. 1AeC; p <0.05).
Thus, blockade of NMDA receptors in the NAc did not disrupt
initiation or performance of sexual behavior or the experience-
induced facilitation of sexual behavior.
4.2. Intra-NAc MK 801 prevented sex experience-induced Amph CPP
One week following sexual experience, all groups were tested
for Amph CPP (Fig. 1E). There was an overall interaction effect of
sexual experience and MK 801 treatment (F
(2,58)
¼7.774, p ¼0.001).
Specically, sexually experienced control males that received intra-
NAc vehicle injections displayed a signicantly increased CPP score
compared to vehicle-treated naïve controls (p ¼0.042), conrming
our previous ndings (Pitchers et al., 2010a, 2013). Intra-NAc in-
fusions of MK 801 during mating experience prevented this effect.
CPP scores in both of the MK 801-injected sexually experienced
groups did not differ from vehicle-treated naïve males, and CPP
scores of animals injected with 1
m
l MK 801 were signicantly
lower than vehicle-treated experienced males (p ¼0.004). Intra-
NAc infusions of MK 801 in sexually naïve controls did not affect
Amph CPP at the lower volume (0.5
m
l). However, infusions of the
higher volume (1
m
l) of MK 801 in sexually naïve males caused a
signicantly increased CPP score compared to vehicle-infused
sexually naïve males (p ¼0.016). This nding is in agreement
with effects of systemic MK 801 on psychostimulant sensitization
(Rung et al., 2005; Eyjolfsson et al., 2006; Landa et al., 2014), but in
sharp contrast to the effect of NMDA receptor blockade in sexually
experienced males in the current study.
4.3. Intra-NAc MK 801 blocked sex experience-induced deltaFosB
There were signicant main effects of sexual experience (core:
F
(1,40)
¼31.357, p <0.001; shell: F
(1,38)
¼19.129, p <0.001) and MK
801 treatment (core: F
(2,40)
¼13.660, p <0.001; shell:
F
(2,38)
¼16.532, p <0.001) and a signicant interaction effect (core:
F
(2,40)
¼20.949, p <0.001; shell: F
(2,38)
¼18.498, p <0.001) on NAc
deltaFosB. Sexually experienced vehicle-treated males had signi-
cantly more deltaFosB immunoreactive cells in the NAc core
(Fig. 2G) and shell (Fig. 2H) compared to naïve saline-treated con-
trols (p <0.001), as reported previously (Pitchers et al., 2010b,
2013). Intra-NAc MK 801 injections prior to each mating session
prevented this effect. Numbers of deltaFosB cells in both MK 801-
infused sexually experienced groups did not differ from naïve
controls and were signicantly lower compared to vehicle-treated
sexually experienced males (p <0.001). Intra-NAc MK 801 did
not affect deltaFosB expression in naïve males. Finally, the MK 801
infusions appeared to be restricted to the NAc and did not reduce
sex experience-induced deltaFosB expression in the dorsal stria-
tum. Specically, in the medial caudate putamen, an area imme-
diately dorsal to the NAc, both sexually experienced vehicle- and
1
m
l MK 801-treated groups had signicantly higher numbers of
deltaFosB compared to vehicle-treated naïve males and did not
differ from each other (vehicle naive: 5.714 ±4.071; vehicle
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e164 157
experienced: 34.40 ±8.38, p <0.001; 1
m
l MK 801 experienced:
38.67 ±9.39, p <0.001).
4.4. Intra-NAc MK 801 did not prevent mating-induced CPP
The second experiment replicated the nding that intra-NAc MK
801 does not affect the initiation or performance of sexual behavior
(Table 1). Moreover, MK 801 did not prevent mating-induced CPP
(Fig. 3). Mating induced a similar CPP in MK 801- and vehicle-
treated groups, indicated by signicantly increased preference
(percentage of time in the sex-paired chamber) and difference
scores (time in paired minus unpaired chamber) during the post-
test compared to pre-test (p <0.05). The 0.5
m
l MK 801-injected
group had a trend towards higher preference score during the
Fig. 1. Intra-NAc MK 801 blocks sexual experience-induced Amph CPP but does not affect sexual behavior. (AeC) Quantitative analysis of latencies (in seconds) to rst mount (A),
rst intromission (B) or ejaculation (C) on days 1 and 4 of consecutive days of mating for groups receiving intra-NAc saline (white bars,n¼20), 0.5
m
lMK801(gray bars,n¼8) or
1
m
lMK801(black bars,n¼22). Data represent mean ±SEM. * Denotes signicant difference within group compared to day 1: A) mount latencies: vehicle: p ¼0.040, 0.5
m
lMK801:
p<0.001,1
m
lMK801:p¼0.06; B) intromission latencies: vehicle: p ¼0.043, 0.5
m
lMK801:p<0.0 01, 1
m
lMK801:p¼0.066; C) ejaculation latencies: vehicle: p ¼0.002, 0.5
m
lMK
801: p ¼0.095, 1
m
lMK801:p¼0.021; D) Coronal NAc sections indicating injection sites. Cannulae were placed bilaterally, but are shown unilaterally for easier viewing. Left side:
sexually naïve males (dotted outline); Right side: sexually experienced males (solid outline). White ll: saline; Light gray ll: 0.5
m
lMK801;Dark gray ll:1
m
lMK801.(E) CPP scores,
dened as the time spent in Amph-paired chamber during post-test minus pre-test, for sexually naïve (naive) and sexually experienced (experienced) groups that received intra-NAc
saline (white bars, naïve n ¼15, exp n ¼14), 0.5
m
lMK801(gray bars, naïve n ¼7, exp n ¼7) or 1
m
lMK801(black bars, naïve n ¼7, ex p n ¼14) during mating or handling. Data are
presented as mean ±SEM. * Indicates signicant difference compared to saline naïve group; # indicates signicant difference compared to all other sexually experienced groups.
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e16 4158
post-test compared to pre-test, although this increase did not reach
signicance (p ¼0.055).
4.5. Intra-NAc MK 801 blocked mating-induced NAc cFos
expression
Finally, intra-NAc MK 801 blocked mating-induced cFos
expression. There was a signicant interaction effect of sexual
behavior and MK 801 (F
(3,24)
¼49.196, p <0.0 01). In vehicle-treated
control animals, mating increased cFos expression in the NAc core
(Fig. 4E) and shell (Fig. 4F), compared to non-mating controls
(p <0.001). Both volumes of MK 801 prevented mating-induced
cFos in the NAc shell and core, as numbers of cFos cells in MK
801 groups did not differ from non-mating controls and were
signicantly lower compared to vehicle-treated mating males
(Fig. 4F; p 0.001). Again, intra-NAc MK 801 infusions did not
affect mating-induced cFos expression in nearby brain regions. In
particular, the was analyzed, as mating induces robust cFos in this
region (Veening and Coolen, 2014) and the MPN is located caudal to
the NAc and close to the ventricle. Indeed, in the MPN, there was a
main effect of sex behavior (F
(3,14)
¼36.725, p <0.001) but not of
MK 801 treatment, and mating-induced cFos in the MPN inde-
pendent of the intra-NAc drug infusion (Fig. 5).
5. Discussion
The current study demonstrates that NMDA receptor activation
in the NAc during sexual behavior is critical for the cross-sensitizing
effects of sexual experience on Amph reward as intra-NAc MK 801
injections during sexual experience prevented increased CPP for
Amph. Moreover, NAc NMDA receptor activation during sexual
behavior contributes to mating-induced cFos and deltaFosB
expression but not to sexual behavior or reward. In summary, NAc
NMDA receptor activation during sexual behavior regulates
mating-induced neural activation, neural alterations and behav-
ioral plasticity following sexual experience, but is not critical for the
initiation, expression, facilitation or reward of sex behavior.
Sexual experience causes cross-sensitization of CPP for Amph
after a period of abstinence from sexual reward (Pitchers et al.,
2010a, 2010b, 2013). Moreover, sexual experience upregulates
NR1 NMDA receptor subunit expression in the NAc (Pitchers et al.,
2012), suggesting that NMDA receptors are activated during sexual
experience and mediate these cross-sensitizing effects. Indeed, the
present study shows that activation of NMDA receptors in the NAc
during the acquisition of sexual experience is critical for
experience-induced cross-sensitization of Amph CPP. In previous
studies, NAc NMDA receptor blockade has been shown to block
Fig. 2. Intra-NAc MK 801 blocks sex experience-induced NAc deltaFosB expression.
(AeF), Representative images of NAc deltaFosB expression in sexually naïve and
experienced males that received intra-NAc saline (A,B), 0.5
m
lMK801(C,D)or1
m
lMK
801 (E,F). Scale bar indicates 100
m
m. Quantication of deltaFosB-positive neurons in
NAc core (G) and shell (H): saline: naïve, n ¼15; experienced, n ¼14; 0.5
m
lMK801:
naïve, n ¼7; experienced, or 1
m
l MK 801: naïve, n ¼7; exp, n ¼14. Data are presented
as mean ±SEM. * Indicates signicant difference compared to saline naïve (all
p<0.001).
Table 1
Sexual behavior parameters following intra-NAc MK 801. Latencies (in seconds) to
rst mount, rst intromission or ejaculation during the single mating session con-
ducted during the CPP conditioning trial (sal, n ¼6; 0.5
m
l MK, n ¼9; 1
m
l MK, n ¼7)
and single mating session conducted to investigate mating-induced cFos expression
(sal, n ¼5; 0.5
m
l MK, n ¼5; 1
m
l MK, n ¼5). Data represent mean ±SEM. No sig-
nicant differences between groups were detected.
Latencies (s)
Mount Intromission Ejaculation
CPP conditioning
Saline 593.3 ±291.6 599 ±289.5 961 ±167.3
MK 801 0.5
m
l 318.6 ±152.8 394.4 ±205.7 955 ±249.4
MK 801 1
m
l 231 ±55.1 296.1 ±73.6 852.6 ±135.5
Final mating
Saline 99.4 ±43.8 157.8 ±74.7 708.3 ±110.9
MK 801 0.5
m
l 115.8 ±35.6 126.4 ±31.6 975.5 ±166.8
MK 801 1
m
l 73.2 ±11.4 73.8 ±11.9 1467.5 ±652.6
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e164 159
morphine- (Popik and Kolasiewicz, 1999; Ma et al., 2006; Kao et al.,
2011; Xu et al., 2012) and ethanol-induced CPP (Gremel and
Cunningham, 2009) but the blockade took place during condi-
tioning or reconsolidation of the drug CPP. NMDA receptors in the
NAc have been shown to regulate the development of psychosti-
mulant locomotor sensitization (Wolf, 1998; Vanderschuren and
Kalivas, 2000). Specically, cocaine exposure leads to AMPA re-
ceptor current changes on NAc medium spiny neurons that are
dependent on NMDA receptor activation (Ungless et al., 2001), and
Amph behavioral sensitization is prevented by co-administration
(systemic) of MK 801 (Wolf and Jeziorski, 1993). Sex behavior also
results in locomotor sensitization by Amph (Pitchers et al., 2010a,
2012) and methamphetamine (Frohmader et al., 2011), but it is
Fig. 3. Intra-NAc MK 801 does not affect mating-induced CPP. A, Coronal NAc sections
indicating injection sites, represented unilaterally (dotted outline, on left: no mating on
last mating day, solid, on right: mating; white ll: saline; light gray: 0.5
m
l MK; dark gray:
1
m
l MK). The preference scores (percentage of time in the sex-paired chamber) (B) and
difference scores (time in paired eunpaired chamber) (C) during the pre-test and the
post-test within groups of animals that received intra-NAc saline (white bars,n¼6),
0.5
m
lMK801(gray bars,n¼9) or 1
m
lMK801(black bars,n¼10) prior to mating
during conditioning (preference scores: vehicle: p ¼0.048; difference scores: vehicle:
p¼0.038; 0.5
m
lMK801:p¼0.034; 1
m
lMK801:p¼0.033). Data are presented as
mean ±SEM. * Denotes signicant difference within group during post-test compared
to pre-test.
Fig. 4. Intra-NAc MK 801 blocks mating-induced NAc cFos expression. (AeD), Repre-
sentative images of NAc cFos-positive neurons from males that received intra-NAc
saline (B), 0.5
m
lMK801(C)or1
m
lMK801(D) prior to mating (þmating)or
handling (no mating)(A). Scale bar indicates 100
m
mac¼anterior commissure.
Quantitative data of cFos expression in NAc core (E) and shell (F) from males that
received intra-NAc saline (light gray bars,n¼5), 0.5
m
lMK801(dark gray bars,n¼6)
or 1
m
lMK801(black bars,n¼5) prior to mating or no mating (white bars,n¼12; 4
from each drug group). Data are presented as mean ±SEM. * Indicates signicant
difference compared to no mating group (core: 0.5
m
lMK801:p<0.001, 1
m
lMK801:
p<0.001; shell: 0.5
m
lMK801:p<0.0 01, 1
m
lMK801:p¼0.0 01); # shows signicant
difference compared to saline þmating group (p <0.001).
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e16 4160
unknown if NMDA receptor activation during sex experience is
required for this effect.
In addition, the current results demonstrate that sexual
experience-induced deltaFosB expression in the NAc is dependent
on NMDA receptor activation during mating. We have previously
shown that functional blockade of deltaFosB in the NAc, by over-
expression of the dominant-negative binding partner JunD, pre-
vented the cross-sensitization to Amph CPP following sexual
experience and abstinence (Pitchers et al., 2013). Hence, the effects
of NMDA receptor blockade on attenuation of Amph CPP sensiti-
zation may be mediated via blockade of deltaFosB expression
during the mating session. Drug- and natural reward-induced
deltaFosB expression has been shown to be expressed in D1 re-
ceptor neurons and is dependent on D1 receptor activation (Zhang
et al., 2002; Muller and Unterwald, 2005; Lobo et al., 2013). In
particular, our lab has shown that NAc D1 dopamine receptor
activation is required for sex experience-induced cross-sensitiza-
tion to Amph CPP (Pitchers et al., 2013). The current ndings show
that in addition to the role of dopamine receptor activation, NMDA
receptor activation is also functionally involved in sex experience-
induced deltaFosB upregulation in the NAc. Although there is
currently no direct evidence for glutamate release in the NAc dur-
ing mating, is it well established that dopamine is released in the
NAc during mating (Pfaus et al., 1990; Meisel et al., 1993; Fumero
et al., 1994) and that dopamine and glutamate are co-released in
mesoaccumbens axons (Tecuapetla et al., 2010; Zhang et al., 2015).
Together these ndings suggest a potential interaction between
NMDA and D1 receptor activation during reward experience. In
agreement, Amph locomotor and CPP sensitization is prevented by
NMDA gene deletion in D1-expressing medium spiny neurons
(Beutler et al., 2011), and D1 and NMDA receptors form direct in-
teractions in the striatum, and other areas, such as cortex and
hippocampus (Lee et al., 2002; Ladepeche et al., 2014). In the
striatum NMDA activity upregulates D1 receptor internalization
and trafcking (Scott et al., 2002, 20 06), and D1 activation increases
glutamate transmission (Harvey and Lacey, 1997) and NMDA Ca
2þ
signaling and ERK activation (Cahill et al., 2014).
A third nding of the current study was that mating-induced
cFos expression in the NAc is dependent on NMDA receptor acti-
vation. cFos is an immediate early gene commonly used as a marker
for neuronal activation and is upregulated in the NAc in responseto
drugs of abuse (Hope et al., 1992; Persico et al., 1993; Steiner and
Gerfen, 1993) and sexual behavior (Veening and Coolen, 2014).
We had previously shown that activation of NMDA receptors is
critical for mating-induced cFos in the medial preoptic area (mPOA)
(Dominguez et al., 2007). In this brain area, extra-synaptic levels of
glutamate are elevated during sexual behavior (Dominguez et al.,
2006, 2007); mating leads to phosphorylation of NMDA re-
ceptors; and intra-mPOA MK 801 blocks mating-induced phos-
phorylation of NMDA and cFos immunoreactivity (Dominguez
et al., 2007). cFos has been shown to regulate cocaine behavioral
sensitization and cocaine-induced deltaFosB in the striatum, spe-
cically in D1 receptor-expressing neurons (Zhang et al., 2006) but
the role of cFos induction in the NAc for the expression of sexual
behavior remains unknown. The current study suggests that sex-
induced cFos in the NAc regulates sex experience-induced delta-
FosB expression and cross-sensitization of Amph CPP.
Results of the present study demonstrate that activation of
NMDA receptors in the NAc is not required for initiation and
expression of sexual behavior, or sexual reward as determined by
CPP. This is in apparent contrast to the effects of excitotoxic lesions
of the NAc which disrupt initiation and expression of sexual
behavior in male rats, but not the preference for receptive females
(Kippin et al., 2004). In addition, the NAc is generally thought to be
critical for learning of stimulus-reward associations and mediating
goal-directed behavior (Robinson and Kolb, 2004; Richard et al.,
2013). Hence, our results indicate that NMDA receptor activation
in the NAc during mating is not required for these functions, similar
to the lack of involvement of NAc mGluR5 (Pitchers et al., 2015,
unpublished observations) and dopamine D1 receptors (Pitchers
et al., 2013). Likewise, intra-NAc injections of NMDA antagonists
block the acquisition of food-reinforced instrumental learning
(Kelley et al., 1997; Doerks et al., 2002; Kelley, 2004), but do not
block food intake (Reynolds and KC, 2003). Instead, a role for AMPA
receptor activation in motivation for food intake has been shown
(Reynolds and KC, 2003), suggesting that glutamate acts in the NAc
to mediate goal-directed learning and behavior, but via activation
of AMPA receptors. The present results also indicate that mating-
induced CPP was unaffected by intra-NAc MK 801 during the con-
ditioning day. NAc NMDA receptor antagonism has been shown to
block drug-induced CPP (Popik and Kolasiewicz, 1999; Ma et al.,
2006; Gremel and Cunningham, 2009; Kao et al., 2011; Xu et al.,
2012), without affecting social interaction- or food-induced CPP
(Ma et al., 2006). Together, these results suggests a role for NAc
NMDA receptors primarily in drug reward or cross-sensitization to
drug reward, but not of associated learning and CPP expression for
natural rewards, including sexual behavior.
Finally, the current results demonstratedthat repeated infusions
of MK 801 into the NAc caused a sensitization to Amph CPP in naïve
rats 10 days after nal infusion. It is well established that systemic
administration of MK 801 leads to Amph locomotor sensitization in
Fig. 5. Intra-NAc MK 801 does not affect mating-induced MPN cFos expression. (AeD)
Representative images of cFos-positive neurons in MPN from males that received intra-
NAc saline (B), 0.5
m
lMK801(C)or1
m
lMK801(D) prior to mating (þmating)or
handling (no mating)(A). 3V ¼third ventricle. Scale bar indicates 100
m
m. (E) Quan-
tication of MPN cFos-positive neurons in the same animals as in Fig. 4. Data are
presented as mean ±SEM. * Indicates signicant difference compared to no mating
group (all p <0.001).
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e164 161
rats (Rung et al., 2005; Eyjolfsson et al., 2006; Landa et al., 2014) but
its effects on cross-sensitization to drug CPP has not been
demonstrated. Therefore, the current ndings expand on this
knowledge by showing that MK 801 results in sensitization of
Amph reward, and specically by acting in the NAc. It is of note that
this effect of MK 801 in sexually naïve animals caused an opposite
effect to that in mating animals; i.e. a reward sensitization in
sexually naïve males instead of a blockade of cross-sensitization
following sexual experience. This may be explained by the ac-
tions of MK 801 in the absence of the endogenous ligand for NMDA
receptors. Indeed, studies suggest that MK 801's actions depend on
the activity state of NMDA receptors (Huettner and Bean, 1988;
Yuzaki et al., 1990; Reynolds and Miller, 1998). Furthermore, cell
culture studies have shown that MK 801 binding to the NMDA re-
ceptor is long lasting in the absence of glutamate or Mg
2þ
, while
dissociation of the drug from the receptor is greatly enhanced by
the presence of Mg
2þ
and opening of the channel (McKay et al.,
2013). Hence, the pharmacokinetics of MK 801 action may have
drastically differed in mating versus naïve animals.
5.1. Conclusions
In conclusion, the current study provides evidence for NAc
NMDA receptor regulation of mating-induced deltaFosB and cFos
and sexual experience-induced psychostimulant reward cross-
sensitization. Together, the results suggest that NAc NMDA recep-
tor activation during mating regulates the long-term effects of
natural reward experience via induction of cFos and deltaFosB.
References
Adams, E., Klug, J., Quast, M., Stairs, D.J., 2013. Effects of environmental enrichment
on nicotine-induced sensitization and cross-sensitization to
D
-Amphetamine in
rats. Drug Alcohol Depend. 129 (3), 247e253.
Aragona, B.J., Detwiler, J.M., Wang, Z., 2007. Amphetamine reward in the monog-
amous prairie vole. Neurosci. Lett. 418 (2), 190e194.
Bardo, M.T., Neisewander, J.L., Kelly, T.H., 2013. Individual differences and social
inuences on the neurobehavioral pharmacology of abused drugs. Pharmacol.
Rev. 65 (1), 255e290.
Beutler, L.R., Wanat, M.J., Quintana, A., Sanz, E., Bamford, N.S., Zweifel, L.S.,
Palmiter, R.D., 2011. Balanced NMDA receptor activity in dopamine D1 receptor
(D1R)- and D2R-expressing medium spiny neurons is required for amphet-
amine sensitization. PNAS 108 (10), 4206e4211.
Bradley, K.C., Meisel, R.L., 2001. Sexual behavior induction of c-Fos in the nucleus
accumbens and Amphetamine-stimulated locomotor activity are sensitized by
previous sexual experience in female Syrian hamsters. J. Neurosci. 21 (6),
2123e2130 .
Britt, J.P., Benaliouad, F., McDevitt, R.A., Stuber, G.D., Wise, R.A., Bonci, A., 2012.
Synaptic and behavioral prole of multiple glutamatergic inputs to the nucleus
accumbens. Neuron 76 (4), 790e803.
Burkett, J.P., Young, L.J., 2012. The behavioral, anatomical and pharmacological
parallels between social attachment, love and addiction. Psychopharmacology
(Berl) 224 (1), 1e26.
Cahill, E., Pascoli, V., Trilieff, P., Savoldi, D., Kappes, V., Luscher, C., Caboche, J.,
Vanhoutte, P., 2014. D1R/GluN1 complexes in the striatum integrate dopamine
and glutamate signalling to control synaptic plasticity and cocaine-induced
responses. Mol. Psychiatry 19 (12), 1295e1304.
Chen, B.T., Hopf, F.W., Bonci, A., 2010. Synaptic plasticity in the mesolimbic system:
therapeutic implications for substance abuse. Ann. N. Y. Acad. Sci. 1187,
129e139.
Christiansen, A.M., Dekloet, A.D., Ulrich-Lai, Y.M., Herman, J.P., 2011. Snacking
causes long term attenuation of HPA axis stress responses and enhancement of
brain FosB/deltaFosB expression in rats. Physiol. Behav. 103 (1), 111e116 .
Davis, J.F., Loos, M., Di Sebastiano, A.R., Brown, J.L., Lehman, M.N., Coolen, L.M., 2010.
Lesions of the medial prefrontal cortex cause maladaptive sexual behavior in
male rats. Biol. Psychiatry 67 (12), 1199e1204.
Di Sebastiano, A.R., Wilson-Perez, H.E., Lehman, M.N., Coolen, L.M., 2011. Lesions of
orexin neurons block conditioned place preference for sexual behavior in male
rats. Horm. Behav. 59 (1), 1e8.
Di Sebastiano, A.R., Yong-Yow, S., Wagner, L., Lehman, M.N., Coolen, L.M., 2010.
Orexin mediates initiation of sexual behavior in sexually naive male rats, but is
not critical for sexual performance. Horm. Behav. 58 (3), 397e404.
Doerks, T., Copley, R.R., Schultz, J., Ponting, C.P., Bork, P., 2002. Systematic identi-
cation of novel protein domain families associated with nuclear functions.
Genome Res. 12 (1), 47e56.
Dominguez, J.M., Balfour, M.E., Lee, H.S., Brown, J.L., Davis, B.A., Coolen, L.M., 2007.
Mating activates NMDA receptors in the medial preoptic area of male rats.
Behav. Neurosci. 121 (5), 1023e1031.
Dominguez, J.M., Gil, M., Hull, E.M., 2006. Preoptic glutamate facilitates male sexual
behavior. J. Neurosci. 26 (6), 1699e170 3 .
Eyjolfsson, E.M., Brenner, E., Kondziella, D., Sonnewald, U., 2006. Repeated injection
of MK801: an animal model of schizophrenia? Neurochem. Int. 48 (6e7),
541e546.
Feltenstein, M.W., See, R.E., 2013. Systems level neuroplasticity in drug addiction.
Cold Spring Harb. Perspect. Med. 3 (5), a011916.
Frohmader, K.S., Lehman, M.N., Laviolette, S.R., Coolen, L.M., 2011. Concurrent
exposure to methamphetamine and sexual behavior enhances subsequent drug
reward and causes compulsive sexual behavior in male rats. J. Neurosci. 31 (45),
1647 3e16482.
Frohmader, K.S., Pitchers, K.K., Balfour, M.E., Coolen, L.M., 2010. Mixing pleasures:
review of the effects of drugs on sex behavior in humans and animal models.
Horm. Behav. 58 (1), 149e162.
Fumero, B., Fernandez-Vera, J.R., Gonzalez-Mora, J.L., Mas, M., 1994. Changes in
monoamine turnover in forebrain areas associated with masculine sexual
behavior: a microdialysis study. Brain Res. 662 (1e2), 233e239.
Gipson, C.D., Beckmann, J.S., El-Maraghi, S., Marusich, J.A., Bardo, M.T., 2011. Effect of
environmental enrichment on escalation of cocaine self-administration in rats.
Psychopharmacology (Berl) 214 (2), 557e566.
Gipson, C.D., Kupchik, Y.M., Kalivas, P.W., 2014. Rapid, transient synaptic plasticity
in addiction. Neuropharmacology 76 Pt B, 276e286.
Greenwood, B.N., Foley, T.E., Le, T.V., Strong, P.V., Loughridge, A.B., Day, H.E.,
Fleshner, M., 2011. Long-term voluntary wheel running is rewarding and pro-
duces plasticity in the mesolimbic reward pathway. Behav. Brain Res. 217 (2),
354e362.
Gremel, C.M., Cunningham, C.L., 2009. Involvement of amygdala dopamine and
nucleus accumbens NMDA receptors in ethanol-seeking behavior in mice.
Neuropsychopharmacology 34 (6), 1443e1453.
Grueter, B.A., Robison, A.J., Neve, R.L., Nestler, E.J., Malenka, R.C., 2013. FosB differ-
entially modulates nucleus accumbens direct and indirect pathway function.
Proc. Natl. Acad. Sci. U. S. A. 110 (5), 1923e1928.
Grueter, B.A., Rothwell, P.E., Malenka, R.C., 2012. Integrating synaptic plasticity and
striatal circuit function in addiction. Curr. Opin. Neurobiol. 22 (3), 545e551.
Harvey, J., Lacey, M.G., 1997. A postsynaptic interaction between dopamine d1 and
NMDA receptors promotes presynaptic inhibition in the rat nucleus accumbens
via adenosine release. J. Neurosci. 17 (14), 5271e5280.
Hollen, K.M., Nakabeppu, Y., Davies, S.W., 1997. Changes in expression of deltaFosB
and the Fos family proteins following NMDA receptor activation in the rat
striatum. Brain Res. Mol. Brain Res. 47 (1e2), 31e43.
Hope, B., Kosofsky, B., Hyman, S.E., Nestler, E.J., 1992. Regulation of immediate early
gene expression and AP-1 binding in the rat nucleus accumbens by chronic
cocaine. Proc. Natl. Acad. Sci. U. S. A. 89, 5764e5768.
Huettner, J.E., Bean, B.P., 1988. Block of N-methyl-
D
-aspartate-activated current by
the anticonvulsant MK-801: selective binding to open channels. Proc. Natl.
Acad. Sci. U. S. A. 85, 1307e1311.
Hunt, M., Falinska, M., Kasicki, S., 2010. Local injection of MK801 modies oscilla-
tory activity in the nucleus accumbens in awake rats. Psychopharmacology 24
(6), 931e941.
Hussain, N., Flumerfelt, B.A., Rajakumar, N., 2001. Glutamatergic regulation of
haloperidol-induced c-fos expression in the rat striatum and nucleus accum-
bens. Neuroscience 102 (2), 391e399.
Kalivas, P.W., Volkow, N.D., 2011. New medications for drug addiction hiding in
glutamatergic neuroplasticity. Mol. Psychiatry 16 (10), 974e986.
Kao, J.H., Huang, E.Y., Tao, P.L., 2011. NR2B subunit of NMDA receptor at nucleus
accumbens is involved in morphine rewarding effect by siRNA study. Drug
Alcohol Depend. 118 (2e3), 366e374.
Kawasaki, Y., Ishida, S., Jin, C., Kitamura, Y., Kawasaki, H., Gomita, Y., Sendo, T.,
Araki, H., 2011. Effect of glutamate receptor antagonists microinjected into the
nucleus accumbens on place aversion induced by naloxone in single-dose,
morphine-treated rats. Eur. J. Pharmacol. 666 (1e3), 131e13 4.
Kelley, A.E., 2004. Ventral striatal control of appetitive motivation: role in ingestive
behavior and reward-related learning. Neurosci. Biobehav. Rev. 27 (8), 765e776.
Kelley, A.E., Smith-Roe, S.L., Holahan, M.R., 1997. Response-reinforcement learning
is dependent on N-methyl-
D
-aspartate receptor activation in the nucleus
accumbens core. Proc. Natl. Acad. Sci. U. S. A. 94, 12174e12179.
Kelz, M., Chen, J., Carlezon, W.A., Whisler, K., Gilden, L., Beckmann, A.M., Steffen, C.,
Zhang, Y.-J., Marotti, L., Self, D.W., Tkatch, T., Baranauskas, G., Surmeier, D.J.,
Neve, R.L., Duman, R.S., Picciotto, M.R., EJ, N., 1999. Expression of the tran-
scription factor deltaFosB in the brain controls sensitivity to cocaine. Nat. Lett.
401, 272e276.
Kim, Y., Teylan, M.A., Baron, M., Sands, A., Nairn, A.C., Greengard, P., 20 09. Meth-
ylphenidate-induced dendritic spine formation and DeltaFosB expression in
nucleus accumbens. Proc. Natl. Acad. Sci. U. S. A. 106 (8), 2915e2920.
Kippin, T.E., Sotiropoulos, V., Badih, J., Pfaus, J.G., 2004. Opposing roles of the nu-
cleus accumbens and anterior lateral hypothalamic area in the control of sexual
behaviour in the male rat. Eur. J. Neurosci. 19, 698e704.
Koob, G.F., Volkow, N.D., 2010. Neurocircuitry of addiction. Neuro-
psychopharmacology 35 (1), 217e238.
Ladepeche, L., Dupuis, J.P., Groc, L., 2014. Surface trafcking of NMDA receptors:
gathering from a partner to another. Semin. Cell Dev. Biol. 27, 3e13.
Landa, L., Machalova, A., Sulcova, A., 2014. Implication of NMDA receptors in
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e16 4162
behavioural sensitization to psychostimulants: a short review. Eur. J. Pharmacol.
730, 77e81.
Lee, F.J.S., Xue, S., Pei, L., Vukusic, B., Chery, N., Wang, Y., Wang, Y.T., Niznik, H.B.,
Yu, X.-M., Liu, F., 2002. Dual regulation of NMDA receptor functions by direct
protein-protein interactions with the dopamine D1 receptor. Cell 111, 219e230.
Lee, K.W., Kim, Y., Kim, A.M., Helmin, K., Nairn, A.C., Greengard, P., 2006. Cocaine-
induced dendritic spine formation in D1 and D2 dopamine receptor-containing
medium spiny neurons in nucleus accumbens. Proc. Natl. Acad. Sci. U. S. A. 103
(9), 3399e3404.
Leonibus, E., Mele, A., Oliverio, A., Pert, A., 2002. Distinct pattern of c-FOS mRNA
expression after systemic and intra-accumbens amphetamine and mk-801.
Neuroscience 115 (1), 67e78.
Liu, J., Nickolenko, J., Sharp, F.R., 1994. Morphine induces c-fos and junB in striatum
and nucleus accumbens via D1 and N-methyl-
D
-aspartate receptors. Proc. Natl.
Acad. Sci. U. S. A. 91, 8537e8541.
Liu, Y., Young, K.A., Curtis, J.T., Aragona, B.J., Wang, Z., 2011. Social bonding decreases
the rewarding properties of amphetamine through a dopamine D1 receptor-
mediated mechanism. J. Neurosci. 31 (22), 7960e7966.
Lobo, M.K., Zaman, S., Damez-Werno, D.M., Koo, J.W., Bagot, R.C., DiNieri, J.A.,
Nugent, A., Finkel, E., Chaudhury, D., Chandra, R., Riberio, E., Rabkin, J.,
Mouzon, E., Cachope, R., Cheer, J.F., Han, M.H., Dietz, D.M., Self, D.W., Hurd, Y.L.,
Vialou, V., Nestler, E.J., 2013. DeltaFosB induction in striatal medium spiny
neuron subtypes in response to chronic pharmacological, emotional, and
optogenetic stimuli. J. Neurosci. 33 (47), 18381e18395.
Loweth, J.A., Tseng, K.Y., Wolf, M.E., 2014. Adaptations in AMPA receptor trans-
mission in the nucleus accumbens contributing to incubation of cocaine
craving. Neuropharmacology 76 Pt B, 287e300.
Lu, L., Shepard, J.D., Scott Hall, F., Shaham, Y., 2003. Effect of environmental stressors
on opiate and psychostimulant reinforcement, reinstatement and discrimina-
tion in rats: a review. Neurosci. Biobehav. Rev. 27 (5), 457e491.
Ma, Y.Y., Guo, C.Y., Yu, P., Lee, D.Y., Han, J.S., Cui, C.L., 2006. The role of NR2B con-
taining NMDA receptor in place preference conditioned with morphine and
natural reinforcers in rats. Exp. Neurol. 200 (2), 343e355.
Mameli, M., Luscher, C., 2011. Synaptic plasticity and addiction: learning mecha-
nisms gone awry. Neuropharmacology 61 (7), 1052e1059.
McKay, S., Bengtson, C.P., Bading, H., Wyllie, D.J., Hardingham, G.E., 2013. Recovery
of NMDA receptor currents from MK-801 blockade is accelerated by Mg
2þ
and
memantine under conditions of agonist exposure. Neuropharmacology 74,
119 e125.
Meisel, R., Mullins, A., 2006. Sexual experience in female rodents: cellular mecha-
nisms and functional consequences. Brain Res. 1126 (1), 56e65.
Meisel, R.L., Camp, D.M., Robinson, T.E., 1993. A microdialysis study of ventral
striatal dopamine during sexual behavior in female Syrian hamsters. Behav.
Brain Res. 55 (2), 151e15 7.
Muller, D.L., Unterwald, E.M., 2005. D1 dopamine receptors modulate deltaFosB
induction in rat striatum after intermittent morphine administration.
J. Pharmacol. Exp. Ther. 314 (1), 148e154 .
Nader, J., Chauvet, C., Rawas, R.E., Favot, L., Jaber, M., Thiriet, N., Solinas, M., 2012.
Loss of environmental enrichment increases vulnerability to cocaine addiction.
Neuropsychopharmacology 37 (7), 1579e1587.
Neisewander, J.L., Peartree, N.A., Pentkowski, N.S., 2012. Emotional valence and
context of social inuences on drug abuse-related behavior in animal models of
social stress and prosocial interaction. Psychopharmacology (Berl) 224 (1),
33e56.
Nestler, E.J., 2008. Review. Transcriptional mechanisms of addiction: role of Del-
taFosB. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363 (1507), 3245e3255.
Olsen, C.M., 2011. Natural rewards, neuroplasticity, and non-drug addictions.
Neuropharmacology 61 (7), 1109e112 2 .
Papp, E., Borhegyi, Z., Tomioka, R., Rockland, K.S., Mody, I., Freund, T.F., 2012. Glu-
tamatergic input from specic sources inuences the nucleus accumbens-
ventral pallidum information ow. Brain Struct. Funct. 217 (1), 37e48.
Perrotti, L.I., Hadeishi, Y., Ulery, P.G., Barrot, M., Monteggia, L., Duman, R.S.,
Nestler, E.J., 2004. Induction of deltaFosB in reward-related brain structures
after chronic stress. J. Neurosci. 24 (47), 10594e10602.
Perrotti, L.I., Weaver, R.R., Robison, B., Renthal, W., Maze, I., Yazdani, S., Elmore, R.G.,
Knapp, D.J., Selley, D.E., Martin, B.R., Sim-Selley, L., Bachtell, R.K., Self, D.W.,
Nestler, E.J., 2008. Distinct patterns of DeltaFosB induction in brain by drugs of
abuse. Synapse 62 (5), 358e369.
Persico, A.M., Schindler, C.W., O'Hara, B.F., Brannock, M.T., Uhl, G.R., 1993. Brain
transcription factor expression: effects of acute and chronic amphetamine and
injection stress. Brain Res. Mol. Brain Res. 20 (1e2), 91e10 0 .
Pfaus, J.G., Damsma, G., Nomikos, G.G., Wenkstern, D.G., Blaha, C.D., Phillips, A.G.,
Fibiger, H.C., 1990. Sexual behavior enhances central dopamine transmission in
the male rat. Brain Res. 530 (2), 345e348.
Pitchers, K.K., Balfour, M.E., Lehman, M.N., Richtand, N.M., Yu, L., Coolen, L.M.,
2010a. Neuroplasticity in the mesolimbic system induced by natural reward and
subsequent reward abstinence. Biol. Psychiatry 67 (9), 872e879.
Pitchers, K.K., Coppens, C.M., Beloate, L.N., Fuller, J., Van, S., Frohmader, K.S.,
Laviolette, S.R., Lehman, M.N., Coolen, L.M., 2014. Endogenous opioid-induced
neuroplasticity of dopaminergic neurons in the ventral tegmental area in-
uences natural and opiate reward. J. Neurosci. 34 (26), 8825e8836.
Pitchers, K.K., Frohmader, K.S., Vialou, V., Mouzon, E., Nestler, E.J., Lehman, M.N.,
Coolen, L.M., 2010b. DeltaFosB in the nucleus accumbens is critical for rein-
forcing effects of sexual reward. Genes Brain Behav. 9 (7), 831e840.
Pitchers, K.K., Schmid, S., Di Sebastiano, A.R., Wang, X., Laviolette, S.R.,
Lehman, M.N., Coolen, L.M., 2012. Natural reward experience alters AMPA and
NMDA receptor distribution and function in the nucleus accumbens. PLoS One 7
(4), e34700.
Pitchers, K.K., Vialou, V., Nestler, E.J., Laviolette, S.R., Lehman, M.N., Coolen, L.M.,
2013. Natural and drug rewards act on common neural plasticity mechanisms
with DeltaFosB as a key mediator. J. Neurosci. 33 (8), 3434e3442.
Pitchers, K.K., DiSebastiano, A.R., Coolen, L.M., 2015. mGluR5 activation in the nu-
cleus accumbens is not essential for sexual behavior, but reduced mGluR5 ac-
tivity may contribute to the cross sensitization of amphetamine responses by
sexual experience. Submitted to Neuropharmacology [unpublished
observations].
Pomierny-Chamiolo, L., Rup, K., Pomierny, B., Niedzielska, E., Kalivas, P.W., Filip, M.,
2014. Metabotropic glutamatergic receptors and their ligands in drug addiction.
Pharmacol. Ther. 142 (3), 281e305.
Popik, P., Kolasiewicz, W., 1999. Mesolimbic NMDA receptors are implicated in the
expression of conditioned morphine reward. Naunyn Schmiedebergs Arch.
Pharmacol. 359 (4), 288e294.
Puhl, M.D., Blum, J.S., Acosta-Torres, S., Grigson, P.S., 2012. Environmental enrich-
ment protects against the acquisition of cocaine self-administration in adult
male rats, but does not eliminate avoidance of a drug-associated saccharin cue.
Behav. Pharmacol. 23 (1), 43e53.
Raz, S., Berger, B.D., 2010. Social isolation increases morphine intake: behavioral
and psychopharmacological aspects. Behav. Pharmacol. 21 (1), 39e46.
Reynolds, I.J., Miller, R.J., 1998. Multiple sites for the regulation of the N-methyl-
D
-
aspartate receptor. Mol. Pharmacol. 33, 581e584.
Reynolds, S.M., KC, B., 2003. Glutamate motivational ensembles in nucleus
accumbens: rostrocaudal shell gradients of fear and feeding. Eur. J. Neurosci.17,
2187e2200.
Richard, J.M., Castro, D.C., Difeliceantonio, A.G., Robinson, M.J., Berridge, K.C., 2013.
Mapping brain circuits of reward and motivation: in the footsteps of Ann Kelley.
Neurosci. Biobehav. Rev. 37 (9 Pt A), 1919e1931.
Robinson, T.E., Kolb, B., 2004. Structural plasticity associated with exposure to drugs
of abuse. Neuropharmacology 47 (Suppl. 1), 33e46.
Robison, A.J., Nestler, E.J., 2011. Transcriptional and epigenetic mechanisms of
addiction. Nat. Rev. Neurosci. 12 (11), 623e637.
Robison, A.J., Vialou, V., Mazei-Robison, M., Feng, J., Kourrich, S., Collins, M., Wee, S.,
Koob, G., Turecki, G., Neve, R., Thomas, M., Nestler, E.J., 2013. Behavioral and
structural responses to chronic cocaine require a feedforward loop involving
DeltaFosB and calcium/calmodulin-dependent protein kinase II in the nucleus
accumbens shell. J. Neurosci. 33 (10), 4295e4307.
Rung, J.P., Carlsson, A., Ryden Markinhuhta, K., Carlsson, M.L., 2005. (þ)-MK-801
induced social withdrawal in rats; a model for negative symptoms of schizo-
phrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 29 (5), 827e832.
Scott, L., Kruse, M.S., Forssberg, H., Brismar, H., Greengard, P., Aperia, A., 20 02. Se-
lective up-regulation of dopamine D1 receptors in dendritic spines by NMDA
receptor activation. Proc. Natl. Acad. Sci. U. S. A. 99 (3), 1661e16 64 .
Scott, L., Zelenin, S., Malmersjo, S., Kowalewski, J.M., Markus, E.Z., Nairn, A.C.,
Greengard, P., Brismar, H., Aperia, A., 2006. Allosteric changes of the NMDA
receptor trap diffusible dopamine 1 receptors in spines. Proc. Natl. Acad. Sci. U.
S. A. 103 (3), 762e767.
Solinas, M., Chauvet, C., Thiriet, N., El Rawas, R., Jaber, M., 2008. Reversal of cocaine
addiction by environmental enrichment. Proc. Natl. Acad. Sci. U. S. A. 105 (44),
1714 5 e1715 0 .
Staffend, N.A., Hedges, V.L., Chemel, B.R., Watts, V.J., Meisel, R.L., 2014. Cell-type
specic increases in female hamster nucleus accumbens spine density
following female sexual experience. Brain Struct. Funct. 219 (6), 2071e2081.
Steiner, H., Gerfen, C.R., 1993. Cocaine-induced c-fos messenger RNA is inversely
related to dynorphin expression in striatum. J. Neurosci. 13 (12), 506 6e5081.
Tecuapetla, F., Patel, J.C., Xenias, H., English, D., Tadros, I., Shah, F., Berlin, J.,
Deisseroth, K., Rice, M.E., Tepper, J.M., Koos, T., 2010. Glutamatergic signaling by
mesolimbic dopamine neurons in the nucleus accumbens. J. Neurosci. 30 (20),
7105e7110.
Teegarden, S.L., Scott, A.N., Bale, T.L., 2009. Early life exposure to a high fat diet
promotes long-term changes in dietary preferences and central reward
signaling. Neuroscience 162 (4), 924e932.
Tenk, C.M., Wilson, H., Zhang, Q., Pitchers, K.K., Coolen, L.M., 2009. Sexual reward in
male rats: effects of sexual experience on conditioned place preferences asso-
ciated with ejaculation and intromissions. Horm. Behav 55 (1), 93e97.
Tye, K.M., 2012. Glutamate inputs to the nucleus accumbens: does source matter?
Neuron 76 (4), 671e673.
Ungless, M.A., Whistler, J.L., Malenka, R.C., Bonci, A., 2001. Single cocaine exposure
in vivo induces long-term potentiation in dopamine neurons. Nat. Lett. 411,
583e587.
van Huijstee, A.N., Mansvelder, H.D., 2014. Glutamatergic synaptic plasticity in the
mesocorticolimbic system in addiction. Front. Cell. Neurosci. 8, 466.
Vanderschuren, L.J.M.J., Kalivas, P., 2000. Alterations in dopaminergic and gluta-
matergic transmission in the induction and expression of behavioral sensiti-
zation: a critical review of preclinical studies. Psychopharmacology 151,
99e120.
Veening, J.G., Coolen, L.M., 2014. Neural mechanisms of sexual behavior in the male
rat: emphasis on ejaculation-related circuits. Pharmacol. Biochem. Behav. 121,
170 e183 .
Volkow, N.D., Wang, G.J., Fowler, J.S., Tomasi, D., Telang, F., 2011. Addiction: beyond
dopamine reward circuitry. Proc. Natl. Acad. Sci. U. S. A. 108 (37), 15037e15042.
Volman, S.F., Lammel, S., Margolis, E.B., Kim, Y., Richard, J.M., Roitman, M.F.,
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e164 163
Lobo, M.K., 2013. New insights into the specicity and plasticity of reward and
aversion encoding in the mesolimbic system. J. Neurosci. 33 (45), 17569e1757 6 .
Wallace, D.L., Vialou, V., Rios, L., Carle-Florence, T.L., Chakravarty, S., Kumar, A.,
Graham, D.L., Green, T.A., Kirk, A., Iniguez, S.D., Perrotti, L.I., Barrot, M.,
DiLeone, R.J., Nestler, E.J., Bolanos-Guzman, C.A., 2008. The inuence of Delta-
FosB in the nucleus accumbens on natural reward-related behavior. J. Neurosci.
28 (41), 10272e1027 7.
Wang, H.L., Xiang, X.H., Guo, Y., Wu, W.R., Cao, D.Y., Wang, H.S., Zhao, Y., 2005.
Ionotropic glutamatergic neurotransmission in the ventral tegmental area
modulates DeltaFosB expression in the nucleus accumbens and abstinence
syndrome in morphine withdrawal rats. Eur. J. Pharmacol. 527 (1e3), 94e104.
Werme, M., Messer, C., Olson, L., Gilden, L., Thoren, P., Nester, E., Brene, S., 20 02.
DeltaFosB regulates wheel running. J. Neurosci. 22 (18), 8133e8138.
Wolf, M.E., 1998. The role of excitatory amino acids in behavioral sensitization to
psychomotor stimulants. Prog. Neurobiol. 54, 679e720.
Wolf, M.E., Jeziorski, M., 1993. Coadministration of MK-801 with amphetamine,
cocaine or morphine prevents rather than transiently masks the development
of behavioral sensitization. Brain Res. 613 (2), 291e294.
Xu, Y., Lu, X.F., Cui, C.L., Ge, F.F., Li, Y.J., Zhang, H.L., 2012. Essential role of NR2B-
containing NMDA receptor-ERK pathway in nucleus accumbens shell in
morphine-associated contextual memory. Brain Res. Bull. 89 (1e2), 22e30.
Yanahashi, S., Hashimoto, K., Hattori, K., Yuasa, S., Iyo, M., 2004. Role of NMDA
receptor subtypes in the induction of catalepsy and increase in Fos protein
expression after administration of haloperidol. Brain Res. 1011 (1), 84e93.
Yuzaki, M., Miyawaki, A., Akita, K., Kudo, Y., Ogura, A., Ino, H., Mikoshiba, K., 1990.
Mode of blockade by MK-801 of N-methyl-
D
-aspartate-induced increase in
intracellular Ca
2þ
in cultured mouse hippocampal neurons. Brain Res. 517
(1e2), 51e56.
Zhang, D., Zhang, L., Lou, D.W., Nakabeppu, Y., Zhang, J., Xu, M., 2002. The dopamine
D1 receptor is a critical mediator for cocaine-induced gene expression.
J. Neurochem. 82, 1453e1564.
Zhang, J., Zhang, L., Jiao, H., Zhang, Q., Zhang, D., Lou, D., Katz, J.L., Xu, M., 2006. c-Fos
facilitates the acquisition and extinction of cocaine-induced persistent changes.
J. Neurosci. 26 (51), 13287e13296.
Zhang, S., Qi, J., Li, X., Wang, H.L., Britt, J.P., Hoffman, A.F., Bonci, A., Lupica, C.R.,
Morales, M., 2015. Dopaminergic and glutamatergic microdomains in a subset
of rodent mesoaccumbens axons. Nat. Neurosci. 18 (3), 386e392.
L.N. Beloate et al. / Neuropharmacology 101 (2016) 154e16 4164
... Sexual experience in male rodents also leads to transient increases in dendritic arborization and number of spines in NAc Pitchers et al., 2013), decreases in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/N-methyl-Daspartate (AMPA/NMDA) ratios in the NAc (Pitchers et al., 2012), changes in NMDA (GluN) and AMPA (GluA) receptors in NAc (Pitchers et al., 2012), and a reduction in soma size of VTA dopamine neurons (Beloate, Omrani, Adan, Webb, & Coolen, 2016;Pitchers et al., 2014). Furthermore, both VTA dopamine activity and NAc dopamine and glutamate receptor activation during sexual experience regulates sex-induced changes in drugrelated behaviors and neural alterations (Beloate, Omrani, et al., 2016;Beloate, Weems, Casey, Webb, & Coolen, 2016;Pitchers et al., 2013). ...
... Males will typically reduce the numbers of mounts prior to ejaculation and thus increase copulation efficiency, and latencies to ejaculation are reduced (Beloate, Omrani, et al., 2016;Pitchers et al., 2014Pitchers et al., , 2012. This effect of sexual experience on decreased latency to mount, intromission and ejaculation, i.e., facilitation of sexual behavior, is evident even after only one mating experience, but typically requires several sessions to reach reliable detection of statistically significant differences (Beloate, Omrani, et al., 2016;Beloate, Weems, et al., 2016;Pitchers et al., 2014Pitchers et al., , 2012Pitchers et al., , 2013Pitchers, Di Sebastiano, & Coolen, 2016;Pitchers, Frohmader, et al., 2010). Finally, experience-induced facilitation of sexual behavior is expressed in a long-term manner, as it is maintained for at least a month after the last mating session (Pitchers et al., 2014(Pitchers et al., , 2012). ...
... Chemogenetic inhibition of VTA dopamine signaling blocks mating-induced cFos, in the NAc and mPFC, but not the BLA, suggesting that dopamine activation and release in specific brain areas contributes to mating-induced cFos expression (Beloate, Omrani, et al., 2016). In addition, mating-induced cFos in the NAc is dependent on GluN activation, as it is prevented by NAc GluN antagonism (Beloate, Weems, et al., 2016). Infusion of the GluN antagonist, MK-801, in the NAc does not affect initiation or expression of sexual behavior (Beloate, Weems, et al., 2016). ...
Chapter
Many factors, including social elements, influence drug addiction in humans and can be modeled in laboratory rodents. In general, the presence of social reward is protective against drug abuse and the absence or removal of social reward in both humans and rodents increases vulnerability to drug addiction. The current review chapter is focused on studies from our lab that have examined the effects of sociosexual behavior in male rats on drug-induced behaviors, including changes in both psychostimulant and opiate behavior. Furthermore, we review the underlying neural mechanisms by which these effects occur. Together, these results may help elucidate the neural mechanisms underlying the interaction between social and drug rewards and the mechanisms by which a loss of social rewards increase the vulnerability to drug addiction development.
... Therefore, we hypothesized that sex-and sexual abstinenceinduced changes in NAc glutamate receptor expression could regulate the subsequent effects on cross-sensitization to psychostimulant CPP. Indeed, NAc NMDA receptor activation during sexual experience is important for sexual experience-induced neuroplasticity and crosssensitization to psychostimulant preference, as intra-NAc NMDA antagonism prior to each 4 days of sexual behavior prevents experienceinduced Amph CPP (Beloate et al., 2016c). Furthermore, intra-NAc NMDA antagonism blocks mating-induced neural activation in the NAc, as measured by cFos immunoreactivity, but does not block sexual behavior or CPP (Beloate et al., 2016c). ...
... Indeed, NAc NMDA receptor activation during sexual experience is important for sexual experience-induced neuroplasticity and crosssensitization to psychostimulant preference, as intra-NAc NMDA antagonism prior to each 4 days of sexual behavior prevents experienceinduced Amph CPP (Beloate et al., 2016c). Furthermore, intra-NAc NMDA antagonism blocks mating-induced neural activation in the NAc, as measured by cFos immunoreactivity, but does not block sexual behavior or CPP (Beloate et al., 2016c). However, the functional role of other glutamate receptor changes throughout the abstinence period and the source of glutamate acting in the NAc during sexual activity remain unknown. ...
... DeltaFos expression is dependent on dopamine activity in the VTA during sexual behavior, as chemogenetic inactivation of VTA dopamine prevents deltaFosB expression in the NAc core and shell and mPFC (Beloate et al., 2016b). Furthermore, mating-induced deltaFosB expression in the NAc is regulated by both NAc D1 dopamine and NMDA receptors (Pitchers et al., 2010b(Pitchers et al., , 2013Beloate et al., 2016c). Along with the behavioral effects, these results suggest that neuroplasticity in the dopaminergic D1 pathway from the VTA to the NAc, in interaction with NMDA receptors signaling, plays an important role in regulating sexual experience-induced cross-sensitization to Amph CPP (Beloate et al., 2016b). ...
Article
Different factors influence the development of drug addiction in humans, including social reward experiences. In animals, experience with social rewards, such as sexual behavior, pair bonding, social and environmental enrichment, can be protective. However, loss or lack of social rewards can lead to a vulnerability to drug-seeking behavior. The effects of social reward experience on drug-seeking behavior are associated with changes in the neural pathways that control drug-related behavior. This review will provide an introduction and overview of the mesolimbic pathway and the influence of social reward experience on drug-seeking behavior in rodents. Moreover, the research from our laboratory on effects of sexual experience and loss of sex reward on psychostimulant and opiate reward will be reviewed. Finally, we will review current knowledge of the neural mechanisms that underlie these interactions. Investigations of the neural underpinnings by which social and drug rewards interact contribute to improved understanding of the neural basis of vulnerability for drug addiction and reward-related behaviors in general.
... In another study, Beloate et al. (2016b) showed that high but not low volume of MK-801 (noncompetitive NMDAR antagonist) reversed mating-experience-induced sensitization of D-amphetamine CPP, and both volumes reversed the effect of mating experience on NAc d -FosB expression. MK-801 had no effect on sexual behavior and mating-induced CPP. ...
... Still unknown is the mechanistic connection between decreased NAc mGluR5 expression and sensitization of D-amphetamine CPP. Beloate et al. (2016b), using TH-Cre rats and inhibitory DREADDs, showed that dopamine neurons in VTA are critical for mating-experience-induced sensitization of D-amphetamine CPP, and that inhibition of these neurons reversed or decreased the effect of mating experience on d -FosB expression in NAc and mPFC, and VTA dopamine neurons' soma size. Finally, Pitchers et al. (2014) reported that, unlike D-amphetamine CPP, morphine CPP showed tolerance after mating experience when acquisition of morphine CPP occurred 1 d after the last mating session. ...
Article
Until recently, most modern neuroscience research on addiction using animal models did not incorporate manipulations of social factors. Social factors play a critical role in human addiction: social isolation and exclusion can promote drug use and relapse, while social connections and inclusion tend to be protective. Here, we discuss the state of the literature on social factors in animal models of opioid and psychostimulant preference, self-administration, and relapse. We first summarize results from rodent studies on behavioral, pharmacological, and circuit mechanisms of the protective effect of traditional experimenter-controlled social interaction procedures on opioid and psychostimulant conditioned place preference, self-administration, and relapse. Next, we summarize behavioral and brain-mechanism results from studies using newer operant social-interaction procedures that inhibit opioid and psychostimulant self-administration and relapse. We conclude by discussing how the reviewed studies point to future directions for the addiction field and other neuroscience and psychiatric fields, and their implications for mechanistic understanding of addiction and development of new treatments. SIGNIFICANCE STATEMENT In this review, we propose that incorporating social factors into modern neuroscience research on addiction could improve mechanistic accounts of addiction and help close gaps in translating discovery to treatment. We first summarize rodent studies on behavioral, pharmacological, and circuit mechanisms of the protective effect of both traditional experimenter-controlled and newer operant social-interaction procedures. We then discuss potential future directions and clinical implications.
... Administration of MK801, either systemically or via microinjections into the brain (mPOA and PVN), diminished male rat sexual behavior (Fleming and Kucera, 1991;Powell et al., 2003;Melis et al., 2004;Dominguez et al., 2007;Vigdorchik et al., 2012;Beloate et al., 2016). In Table 1, we summarized the above studies, as well as subsequent studies reviewed. ...
... MK801 administration (0.6 µg) to the preoptic-anterior hypothalamus in male Wistar rats reduced 50-kHz calls (Brudzynski and Pniak, 2002). In contrast to these studies, infusions of 1 or 2 µg of MK801 to the NAc for 4 consecutive days did not change sexual behavior in male Sprague-Dawley rats (Beloate et al., 2016). The different brain regions targeted may explain this discrepancy between these studies. ...
Article
Full-text available
The survival of animal species predicates on the success of sexual reproduction. Neurotransmitters play an integral role in the expression of these sexual behaviors in the brain. Here, we review the role of glutamate in sexual behavior in rodents and non-rodent species for both males and females. These encompass the release of glutamate and correlations with glutamate receptor expression during sexual behavior. We then present the effects of glutamate on sexual behavior, as well as the effects of antagonists and agonists on different glutamate transporters and receptors. Following that, we discuss the potential role of glutamate on steroid-independent sexual behavior. Finally, we demonstrate the interaction of glutamate with other neurotransmitters to impact sexual behavior. These sexual behavior studies are crucial in the development of novel treatments of sexual dysfunction and in furthering our understanding of the complexity of sexual diversity. In the past decade, we have witnessed the burgeoning of novel techniques to study and manipulate neuron activity, to decode molecular events at the single-cell level, and to analyze behavioral data. They pose exciting avenues to gain further insight into future sexual behavior research. Taken together, this work conveys the essential role of glutamate in sexual behavior.
... The changes in spine morphology from the thin to the mushroom type are related to the expression of ΔFosB. It has been demonstrated that ΔFosB is regulated by D1 dopamine and NMDA receptors [12,18,58]. In addition, some studies have shown that ΔFosB induction inside the mPFC and NAcc appears to be an element of the restructuring of neural circuits that underlies long-term behavioral adaptations with sexual experience [37]. ...
Article
Sexual experience improves copulatory performance in male rats. Copulatory performance has been associated with dendritic spines density in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAcc), structures involved in the processing of sexual stimuli and the manifestation of sexual behavior. Dendritic spines modulate excitatory synaptic contacts, and their morphology is associated with the ability to learn from experience. This study was designed to determine the effect of sexual experience on the density of different types or shapes of dendritic spines in the mPFC and NAcc of male rats. A total of 16 male rats were used, half of them were sexually experienced while the other half were sexually inexperienced. After three sessions of sexual interaction to ejaculation, the sexually-experienced males presented shorter mount, intromission, and ejaculation latencies. Those rats presented a higher total dendritic density in the mPFC, and a higher numerical density of thin, mushroom, stubby, and wide spines. Sexual experience also increased the numerical density of mushroom spines in the NAcc. In both the mPFC and NAcc of the sexually experienced rats, there was a lower proportional density of thin spines and a higher proportional density of mushroom spines. Results show that the improvement in copulatory efficiency resulting from prior sexual experience in male rats is associated with changes in the proportional density of thin and mushroom dendritic spines in the mPFC and NAcc. This could represent the consolidation of afferent synaptic information in these brain regions, derived from the stimulus-sexual reward association.
... A number of neurochemical systems are also implicated in experience-dependent plasticity in social behavior, including oxytocin (Gil et al., 2011), glutamate (e.g., Beloate et al., 2016;Powell et al., 2003), and opioids (reviewed in Pfaus et al., 2012). However, catecholamines like dopamine (DA) and norepinephrine (NE) have been most extensively studied. ...
Article
Revealing the mechanisms underlying experience-dependent plasticity is a hallmark of behavioral neuroscience. While the study of social behavior has focused primarily on the neuroendocrine and neural control of social behaviors, the plasticity of these innate behaviors has received relatively less attention. Here, we review studies on mating-dependent changes to social behavior and neural circuitry across mammals, birds, and reptiles. We provide an overview of species similarities and differences in the effects of mating experiences on motivational and performative aspects of sexual behaviors, on sensory processing and preferences, and on the experience-dependent consolidation of sexual behavior. We also discuss recent insights into the neural mechanisms of and developmental influences on mating-dependent changes and outline promising approaches to investigate evolutionary parallels and divergences in experience-dependent plasticity. RESEARCH HIGHLIGHTS • Social experiences alter the performance and hormonal and neural control of social behavior • Nonmammalian vertebrates can demonstrate similar types of experience-dependent changes as mammals • Similarities and differences in brain mechanisms of behavioral plasticity between mammals and nonmammalian vertebrates are discussed
... Treating tertiary drug-induced chronic symptomology is short-sighted at best when compared to the consideration of newer epidemiological targets for long-term prophylaxis linked to early interventions. For example, Delta FosB (ΔFosB), a gene transcription factor, is a central mediator in the epigenetic development of all drug and non-drug addictions [24][25][26][27]. Based on an intensive literature review, ΔFosB modulates compulsivity during the acquisition of addiction [28,29]. ...
Article
The consensus of the current literature strongly supports the concept that brain neurotransmitters, and second messengers involved in the net release of dopamine in the mesolimbic region, especially the Nucleus Accumbens (NAc), is directly linked to motivation, anti-stress, incentive salience (wanting), and well-being. The role of dopamine in terms of alcohol withdrawal symptomology, cocaine craving behavior, dopamine -condensation products (TIQs), and more recently, the genetic aspects of drug-seeking and pro-dopamine regulation, provide compelling evidence of the relevant molecular neurological correlates of dopaminergic /endorphinergic mechanisms in reward circuitry due to genetic polymorphisms and epigenetic insults. In the face of an Americans opioid epidemic, the clinical consensus is to treat Opioid Use Disorder (OUD) with life-long opioid substitution therapy. However, the authors suggest a paradigm shift involving novel modalities like targeting the endorphinergic system linked to dopamine release at the NAc, in terms of the induction of required "dopamine homeostasis." Utilizing the known genetic - environmental interaction theorem P = G +E, the authors provide a clear rationale for the adoption of genetic risk testing coupled with endorphinergic/dopamine regulation to address dysfunction across the brain reward circuitry. The goal of altering resting-state, functional connectivity may require a gentle "neurotransmitter fix" vis enkephalinase inhibition to overcome or combat - self-induction of acute dopamine release via psychoactive substance misuse resulting in chronic dopamine down-regulation. As subsets of reward deficiency, we are poised to provide novel, genetically guided therapy for endorphinergic, opioidergic, and dopaminergic deficiencies and related syndromes, utilizing "Precision Addiction Management.
... Moreover, we have demonstrated that D1R blockage in the NAcc during the conditioning phase reduced the olfactory preference of males toward female chemosignals more than 10 days later. The reinforcing effects of sexual stimuli are mediated by long-lasting neuroplasticity changes in the NAc (Beloate et al., 2016;Pitchers et al., 2012;Staffend et al., 2014), which are dependent on D1R signaling and its down-stream effects (Pitchers et al., 2013;Staffend et al., 2014). Our results suggest that blocking D1R signaling during the initial interactions with female chemosignals prevented the occurrence of female-induced neuroplasticity in the NAcc, and this altered the olfactory preference of males days later. ...
Article
Full-text available
Sexual preference for the opposite sex is a fundamental behavior underlying reproductive success, but the neural mechanisms remain unclear. Here, we examined the role of dopamine signaling in the nucleus accumbens core (NAcc) in governing chemosensory-mediated preference for females in TrpC2-/- and wild-type male mice. TrpC2-/- males, deficient in VNO-mediated signaling, do not display mating or olfactory preference toward females. We found that, during social interaction with females, TrpC2-/- males do not show increased NAcc dopamine levels, observed in wild-type males. Optogenetic stimulation of VTA-NAcc dopaminergic neurons in TrpC2-/- males during exposure to a female promoted preference response to female pheromones and elevated copulatory behavior toward females. Additionally, we found that signaling through the D1 receptor in the NAcc is necessary for the olfactory preference for female-soiled bedding. Our study establishes a critical role for the mesolimbic dopaminergic system in governing pheromone-mediated responses and mate choice in male mice.
Article
The role of experience in the process of behavioral refinement has been undertheorized by philosophers of neuroscience and neuroscientists. By examining sleep studies in behavioral neurobiology, I show that scientists frequently invoke a variety of lived experiences—what I call experientially derived notions—to refine the behavior under investigation. Of note, these behaviors must remain sufficiently fuzzy throughout experimentation to permit refinement. The aim of this article is to recognize that neuroscientists’ use of lived experience necessarily helps refine behaviors and render those behavioral terms relevant to human life.
Article
Full-text available
Vulnerability to develop addiction is influenced by numerous factors, including social behavior. Specifically, in human users, drug taking in a socio-sexual context appears to enhance further drug-seeking behavior. Users report heightened sexual pleasure as a motivation for further drug use and display risk behaviors even when tested in drug-free state. Here, using a preclinical model of limited voluntary drug use in rats, the hypothesis was tested that methamphetamine (Meth)-taking concurrently with socio-sexual experience increases vulnerability to addiction. Male Sprague Dawley rats were socially housed and underwent limited-access Meth self-administration (maximum 1 mg/kg/session). Meth-taking was either concurrent or non-concurrent with sexual behavior: concurrent animals were mated with a receptive female immediately after each session, while non-concurrent animals gained equivalent sexual experience the week prior. Next, drug-seeking behaviors were measured during cue reactivity, extinction, and reinstatement sessions using different extinction and reinstatement protocols in 4 separate studies. Both groups equally acquired Meth self-administration and did not differ in total Meth intake. However, drug-seeking behavior was significantly higher in concurrent animals during cue reactivity tasks, extinction sessions, and cue- or Meth-induced reinstatement tests. In addition, sexual behavior in the absence of Meth triggered reinstatement of drug-seeking in concurrent animals. These results indicate that Meth-taking in a socio-sexual context significantly enhances vulnerability for drug addiction in male rats. This preclinical paradigm of drug self-administration concurrent with socio-sexual behavior provides a useful model for studying the underlying neurobiology of socially driven vulnerability to drug addiction.
Article
Full-text available
Mesoaccumbens fibers are thought to co-release dopamine and glutamate. However, the mechanism is unclear, and co-release by mesoaccumbens fibers has not been documented. Using electron microcopy, we found that some mesoaccumbens fibers have vesicular transporters for dopamine (VMAT2) in axon segments that are continuous with axon terminals that lack VMAT2, but contain vesicular glutamate transporters type 2 (VGluT2). In vivo overexpression of VMAT2 did not change the segregation of the two vesicular types, suggesting the existence of highly regulated mechanisms for maintaining this segregation. The mesoaccumbens axon terminals containing VGluT2 vesicles make asymmetric synapses, commonly associated with excitatory signaling. Using optogenetics, we found that dopamine and glutamate were released from the same mesoaccumbens fibers. These findings reveal a complex type of signaling by mesoaccumbens fibers in which dopamine and glutamate can be released from the same axons, but are not normally released at the same site or from the same synaptic vesicles.
Article
Full-text available
Addictive drugs remodel the brain's reward circuitry, the mesocorticolimbic dopamine (DA) system, by inducing widespread adaptations of glutamatergic synapses. This drug-induced synaptic plasticity is thought to contribute to both the development and the persistence of addiction. This review highlights the synaptic modifications that are induced by in vivo exposure to addictive drugs and describes how these drug-induced synaptic changes may contribute to the different components of addictive behavior, such as compulsive drug use despite negative consequences and relapse. Initially, exposure to an addictive drug induces synaptic changes in the ventral tegmental area (VTA). This drug-induced synaptic potentiation in the VTA subsequently triggers synaptic changes in downstream areas of the mesocorticolimbic system, such as the nucleus accumbens (NAc) and the prefrontal cortex (PFC), with further drug exposure. These glutamatergic synaptic alterations are then thought to mediate many of the behavioral symptoms that characterize addiction. The later stages of glutamatergic synaptic plasticity in the NAc and in particular in the PFC play a role in maintaining addiction and drive relapse to drug-taking induced by drug-associated cues. Remodeling of PFC glutamatergic circuits can persist into adulthood, causing a lasting vulnerability to relapse. We will discuss how these neurobiological changes produced by drugs of abuse may provide novel targets for potential treatment strategies for addiction.
Article
Full-text available
Convergent dopamine and glutamate signalling onto the extracellular signal-regulated kinase (ERK) pathway in medium spiny neurons (MSNs) of the striatum controls psychostimulant-initiated adaptive processes underlying long-lasting behavioural changes. We hypothesised that the physical proximity of dopamine D1 (D1R) and glutamate NMDA (NMDAR) receptors, achieved through the formation of D1R/NMDAR complexes, may act as a molecular bridge that controls the synergistic action of dopamine and glutamate on striatal plasticity and behavioural responses to drugs of abuse. We found that concomitant stimulation of D1R and NMDAR drove complex formation between endogenous D1R and the GluN1 subunit of NMDAR. Conversely, preventing D1R/GluN1 association with a cell-permeable peptide (TAT-GluN1C1) left individual D1R and NMDAR-dependent signalling intact, but prevented D1R-mediated facilitation of NMDAR-calcium influx and subsequent ERK activation. Electrophysiological recordings in striatal slices from mice revealed that D1R/GluN1 complexes control the D1R-dependent enhancement of NMDAR currents and long-term potentiation in D1R-MSN. Finally, intra-striatal delivery of TAT-GluN1C1 did not affect acute responses to cocaine but reduced behavioural sensitization. Our findings uncover D1R/GluN1 complexes as a major substrate for the dopamine-glutamate interaction in MSN that is usurped by addictive drugs to elicit persistent behavioural alterations. They also identify D1R/GluN1 complexes as molecular targets with a therapeutic potential for the vast spectrum of psychiatric diseases associated with an imbalance between dopamine and glutamate transmission.Molecular Psychiatry advance online publication, 29 July 2014; doi:10.1038/mp.2014.73.
Article
Full-text available
Natural reward and drugs of abuse converge on the mesolimbic pathway and activate common mechanism of neural plasticity in the nucleus accumbens. Chronic exposure to opiates induces plasticity in dopaminergic neurons of the ventral tegmental area (VTA), which regulates morphine reward tolerance. Here, we test the hypotheses that mating-induced release of endogenous opioids in the VTA causes morphological changes of VTA dopamine cells in male rats, which in-turn regulate the long-term expression of experience-induced reinforcement of sexual behavior. First, sexual experience decreased VTA dopamine soma size 1 and 7 days, but not 30 days after the last mating session. This effect was blocked with naloxone before each mating session; thus, VTA dopamine cell plasticity was dependent on action of endogenous opioids. In turn, VTA plasticity was associated with altered opiate reward, as sexually experienced males did not form conditioned place preference for 0.5 mg/kg morphine. Next, it was determined whether endogenous opioid action mediates sexual reward and memory in male rats treated with naloxone during mating experience, either systemically or intra-VTA. Naloxone did not prevent the initial experience-induced facilitation of sexual behavior over repeated mating sessions, or conditioned place preference for mating. However, naloxone treatment attenuated the longer-term expression of experience-induced facilitation of sexual behavior and neural activation in mesolimbic areas induced by mating-associated conditioned cues. Together, these data demonstrate that endogenous opioids during mating induce neural plasticity in VTA dopamine neurons that appear critical for morphine reward and long-term memory for natural reward behavior.
Article
Natural rewards and psychostimulants cause similar neural plasticity in the nucleus accumbens (NAc). In particular, sexual experience in male rats causes increased locomotor activity and conditioned place preference (CPP) induced by d-Amphetamine (amph). The latter is dependent on a period of abstinence from sexual reward. In this study, the role of mGluR5 activation in the NAc for expression of mating and the cross-sensitizing effects of sexual experience was tested. First, intra-NAc infusions of mGluR5 antagonists MPEP (1 or 10 μg/μL) or MTEP (1 μg/μL) 15 minutes prior to mating during 4 daily sessions had no effect on male rat sexual behavior. Subsequently, these sexually experienced males were tested for amph-induced locomotor activity and CPP after one week of abstinence from sexual reward. In addition, sexually naïve males that received MPEP, MTEP or vehicle infusions prior to 4 daily handling sessions were included. Cross-sensitization of locomotion or CPP was not prevented by NAc mGluR5 antagonism during acquisition of sexual experience. Instead, sexually naive animals that received NAc mGluR5 antagonists without mating demonstrated sensitized amph-induced locomotor responses and enhanced CPP on par with sexually experienced males. Finally, we showed that sexual experience caused prolonged down-regulation of mGluR5 protein in the NAc, dependent on abstinence from sexual behavior. Together, these findings suggest that mGluR5 activation in the NAc is not essential for the expression of mating, but that experience-induced reduction in mGluR5 protein may contribute to the cross-sensitization of amph responses by sexual experience and abstinence.
Article
Sexual behavior of the male rat can be described as a 'sequence': a series of behavioral transitions eventually leading to a consummatory act: ejaculation. A 'funnel-model' is presented to describe the behavioral progression during the sexual sequence. The ejaculation itself is extensively controlled by the 'spinal ejaculation generator', consisting of several elements with afferent sources of genitosensory information, with ascending projection fibers to inform the brainstem and forebrain as well as with descending afferent fibers providing the supraspinal control mechanisms with the opportunity to restrict ejaculations to the optimal moments and circumstances. The messages ascending from the spinal cord reach several interconnected thalamic, hypothalamic and limbic brain areas and are 'mixed' with olfactory information. These brain areas play a role in mechanisms like 'sexual satiety' or a temporary interruption of sexual activities ('post-ejaculatoryinterval'), but the exact facilitatory and inhibitory mechanisms involved have not been elucidated yet. In the 'downward' mechanisms controlling the spinal 'release' of an ejaculation, the medial preoptic nucleus plays an important role in cooperation with a number of brainstem areas. This nucleus is also explicitly involved in the rewarding experiences coming with an ejaculation. Finally, the role of several neurotransmitters and - peptides on male sexual behavior are discussed shortly, because sometimes they show remarkable effects on specific aspects of the behavioral sequence. We conclude that, despite our increased knowledge about the brain mechanisms involved in the control of ejaculation, we are still far away from a complete understanding and quite a few questions remain to be resolved.