ArticlePDF Available

Amylin Receptor Signaling in the Nucleus Accumbens Negatively Modulates μ-opioid-Driven Feeding

Authors:

Abstract and Figures

Amylin is a peptide co-secreted with insulin that penetrates into the brain, and produces satiation-like effects via actions in the brainstem, hypothalamus, and mesencephalon. Little is known, however, about the effects of amylin in the nucleus accumbens shell (AcbSh), where a circumscribed zone of intense amylin receptor (AMY-R) binding overlaps reported mappings of a 'hotspot' for μ-opioid receptor (μ-OR) amplification of food reward. Here, the ability of intra-AcbSh AMY-R signaling to modulate μ-OR-driven feeding was explored. Amylin (1-30 ng) was administered with the μ-OR agonist, DAMGO (0.25 μg), directly into the AcbSh of ad-libitum-maintained rats. Amylin dose-dependently reversed DAMGO-induced hyperphagia; 3-ng of amylin reduced DAMGO-mediated feeding by nearly 50%. This dose was, however, completely ineffective at altering DAMGO-induced feeding in the anterior dorsal striatum (ADS). Intra-AcbSh amylin alone (3-30 ng) modestly suppressed 10% sucrose intake in ad-libitum-maintained rats, and chow in food-deprived rats, but only at the 30-ng dose. This result indicates that reversal of AcbSh DAMGO-induced feeding at a 10-fold lower dose was due neither to malaise nor motoric impairment. Finally, intra-AcbSh infusion of the AMY-R antagonist, AC187 (20 μg), significantly attenuated the ability of pre-feeding to suppress DAMGO-induced food intake, with no effects in non-prefed rats. Hence, AMY-R signaling negatively modulates μ-OR-mediated appetitive responses at the level of the AcbSh. The results with AC187 indicate that endogenous AMY-R transmission in the AcbSh curtails opioid function in the post-prandial period, suggesting a novel pathway for peripheral-central integration in the control of appetitive motivation and opioid reward.Neuropsychopharmacology accepted article preview online, 24 June 2014; doi:10.1038/npp.2014.153.
Content may be subject to copyright.
Amylin Receptor Signaling in the Nucleus Accumbens
Negatively Modulates l-opioid-Driven Feeding
Sarah K Baisley
1
and Brian A Baldo*
,1,2
1
Neuroscience Training Program, University of Wisconsin-Madison, Medical Sciences Center, University Ave Madison, WI, USA;
2
Department of
Psychiatry University of Wisconsin-Madison, Research Park Blvd, Madison, WI, USA
Amylin is a peptide co-secreted with insulin that penetrates into the brain, and produces satiation-like effects via actions in the brainstem,
hypothalamus, and mesencephalon. Little is known, however, about the effects of amylin in the nucleus accumbens shell (AcbSh), where
a circumscribed zone of intense amylin receptor (AMY-R) binding overlaps reported mappings of a ‘hotspot’ for m-opioid receptor
(m-OR) amplification of food reward. Here, the ability of intra-AcbSh AMY-R signaling to modulate m-OR-driven feeding was explored.
Amylin (1–30 ng) was administered with the m-OR agonist, D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) (0.25 mg), directly into the
AcbSh of ad libitum-maintained rats. Amylin dose-dependently reversed DAMGO-induced hyperphagia; 3 ng of amylin reduced
DAMGO-mediated feeding by nearly 50%. This dose was, however, completely ineffective at altering DAMGO-induced feeding in the
anterior dorsal striatum. Intra-AcbSh amylin alone (3–30 ng) modestly suppressed 10% sucrose intake in ad libitum-maintained rats, and
chow in food-deprived rats, but only at the 30-ng dose. This result indicates that reversal of AcbSh DAMGO-induced feeding at a 10-fold
lower dose was neither due to malaise nor motoric impairment. Finally, intra-AcbSh infusion of the AMY-R antagonist, AC187 (20 mg),
significantly attenuated the ability of prefeeding to suppress DAMGO-induced food intake, with no effects in non-prefed rats. Hence,
AMY-R signaling negatively modulates m-OR-mediated appetitive responses at the level of the AcbSh. The results with AC187 indicate
that endogenous AMY-R transmission in the AcbSh curtails opioid function in the postprandial period, suggesting a novel pathway for
peripheral-central integration in the control of appetitive motivation and opioid reward.
Neuropsychopharmacology (2014) 39, 3009–3017; doi:10.1038/npp.2014.153; published online 13 August 2014
INTRODUCTION
Amylin, a 37-amino-acid peptide that belongs to the calci-
tonin gene-related peptide (CGRP) family (van Rossum et al,
1997), is co-secreted with insulin from pancreatic
beta cells in coordination with prandial stimuli (Butler
et al, 1990; Moore and Cooper, 1991; Ahren and Sundler,
1992). Once secreted, amylin modulates insulin’s effects
on glycogen synthesis and glucose uptake in muscle, and
therefore has an important role in glycemic control (Singh-
Franco et al, 2011). In addition to these metabolic effects,
amylin also modulates food intake via actions at multiple
levels of the central nervous system (CNS). Amylin penetrates
into the brain at least as well as insulin, and accumulates in
sites throughout the neural axis (Banks and Kastin, 1998).
Because CNS amylin receptors (AMY-Rs) show regional
differences and localization to discrete neural pathways and
structures, it is hypothesized that amylin and related peptides
have a role in neuroregulation (Beaumont et al, 1993; Sexton
et al, 1994; van Rossum et al, 1994; Christopoulos et al, 1995).
Accordingly, AMY-R ligands cause a satiation-like suppres-
sion of feeding when infused into the lateral ventricle, third
ventricle, hypothalamus, and ventral tegmental area (VTA)
(Chance et al, 1991; Morley and Flood, 1991; Bouali et al,
1995; Lutz et al,1998a;Rushinget al, 2000; Mietlicki-Baase
et al, 2013). Perhaps the most extensively studied site for
feeding-modulatory actions of amylin is the area postrema;
blockade of area postrema AMY-Rs and lesions specific to
the area postrema both attenuate the anorectic effect of
systemically administered amylin (Lutz et al, 1998b, 2001;
Mollet et al, 2004).
Less is known about feeding-modulatory effects of
amylin in the telencephalon, despite the fact that one of
the densest concentrations of high-affinity amylin-binding
sites, and expression of component genes encoding the
high-affinity AMY-R (Poyner et al, 2002) is found in the
medial nucleus accumbens shell (AcbSh) (Sexton et al, 1994;
van Rossum et al, 1994; Baisley et al, 2014). This zone of
intense AMY-R binding conforms remarkably well with the
circumscribed medial AcbSh area from which intense
feeding responses are elicited by GABA or m-opioid receptor
(m-OR) stimulation (Bakshi and Kelley, 1993; Stratford
and Kelley, 1997; Zhang and Kelley, 2000). Moreover,
the reported ‘hotspot’ for amplification of hedonic taste
reactions by m-OR stimulation (Pecina and Berridge, 2005)
*Correspondence: Dr BA Baldo, Department of Psychiatry, University
of Wisconsin-Madison, School of Medicine and Public Health, 6001
Research Park Blvd, Madison, WI 53719 USA. Tel: +1 608 263 4019,
Fax: +1 608 265 3050, E-mail: babaldo@wisc.edu
Received 20 March 2014; revised 16 June 2014; accepted 17 June
2014; accepted article preview online 24 June 2014
Neuropsychopharmacology (2014) 39, 3009 –3017
&
2014 American College of Neuropsychopharmacology. All rights reserved 0893-133X/14
www.neuropsychopharmacology.org
overlaps the AMY-R distribution. Because of this overlap,
AcbSh-localized AMY-Rs are well-positioned to modulate
food intake and hedonic taste reward by interacting with the
m-opioid system.
To date, only one study (Baldo and Kelley, 2001) has
investigated the role of AcbSh-localized AMY-Rs in control-
ling feeding behavior; this study showed that exogenously
administered amylin in the 30–100 ng range suppressed
feeding. Nevertheless, the interaction of AMY-Rs with other
Acb-localized neuromodulator systems, and, importantly,
the role of endogenous Acb AMY-R signaling in modulating
feeding behavior, remain unknown. Here, interactions
between AMY-Rs and m-ORs were studied, both in the
AcbSh where dense AMY-R binding is found, and the
anterior dorsal striatum (ADS), lacking high-affinity AMY-
R binding but where m-ORs also modulate feeding (Bakshi
and Kelley, 1993; DiFeliceantonio et al, 2012). We also
examined the effects of AMY-R blockade on m-OR-driven
feeding, during either a food-deprived state or immediately
after a prefeeding session (when circulating amylin levels
are high) (Alam et al, 1992; Arnelo et al, 1998), to explore
whether an endogenous ‘tone’ of AMY-R signaling at the
level of the AcbSh interacts with the behavioral functions
of m-ORs.
MATERIALS AND METHODS
Subjects
Subjects in all experiments were male Sprague-Dawley rats,
obtained from Harlan (Madison, WI), weighing 300–325 g
upon arrival at the laboratory. The rats were pair-housed
in clear polycarbonate cages (9.5-inch width 17-inch
length 8-inch height), with cob bedding, in a light- and
temperature-controlled vivarium. Animals were maintained
under a 12 : 12-h light–dark cycle (lights on at 7 : 00 AM).
Food and water were available ad libitum, except as
indicated for the various experiments. Animals were
handled daily to reduce stress. Testing occurred between
1200–1800 h. All facilities and procedures were in accor-
dance with the guidelines regarding animal use and care
put forth by the National Institutes of Health, and were
supervised and approved by the Institutional Animal
Care and Use Committee of the University of Wisconsin.
Surgical Procedure
Rats (weighing 300–325 g) underwent stereotaxic surgery
under isoflurane anesthesia according to standard techni-
ques (Perry et al, 2009). Bilateral stainless-steel cannulae
(10-mm long, 23 gauge) were aimed bilaterally either
at the AcbSh or at the ADS and anchored in place with
dental acrylic (New Truliner, Skokie, IL) and skull screws
(Plastics One, Roanoke, VA). Surgeries were conducted with
the nosebar set to 3.3 mm below interaural zero. For the
AcbSh, the coordinates from bregma were: þ3.2-mm
anteroposterior (AP); ±1.0-mm lateromedial (LM); and
5.2-mm dorsoventral (DV) (with injectors extending an
additional 2.5 mm beyond cannulae tips for a final DV
coordinate of 7.7). For ADS surgeries, the coordinates
from bregma were: þ1.6 mm AP; ±2.4 mm LM; and
1.7 mm DV (with injectors extending an additional
2.5 mm beyond cannulae tips for a final DV coordinate
of 4.2). Wire stylets (10-mm long, 30 gauge) were placed
in the cannulae to prevent blockage. Animals were given an
intramuscular injection of penicillin (0.3 ml of a 300 000 U/
ml suspension; Phoenix Pharmaceuticals, St Joseph, MO),
placed in a warm recovery cage, returned to their home
cages on awakening, and given a recovery period of no o5
days (with daily health checks) before behavioral testing
commenced.
Drugs and Microinfusions
Amylin (Bachem, Torrance, CA) and ([D-Ala2, N-MePhe4,
Gly-ol]-enkephalin) (DAMGO) (Bachem) were dissolved in
sterile isotonic saline, whereas AC187 (Tocris Bioscience,
Ellisville, MO) was dissolved in sterile H
2
O. The 0.25 mg/
0.5 ml/side dose of DAMGO was chosen because it has been
shown to elicit robust feeding in satiated rats (eg, Perry
et al, 2009). The dose of the AMY-R antagonist AC187 (Hay
et al, 2005) was chosen because in our laboratory it altered
prepulse inhibition upon intra-AcbSh infusion (Baisley et al,
2014). In other literature, an AC187 dose of 30 mg but not
10 mg delivered into lower levels of the CNS increased food
intake in rats (Lutz et al, 1997; Mollet et al, 2004). Hence, the
present experiment used an AC187 dose of 20 mg/side. All
three drugs were infused directly into specific brain regions
in accordance with the experimental designs. For micro-
infusions, injectors (connected via tubing to a microdrive
pump) extended 2.5 mm past cannulae tips, and delivered
drugs at 0.32 ml/min over 1 min 33 s, with a 1-min post-
infusion period before reinsertion of stylets and placement
of rats into testing chambers.
Experimental Design
In all experiments, 30-min free-feeding test sessions were
conducted in wire-bottom polycarbonate cages with rat
chow pellets and water available, as previously described
(Baldo and Kelley, 2001). Prior to all the experiments, rats
were habituated to the testing cages to minimize stress.
Testing occurred on every other day.
Interactions between Amylin and DAMGO in the AcbSh
Effects of amylin were tested in two dose ranges (0, 1, 3 ng
and 0, 10, 30 ng), in separate groups of rats (n¼7 for the
low-dose range; n¼10 for the high-dose range) to minimize
the number of infusions per experiment. For both experi-
ments, rats were surgically implanted with cannulae aimed
at AcbSh. Rats were infused with saline or 0.25 mg/0.5 ml
DAMGO into the AcbSh 10 min before testing. Five minutes
before testing, rats received a second infusion of saline or
amylin into the AcbSh. DAMGO dose and amylin dose were
both within-subjects factors; hence, each rat received all
amylin doses with and without DAMGO (for a total of six
infusions). Treatments were counterbalanced across test
days according to Latin-square designs.
Interactions between Amylin and DAMGO in the ADS
The same basic protocol as Experiment 1 was followed,
except rats (n¼6) were implanted with cannulae aimed at
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3010
Neuropsychopharmacology
the ADS instead of the AcbSh. ADS coordinates were chosen
based on prior literature describing a m-opioid-sensitive
zone dorsal to the Acb (Bakshi and Kelley, 1993a;
DiFeliceantonio et al, 2012). Rats received a total of four
infusions.
Effects of Intra-AcbSh Amylin (Without DAMGO) on
Palatability- and Hunger-Induced Feeding
Eight rats were used to test the effects of intra-AcbSh
amylin upon sucrose intake in ad libitum-maintained rats.
Prior to experimentation, the rats were exposed daily for
2 days to a 10% sucrose solution until they consumed
a consistent amount over two 30-minute test sessions
spaced 4 h apart. Once stable intake was achieved, rats were
given intra-AcbSh infusion of amylin (0, 3, 10, 30 ng/0.5 ml/
side) administered according to a Latin-square design.
Amylin was infused 5 min before testing, whereupon rats
were placed into the testing cages for 30 min with free
access to chow, water, and a bottle containing 10% sucrose.
Seven of the eight rats in this experiment also underwent
testing for intra-AcbSh amylin effects on hunger-driven
chow intake. In this part of the experiment, rats were food-
deprived for 18 h prior to each testing period, given intra-
AcbSh amylin (0, 3, 10, 30 ng/0.5 ml) infusions, and placed
into the testing cages for 30 min with rat chow and water
present. The two experiments (sucrose intake and hunger-
driven chow intake) were performed in a counterbalanced
order, with half the rats receiving sucrose first, and the
other half, hunger/chow intake first (for a total of eight
infusions).
Effects of AC187 on DAMGO-Induced Feeding, With or
Without Prefeeding
Seven rats were surgically prepared with cannulae aimed at
the AcbSh. After recovery, rats underwent behavioral testing
every other day for a total of eight test days. All rats were
food-deprived for 18 h before each testing day; however, on
each interim testing-free day, they had free access to food.
On each testing day, rats were either given a 30-min
‘prefeeding’ session, or given no prefeeding session, where-
upon they received intra-AcbSh infusions of DAMGO and
AC187. DAMGO (0, 0.25 mg/0.5 ml) was infused bilaterally
into the AcbSh 10 min before testing, followed 5 min later
by an infusion of AC187 (0, 20 mg/0.5 ml). For rats exposed
to the prefeeding session, infusions were given after the
prefeeding session, and the time between the prefeeding and
testing session was 15 min. The environmental contexts for
prefed and non-prefed rats were identical. Following drug
infusions, both prefed and non-prefed rats were tested in a
30-min feeding sessions. Each rat received all treatment
combinations (mock/mock, DAMGO/mock, mock/AC187,
DAMGO/AC187) under both prefed and non-prefed condi-
tions, according to a within-subjects, Latin-square design.
‘Mock’ infusions were used to limit the number of needle
insertions into the tissue of the AcbSh and therefore reduce
tissue damage. Ten-millimeter injectors that did not
protrude beyond the ends of the guide cannulae were
lowered into the cannulae for 2 min and 33 s to mimic a
saline infusion. Not including mock infusions, each rat
received a total of eight infusions.
Data Analysis
Multifactor ANOVAs were used for all experiments.
Significant main effects or interactions were followed by
Fisher’s PLSD post hoc. Experimenters blind to data and
treatments confirmed injector placements in Nissl-stained
sections. Final sample sizes exclude rats with placements
outside of target sites.
RESULTS
Figure 1 depicts histological verification of intra-tissue
injection placements. One rat was removed from Experi-
ment 1 owing to placements that fell outside of the targeted
region. Representative photomicrographs of injector place-
ments into the AcbSh and ADS of cannulated animals reveal
that cannulae and injector tracks are clearly visible with no
unusual damage to the targeted areas. For Acb placements,
although in some cases we would notice some damage to the
lateral ventricles induced by the guide cannulae, injector
tips were found always to be located within the cellular
neuropil of the AcbSh (not in the ventricles).
Amylin Potently Reduced Intra-AcbSh DAMGO-Induced
Feeding
As shown in Figure 2, DAMGO significantly elevated
feeding in both the low-dose and high-dose DAMGO/
amylin interaction studies (main effect of DAMGO:
F(1, 6) ¼50.7, Po0.001 for low-dose study; F(1, 9) ¼17.9,
Po0.01 for high-dose study). Post hoc comparison among
means with Fisher’s PLSD test confirmed that DAMGO-
associated levels of food intake were significantly elevated
relative to saline or to any of the amylin-alone doses
(Ps¼0.0001–0.05).
In both dose ranges tested, amylin significantly attenu-
ated DAMGO-induced hyperphagia (DAMGO amylin
interactions: F(2, 12) ¼4.8, Po0.05 for low-dose study;
F(2, 18) ¼6.6, Po0.01 for high-dose study). Post hoc
comparison among means revealed specific differences
between DAMGO/saline and DAMGO/amylin-3 ng, DAM-
GO/amylin-10 ng, and DAMGO/amylin-30 ng dose-combi-
nations (Figure 2a and b). Note that these doses of amylin
did not suppress feeding when tested in the absence of
DAMGO, as indicated by the lack of significant differences
between vehicle-treated rats and any of the amylin-alone
doses (although there was a small, nonsignificant trend at
the highest dose, 30 ng). Moreover, amylin (either alone or
in combination with DAMGO) did not affect water intake in
either the high-dose or low-dose experiment, as evidenced
by the lack of amylin main effects or amylin DAMGO
interactions (Fs¼0.23–2.5, not significant (NS)). Hence, the
potent reversal of DAMGO-driven feeding by amylin,
particularly at the low, 3-ng amylin dose, was unlikely the
result of nonspecific motor impairment or malaise. It
should be noted that for the group that received lower
doses of amylin, baseline saline/saline and DAMGO/saline
feeding values were higher relative to those for the group
that received higher doses of amylin. However, there
were no systematic differences in injector tip placements
or methodology across groups. These differing values may
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3011
Neuropsychopharmacology
have been due to normally occurring, between-cohort
differences across separate groups of rats.
Amylin Did Not Alter Intra-ADS DAMGO-Induced
Feeding
It has been shown that, outside the Acb, a zone within the
ADS also subserves m-opioid-driven feeding (Bakshi and
Kelley, 1993; DiFeliceantonio et al, 2012). We replicated this
observation, obtaining a main effect of DAMGO in the ADS
(F(1, 5) ¼39.749, Po0.01) on food intake (see Figure 2b,
inset). In contrast to the AcbSh, there was no significant
main effect of 3-ng amylin in the ADS on food intake nor on
DAMGO amylin interaction (Fs¼1.5–2.0, NS). Amylin
also failed to alter water intake, as evidenced by lack of
an amylin main effect or amylin DAMGO interaction
(Fs¼0.6–0.7, NS). Therefore, the same dose of amylin that
reduced the effects of DAMGO on food intake by nearly
50% in the AcbSh was ineffective at reducing DAMGO-
induced feeding in the ADS.
In a Higher Dose Range, Intra-AcbSh Amylin Modestly
Decreased Hunger- or Palatability-Induced Feeding
(Without DAMGO)
There was no main effect of AcbSh amylin on sucrose intake
(F(3, 21) ¼1.9, NS), although a directed contrast showed a
significant difference between the saline condition and the
Amylin 30-ng condition, with the Amylin 30-ng condition
slightly suppressing sucrose intake (Po0.05, Figure 3a).
However, amylin failed to alter water intake in this experi-
ment (F(3, 21) ¼0.7, NS). AcbSh amylin had a significant
main effect on chow intake in food-deprived rats (F(3, 18) ¼
4.2, Po0.02) (see Figure 3b). Post hoc tests showed a
Figure 1 Injector placements for accumbens shell (AcbSh) and anterior dorsal striatum (ADS)-cannulated animals. Photomicrographs at the top of each
column show injector placements into the AcbSh (left) and ADS (right). Arrows indicate location of injector tips. Below the photomicrographs, line drawings
of coronal sections (with position of each section given in mm from bregma) show injection sites from rats with bilateral placements in AcbSh and ADS.
Each injector tip placement is represented by a dot color-coded by experiment; red and orange represent low-dose and high-dose amylin vs
DAMGO experiments, respectively; green represents the ADS study; blue represents hunger- and palatability-driven feeding; violet represents the AC187/
prefeeding study. ac, anterior commissure; cc, corpus callosum; LV, lateral ventricle. Line drawings were adapted from the atlas of Paxinos and Watson
(2007), with permission.
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3012
Neuropsychopharmacology
significant difference between the saline and amylin 30-ng
conditions (Po0.01), but not between saline and other
amylin doses. This was the only experiment in which
amylin affected water intake (F(3, 18) ¼3.3, Po0.05),
producing a significant (50%) decrease at the 30-ng dose
(Po0.008). No other dose significantly altered water intake.
These results further indicate that the reversal of DAMGO-
induced feeding by substantially lower amylin doses
(as observed in the aforementioned experiments) was not
the consequence of a nonspecific motivational or motoric
impairment.
Intra-AcbSh AMY-R Blockade Significantly Reversed
the Ability of Prefeeding to Suppress DAMGO-Induced
Food Intake
As expected, food-deprived rats that were given a 30-min
chow prefeeding session 15 min prior to the 30-min chow
testing session ate less than rats that were not prefed (main
effect of prefeeding: F(1, 6) ¼24.8, Po0.003). Also, DAMGO
had a significant main effect on food intake in both prefed
and non-prefed rats (F(1, 6) ¼268.2, Po0.0001). Again,
as expected, DAMGO-induced hyperphagia was lower after
prefeeding (Po0.0001, Figure 4). There was a significant
interaction between DAMGO and the AMY-R antagonist,
AC187 (F(1, 6) ¼6.1, Po0.05). Comparisons among means
revealed a significant difference between the prefed/
DAMGO condition compared with the prefed/DAMGO/
AC187 condition (Po0.05), with rats in the latter condition
eating more, thus demonstrating that blocking AMY-Rs
partly reverses the ability of prefeeding to diminish
m-opioid-driven food intake (Figure 4). Interestingly,
AC187 did not augment feeding in rats not treated with
DAMGO, suggesting that the modulatory effect of endo-
genous AcbSh AMY-R signaling exhibits some specificity
for excessive, mu-opioid-driven appetitive responses. For
additional means comparisons, see Figure 4 legend. For
water intake, there was no significant main effect of AC187,
AC187 DAMGO interaction, or feeding-status AC187
DAMGO interaction (Fs¼0.02–3.2, NS).
To explore the possibility of carry-over effects arising
from repeated exposure to food-restriction over the course
of the experiment, we conducted directed comparisons with
t-tests on sub-cohorts of rats receiving various treatments
either in the first half (days 1–4) or second half (days 5–8)
of the experiment (recall that the order of treatments was
counterbalanced across subjects). The following treatments
were analyzed with regard to possible differences in the first
vs second half: DAMGO, DAMGO þprefeeding, DAMGO þ
AC187, DAMGO þAC187 þprefeeding. These comparisons
revealed no effect of treatment order (ts ¼0.12–0.9, NS),
indicating a lack of carry-over effects over the duration of
the experiment.
DISCUSSION
These results show for the first time a potent modulatory
influence of AMY-R signaling on m-OR-mediated responses
at the level of the AcbSh. Our results demonstrate that
stimulating AMY-Rs with exogenously administered amylin
strongly reduces m-OR agonist-induced feeding at doses
considerably lower than those required to even modestly
diminish either hunger-associated chow intake or palatable
feeding (sucrose drinking). Moreover, blockade of AMY-Rs
partly reversed the ability of prefeeding to suppress intake
engendered by intra-AcbSh DAMGO. Together, these
results reveal a potent negative modulation of m-ORs by
both exogenous and endogenous AMY-R signaling, and
show for the first time a role of endogenous AMY-R ligands
in post-meal-feeding modulation at the level of the AcbSh.
The reversal of DAMGO-associated feeding seen in the
present study ranks among the most potent of the
behavioral effects of amylin obtained from anywhere in
the brain. The lowest dose of exogenously administered,
intra-AcbSh amylin to significantly reduce DAMGO-driven
feeding was 3 ng/side, or 6 ng/rat (1.52 pmol/rat). This dose
is similar to that required to suppress feeding upon infusion
into the third ventricle, immediately adjacent to the medial
basal hypothalamus (1 pmol/rat; Rushing et al, 2000), and
Figure 2 (a) The effects of intra-accumbens shell (AcbSh) amylin
(Vehicle (Veh), 1, or 3 ng) on chow intake elicited by intra-AcbSh DAMGO
(Veh or 0.25 mg). ***Po0.001 compared with Veh/Veh.
þþ
Po0.01
compared with Veh/DAMGO. Inset: Interaction between DAMGO (Veh
or 0.25 mg) and amylin (Veh or 3 ng) upon infusion of both compounds into
the anterior dorsal striaum (ADS). **Po0.01, main effect of DAMGO.
(b) Interaction between higher doses of amylin (Veh, 10, or 30 ng) and
DAMGO (Veh or 0.25 mg) upon infusion of both compounds into the
AcbSh. ***Po0.01, compared with Veh/Veh.
þ
Po0.05,
þþþ
Po0.001
compared with Veh/DAMGO. All testing sessions were 30-min long.
Error bars depict one SEM.
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3013
Neuropsychopharmacology
even lower than the dose required to reduce feeding in the
area postrema, where 10 pmol/rat amylin is effective but
1 pmol/rat is not (Mollet et al, 2004). We also found that the
3-ng/side amylin dose, which robustly suppressed DAMGO-
induced feeding in the AcbSh, was completely ineffective
at altering DAMGO-driven feeding in the ADS. It has been
shown that m-OR stimulation outside the Acb, in select
dorsal striatal regions, increases feeding (Bakshi and Kelley,
1993a; DiFeliceantonio et al, 2012). However, these striatal
territories possess neither AMY-R binding nor expression
of AMY-R-component genes (Sexton et al, 1994; van
Rossum et al, 1994; Baisley et al, 2014). Therefore, our
results indicate that DAMGO-induced hyperphagia is only
reduced when amylin is infused into striatal regions rich in
AMY-R receptors, suggesting that targeting this receptor
may represent a mechanism for modulating opioid effects in
the ventral striatum specifically.
Interestingly, we found that intra-AcbSh amylin was
considerably less potent in its modulation of sucrose
drinking, compared with its effects on DAMGO-driven
feeding; a 30-ng amylin dose was required to produce a
small reduction in sucrose intake, 10-fold higher than that
required to significantly reverse DAMGO-associated feed-
ing. The 30-ng dose is within the parameters reported in the
only prior study of intra-Acb amylin infusion on hunger-
associated chow intake (Baldo and Kelley, 2001), and is
also consistent with results shown in the present study
for hunger-driven feeding. Considering the evidence that
m-opioid signaling in the Acb robustly modulates palatable
feeding (Zhang and Kelley, 1997; Pecina and Berridge, 2005;
Woolley et al, 2006), our initial hypothesis was that
amylin would reverse sucrose intake in a dose range closer
to that observed for the negative modulation of DAMGO
effects. It is worth considering, however, that whereas
intra-AcbSh DAMGO infusions affect m-ORs only in that
structure, sucrose drinking may recruit m-opioid transmis-
sion in multiple redundant sites (Koch et al, 1995; Kim et al,
2004; Smith and Berridge, 2007; Denbleyker et al, 2009).
Therefore, amylin actions (in the dose range tested) in the
AcbSh may not be sufficient to reduce sucrose solution
intake beyond the modest degree seen here. Accordingly,
Kelley et al (1996) found that intra-Acb infusions of
naloxone or naltrexone effectively reduced sucrose drink-
ing, but only by about 20%. Moreover, whereas intra-AcbSh
naloxone did not significantly reduce chow intake, there
was a trend towards a reduction of about 15%. Hence, the
present results with amylin are not inconsistent with these
opioid antagonist findings, in the sense that both intra-Acb
stimulation of AMY-Rs, and blockade of opioid receptors,
reduced modestly, but did not eliminate, both sucrose
intake and hunger-driven feeding.
To explore the role of endogenous AMY-R signaling, we
tested the ability of prefeeding to suppress AcbSh DAMGO-
induced hyperphagia either with or without intra-AcbSh
infusions of the AMY-R antagonist, AC187 (Hay et al, 2005).
Intra-AcbSh AC187 significantly reversed the ability of
prefeeding to suppress DAMGO-induced food intake;
however, this treatment did not alter food intake in non-
DAMGO-treated rats, nor did it elevate DAMGO-induced
feeding in non-prefed rats. These results suggest some
Figure 3 (a) The effects of intra-accumbens shell (AcbSh) amylin (Amy), (vehicle (Veh), 3, 10 or 30 ng) on intake of a 10% sucrose solution. *Po0.05,
compared with Veh condition. (b) Effects of intra-AcbSh Amy (Veh, 3, 10, or 30 ng) in 18-h food-deprived rats during a 30-minute testing session. **Po0.01
compared with Veh condition. DAMGO was not given in either experiment. All testing sessions were 30-min long. Error bars depict one SEM.
Figure 4 The effects of intra-accumbens shell (AcbSh) infusions of
DAMGO (0.25 mg) plus AC187 (20 mg) combinations on chow intake in
grams (g) during 30 min testing sessions. All rats were food-deprived for
18 h. Non-prefed rats were given either drug or ‘mock’ infusions (see text)
directly before the 30 min feeding test session. Prefed rats ate chow in a
30 min prefeeding session, were given drug infusions, and then were tested
in a second 30-min feeding session. See text for further methodological
details. Values represent means±SEM. *Po0.05, ***Po0.001 compared
with Non-Prefed/DAMGO/Mock condition.
þ
Po0.05 between the
Prefed/DAMGO/Mock and Prefed/DAMGO/AC187 conditions.
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3014
Neuropsychopharmacology
degree of specificity of AMY-R modulation for m-opioid
function. One possible explanation for these effects is
that the AMY-R ligands that negatively modulate m-OR
responses fluctuate according to prandial stimuli, with the
highest levels occurring in the postprandial period. One
candidate ligand is peripherally secreted amylin, which is
co-released with insulin in response to feeding and
macronutrient flux (Ogawa et al, 1990; Arnelo et al, 1998).
According to this hypothesis, prefed rats could have higher
amylin levels than the non-prefed rats and this elevated
amylin ‘tone’ may underlie the reduction of opioid-driven
feeding in the early postprandial period. Given that the non-
prefed, food-deprived rats likely had lower levels of amylin,
the lack of AC187 effect in non-prefed rats (DAMGO-treated
or otherwise) could reflect a paucity of endogenous ligand
in the Acb, and, consequently, negligible levels of endo-
genous AMY-R signaling to block. The question arises,
however, as to whether sufficient levels of amylin cross the
blood-brain barrier to enact behavioral effects. Studies with
radiolabeled peptides showed that intact amylin accumu-
lates in multiple brain structures, including the striatum,
after systemic injection in mice, possibly via a saturable
transport mechanism. Indeed, amylin showed greater over-
all brain penetrance that insulin (Banks and Kastin, 1998).
Nevertheless, caution should be used in interpreting these
results, as only a small fraction of the systemically adminis-
tered amylin reached the brain and the striatum was among
the sites showing relatively lower levels of amylin accumu-
lation. A more definitive answer awaits detailed analysis
of real-time amylin flux in the Acb, using sensitive mass
spectrometry-based methods. Another possibility (though
not mutually exclusive) is that the endogenous AMY-R
ligand is CGRP. There are appreciable densities of
CGRP-like immunoreactive fibers in the Acb, and relatively
high densities of CGRP binding (Kruger et al, 1988; van
Rossum et al, 1997). CGRP binds to the Acb-localized
AMY-R, albeit with less affinity than amylin (Beaumont
et al, 1993). Hence, it is possible that either CGRP, amylin,
or a combination of both ligands participate in postprandial
m-OR modulation through AMY-Rs. Further studies are
needed to clarify this issue. Regardless, the present results
clearly indicate for the first time that there is a negative-
modulatory interaction between endogenous AMY-R and
m-opioid systems at the level of the AcbSh; this interaction
is revealed immediately following a meal.
Note that the lack of AC187-induced feeding augmenta-
tion in DAMGO-treated, non-prefed rats could be attributed
to a ceiling effect. However, close examination of intake
levels in individual rats shows that roughly half of the rats
ate more during the food deprivation þDAMGO þAC187
condition relative to food deprivation þDAMGO—includ-
ing the rat exhibiting the highest intake score under
DAMGO alone—whereas the other half ate less (data not
shown). This pattern would tend to argue against the idea
that there was no room to move upward under the non-
prefed-DAMGO þAC187 condition.
Presently, the mechanism underlying AMY-R and m-OR
interaction is unknown. However, it is interesting to consi-
der that the high-affinity AMY-1 receptor is a G-protein
coupled receptor that increases intracellular cAMP levels,
and that m-ORs are coupled to G(i)-proteins, which decrease
intracellular cAMP levels (Morfis et al, 2008; Williams et al,
2013). Therefore, it is possible that the AMY-Rs may
negatively modulate m-ORs via interactions between post-
receptor cAMP-dependent transduction pathways.
Clinically, our results may be relevant to disorders such
as binge-eating disorder and bulimia nervosa. Mu-opioid
signaling in the CNS is implicated in both disorders;
accordingly, there is some evidence that opioid-blocking
drugs (including selective m-OR antagonists) ameliorate at
least some symptoms of these disorders, and an association
has been reported between binge-eating disorder and a
gain-of-function polymorphism of the m-OR gene (Marrazzi
et al, 1995; Davis et al, 2009; Berner et al, 2011; Ziauddeen
et al, 2013). A theoretical framework has been proposed
stating that intra-Acb m-OR signaling acts to extend
feeding (particularly on palatable foods) beyond physiolo-
gical need, resulting in excess caloric intake (Kelley et al,
2005). Hence, in addition to its established clinical role in
the regulation of Type 2 diabetes mellitus, the FDA-
approved amylin analog, Pramlintide, may be useful
treatment for excessive, m-opioid-driven non-homeostatic
palatable feeding, as occurs putatively in pathological
conditions such as binge-type eating disorders and obesity.
Beyond feeding, AMY-R-based drugs may have therapeutic
effects in opiate and alcohol craving, conditions in which
both the Acb, and m-OR transmission, have been implicated
(O’Brien, 2005).
In summary, this is the first study to examine interactions
between AcbSh m-ORs and amylin. We find that AMY-R
signaling enacts robust negative modulation over m-OR-
mediated responses, highlighting a novel receptor-based
mechanism with which to modulate central m-OR signal-
ing in multiple ‘disorders of appetitive motivation,’ includ-
ing, but not limited to, psychiatric disorders with binge
features.
FUNDING AND DISCLOSURE
The authors declare no conflict of interest.
ACKNOWLEDGEMENTS
This work was supported by R21 MH093824 (BAB), and
SKB was supported by training grant T32 GM007507. We
are grateful to Ken Sadeghian and Ryan Selleck for technical
assistance. Facilities and procedures complied with animal
use and care guidelines from the National Institutes of
Health of the USA, and were approved by the Institutional
Animal Care and Use Committee of the University of
Wisconsin.
REFERENCES
Ahren B, Sundler F (1992). Localization of calcitonin gene-related
peptide and islet amyloid polypeptide in the rat and mouse
pancreas. Cell Tissue Res 269: 315–322.
Alam T, Chen L, Ogawa A, Leffert JD, Unger RH, Luskey KL (1992).
Coordinate regulation of amylin and insulin expression in
response to hypoglycemia and fasting. Diabetes 41: 508–514.
Arnelo U, Reidelberger R, Adrian TE, Larsson J, Permert J (1998).
Sufficiency of postprandial plasma levels of islet amyloid
polypeptide for suppression of feeding in rats. Am J Physiol
275: R1537–R1542.
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3015
Neuropsychopharmacology
Baisley SK, Bremer QZ, Bakshi VP, Baldo BA (2014). Antipsycho-
tic-like actions of the satiety peptide, amylin, in ventral striatal
regions marked by overlapping calcitonin receptor and RAMP-1
gene expression. J Neurosci 34: 4318–4325.
Bakshi VP, Kelley AE (1993). Striatal regulation of morphine-
induced hyperphagia: an anatomical mapping study. Psycho-
pharmacology (Berl) 111: 207–214.
Baldo BA, Kelley AE (2001). Amylin infusion into rat nucleus
accumbens potently depresses motor activity and ingestive behavior.
Am J Physiol Regul Integr Comp Physiol 281: R1232–R1242.
Banks WA, Kastin AJ (1998). Differential permeability of the
blood-brain barrier to two pancreatic peptides: insulin and
amylin. Peptides 19: 883–889.
Beaumont K, Kenney MA, Young AA, Rink TJ (1993). High affinity
amylin binding sites in rat brain. Mol Pharmacol 44: 493–497.
Berner LA, Bocarsly ME, Hoebel BG, Avena NM (2011).
Pharmacological interventions for binge eating: lessons from
animal models, current treatments, and future directions. Curr
Pharm Des 17: 1180–1187.
Bouali SM, Wimalawansa SJ, Jolicoeur FB (1995). In vivo central
actions of rat amylin. Regul Pept 56: 167–174.
Butler PC, Chou J, Carter WB, Wang YN, Bu BH, Chang D et al
(1990). Effects of meal ingestion on plasma amylin concen-
tration in NIDDM and nondiabetic humans. Diabetes 39:
752–756.
Chance WT, Balasubramaniam A, Zhang FS, Wimalawansa SJ,
Fischer JE (1991). Anorexia following the intrahypothalamic
administration of amylin. Brain Res 539: 352–354.
Christopoulos G, Paxinos G, Huang XF, Beaumont K, Toga AW,
Sexton PM (1995). Comparative distribution of receptors for
amylin and the related peptides calcitonin gene related peptide
and calcitonin in rat and monkey brain. Can J Physiol Pharmacol
73: 1037–1041.
Davis CA, Levitan RD, Reid C, Carter JC, Kaplan AS, Patte KA et al
(2009). Dopamine for ‘wanting’ and opioids for ‘liking’: a
comparison of obese adults with and without binge eating.
Obesity (Silver Spring) 17: 1220–1225.
Denbleyker M, Nicklous DM, Wagner PJ, Ward HG, Simansky KJ
(2009). Activating mu-opioid receptors in the lateral para-
brachial nucleus increases c-Fos expression in forebrain areas
associated with caloric regulation, reward and cognition.
Neuroscience 162: 224–233.
DiFeliceantonio AG, Mabrouk OS, Kennedy RT, Berridge KC
(2012). Enkephalin surges in dorsal neostriatum as a signal to
eat. Curr Biol 22: 1918–1924.
Hay DL, Christopoulos G, Christopoulos A, Poyner DR, Sexton PM
(2005). Pharmacological discrimination of calcitonin receptor:
receptor activity-modifying protein complexes. Mol Pharmacol
67: 1655–1665.
Kelley AE, Baldo BA, Pratt WE (2005). A proposed hypothalamic-
thalamic-striatal axis for the integration of energy balance,
arousal, and food reward. J Comp Neurol 493: 72–85.
Kelley AE, Bless EP, Swanson CJ (1996). Investigation of the effects
of opiate antagonists infused into the nucleus accumbens on
feeding and sucrose drinking in rats. J Pharmacol Exp Ther 278:
1499–1507.
Kim EM, Quinn JG, Levine AS, O’Hare E (2004). A bi-directional
mu-opioid-opioid connection between the nucleus of the
accumbens shell and the central nucleus of the amygdala in
the rat. Brain Res 1029: 135–139.
Koch JE, Glass MJ, Cooper ML, Bodnar RJ (1995). Alterations in
deprivation, glucoprivic and sucrose intake following general,
mu and kappa opioid antagonists in the hypothalamic para-
ventricular nucleus of rats. Neuroscience 66: 951–957.
Kruger L, Mantyh PW, Sternini C, Brecha NC, Mantyh CR (1988).
Calcitonin gene-related peptide (CGRP) in the rat central
nervous system: patterns of immunoreactivity and receptor
binding sites. Brain Res 463: 223–244.
Lutz TA, Mollet A, Rushing PA, Riediger T, Scharrer E (2001).
The anorectic effect of a chronic peripheral infusion of amylin
is abolished in area postrema/nucleus of the solitary tract (AP/
NTS) lesioned rats. Int J Obes Relat Metab Disord 25: 1005–1011.
Lutz TA, Rossi R, Althaus J, Del Prete E, Scharrer E (1997).
Evidence for a physiological role of central calcitonin gene-
related peptide (CGRP) receptors in the control of food intake in
rats. Neurosci Lett 230: 159–162.
Lutz TA, Rossi R, Althaus J, Del Prete E, Scharrer E (1998a).
Amylin reduces food intake more potently than calcitonin gene-
related peptide (CGRP) when injected into the lateral brain
ventricle in rats. Peptides 19: 1533–1540.
Lutz TA, Senn M, Althaus J, Del Prete E, Ehrensperger F,
Scharrer E (1998b). Lesion of the area postrema/nucleus of the
solitary tract (AP/NTS) attenuates the anorectic effects of amylin
and calcitonin gene-related peptide (CGRP) in rats. Peptides 19:
309–317.
Marrazzi MA, Bacon JP, Kinzie J, Luby ED (1995). Naltrexone
use in the treatment of anorexia nervosa and bulemia nervosa.
Int Clin Psychopharmacol 10: 163–172.
Mietlicki-Baase EG, Rupprecht LE, Olivos DR, Zimmer DJ, Alter
MD, Pierce RC et al (2013). Amylin receptor signaling in the
ventral tegmental area is physiologically relevant for the control
of food intake. Neuropsychopharmacology 38: 1685–1697.
Mollet A, Gilg S, Riediger T, Lutz TA (2004). Infusion of the amylin
antagonist AC 187 into the area postrema increases food intake
in rats. Physiol Behav 81: 149–155.
Moore CX, Cooper GJ (1991). Co-secretion of amylin and insulin
from cultured islet beta-cells: modulation by nutrient secreta-
gogues, islet hormones and hypoglycemic agents. Biochem
Biophys Res Commun 179: 1–9.
Morfis M, Tilakaratne N, Furness SG, Christopoulos G, Werry TD,
Christopoulos A et al (2008). Receptor activity-modifying
proteins differentially modulate the G protein-coupling effi-
ciency of amylin receptors. Endocrinology 149: 5423–5431.
Morley JE, Flood JF (1991). Amylin decreases food intake in mice.
Peptides 12: 865–869.
Ogawa A, Harris V, McCorkle SK, Unger RH, Luskey KL (1990).
Amylin secretion from the rat pancreas and its selective loss
after streptozotocin treatment. J Clin Invest 85: 973–976.
O’Brien CP (2005). Anticraving medications and relapse preven-
tion: a possible new class of psychoactive medications. Am J
Psychiat 162: 1423–1431.
Paxinos G, Watson C (2007). The rat brain in stereotaxic
coordinates, 6th edn, Academic Press.
Pecina S, Berridge KC (2005). Hedonic hot spot in nucleus
accumbens shell: where do mu-opioids cause increased hedonic
impact of sweetness? J Neurosci 25: 11777–11786.
Perry ML, Baldo BA, Andrzejewski ME, Kelley AE (2009).
Muscarinic receptor antagonism causes a functional alteration
in nucleus accumbens mu-opiate-mediated feeding behavior.
Behav Brain Res 197: 225–229.
Poyner DR, Sexton PM, Marshall I, Smith DM, Quirion R,
Born W et al (2002). International Union of Pharmacology.
XXXII. The mammalian calcitonin gene-related peptides,
adrenomedullin, amylin, and calcitonin receptors. Pharmacol
Rev 54: 233–246.
Rushing PA, Hagan MM, Seeley RJ, Lutz TA, Woods SC (2000).
Amylin: a novel action in the brain to reduce body weight.
Endocrinology 141: 850–853.
Sexton PM, Paxinos G, Kenney MA, Wookey PJ, Beaumont K
(1994). In vitro autoradiographic localization of amylin binding
sites in rat brain. Neuroscience 62: 553–567.
Singh-Franco D, Perez A, Harrington C (2011). The effect of
pramlintide acetate on glycemic control and weight in patients
with type 2 diabetes mellitus and in obese patients without
diabetes: a systematic review and meta-analysis. Diabetes Obes
Metab 13: 169–180.
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3016
Neuropsychopharmacology
Smith KS, Berridge KC (2007). Opioid limbic circuit for reward:
interaction between hedonic hotspots of nucleus accumbens and
ventral pallidum. J Neurosci 27: 1594–1605.
Stratford TR, Kelley AE (1997). GABA in the nucleus accumbens
shell participates in the central regulation of feeding behavior.
J Neurosci 17: 4434–4440.
van Rossum D, Hanisch UK, Quirion R (1997). Neuroanatomical
localization, pharmacological characterization and functions of
CGRP, related peptides and their receptors. Neurosci Biobehav
Rev 21: 649–678.
van Rossum D, Menard DP, Fournier A, St-Pierre S, Quirion R
(1994). Autoradiographic distribution and receptor binding
profile of [125I]Bolton Hunter-rat amylin binding sites in the
rat brain. J Pharmacol Exp Ther 270: 779–787.
Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M,
Schulz S et al (2013). Regulation of mu-opioid receptors:
desensitization, phosphorylation, internalization, and tolerance.
Pharmacol Rev 65: 223–254.
Woolley JD, Lee BS, Fields HL (2006). Nucleus accumbens opioids
regulate flavor-based preferences in food consumption. Neuro-
science 143: 309–317.
Zhang M, Kelley AE (1997). Opiate agonists microinjected into the
nucleus accumbens enhance sucrose drinking in rats. Psycho-
pharmacology (Berl) 132: 350–360.
Zhang M, Kelley AE (2000). Enhanced intake of high-fat food
following striatal mu-opioid stimulation: microinjection map-
ping and Fos expression. Neuroscience 99: 267–277.
Ziauddeen H, Chamberlain SR, Nathan PJ, Koch A, Maltby K,
Bush M et al (2013). Effects of the mu-opioid receptor antagonist
GSK1521498 on hedonic and consummatory eating behaviour:
a proof of mechanism study in binge-eating obese subjects.
Mol Psychiatry 18: 1287–1293.
Intra-accumbens amylin/opioid interactions
SK Baisley and BA Baldo
3017
Neuropsychopharmacology
... In addition, amylin decreases homeostatic and hedonic feeding [for review, see (Hartter et al., 1991;Hay, 2017;Boyle et al., 2018)]. These anorexigenic properties involve amylin receptors (AMYRs) within brain regions highly linked to reward regulation like the laterodorsal tegmental area (LDTg), ventral tegmental area (VTA), and nucleus accumbens (NAc) (Mietlicki-Baase et al., 2013;Baisley and Baldo, 2014;Mietlicki-Baase et al., 2015;Mietlicki-Baase et al., 2017;Reiner et al., 2017). Therefore, AMYR agonists are evaluated as obesity therapies, as they reduce body weight in both rodents and humans (Aronne et al., 2007;Lutz, 2012;Young, 2012). ...
... In humans, cocaine reduces amylin in plasma (Bouhlal et al., 2017;Angarita et al., 2021). Additionally to reward induced by addictive drugs, the amylinergic pathway regulates natural rewards like sexual behaviours (Clementi et al., 1999) and hedonic feeding (Mietlicki-Baase et al., 2013;Baisley and Baldo, 2014;Mietlicki-Baase et al., 2015;Mietlicki-Baase et al., 2017;Reiner et al., 2017). This may be associated with the ability of sCT to prevent a reward from activating the mesolimbic dopamine system, as sCT prevents the VTA-stimulated or drug-induced dopamine release in Nac shell (Whiting et al., 2017;Kalafateli A. L. et al., 2019;Kalafateli et al., 2021) We further found that sCT prevents the acquisition of nicotine-induced locomotor sensitisation when sCT and nicotine are co-administered repeatedly. ...
Article
Full-text available
The behavioural responses to nicotine involve appetite-regulatory hormones; however, the effects of the anorexigenic hormone amylin on reward-related behaviours induced by nicotine remain to be established. Previous studies have shown that the amylinergic pathway regulates behavioural responses to alcohol, amphetamine and cocaine. Here, we evaluated the effects of salmon calcitonin (sCT), an amylin and calcitonin receptor (CTR) agonist, on nicotine-induced locomotor stimulation and sensitisation as well as dopamine release in the nucleus accumbens (NAc) shell. Moreover, we investigated the effects of sCT on the acquisition and expression of nicotine-induced reward in the conditioned place preference (CPP) paradigm. Finally, we performed Western Blot experiments in an attempt to identify the levels of the amylin receptor components CTRa, CTRb, and RAMP1 in reward-related areas of mice responding differently to repeated injections of sCT and nicotine in the locomotor sensitisation test. We found that sCT blocked nicotine’s stimulatory and dopamine-releasing effects and prevented its ability to cause locomotor sensitisation. On the other hand, sCT did not alter nicotine-induced acquisition and expression of CPP. Lastly, sCT-nicotine treated mice from the locomotor sensitisation experiment displayed higher levels of total CTR, i.e. CTRa and CTRb together, in the reward-processing laterodorsal tegmental area (LDTg) of the brain compared to mice treated with vehicle-nicotine. Overall, the present data reveal that activation of CTR or/and amylin receptors attenuates certain nicotine-induced behaviours in male mice, further contributing to the understanding of appetite-regulatory peptides in reward regulation.
... The most plausible confounding factor is malaise, which is a common side effect of the GLP-1R agonists used (Kanoski et al., 2012). Although indirectly excluding this factor, the tested GLP-1R agonists act as follows: 1) they do not cause a CPP per se as an indication of conditioned aversion (Cagniard and Murphy, 2012), 2) they are used in dosages that are not associated with malaise (Cagniard and Murphy, 2012;Kanoski et al., 2012;Mietlicki-Baase et al., 2013;Baisley and Baldo, 2014;Mietlicki-Baase et al., 2017;Rodriguez et al., 2018), 3) they enhance water intake, and 4) they reduce alcohol/drug responses that are unaffected by aversion [for a review, see Shevchouk et al. (2021)]. On a final note, preclinical models are combined to reflect the responses in animals, although animals cannot be diagnosed with an addiction, so this is a limitation associated with preclinical studies. ...
Article
Full-text available
Although the multifaceted mechanisms underlying alcohol use disorder (AUD) have been partially defined, the neurobiological complexity of this disorder is yet to be unraveled. One of the systems that have gained attention in recent times is the gut-brain axis. Although numerous peptides participate in this axis, glucagon-like peptide-1 (GLP-1) plays a central role. GLP-1 is a crucial anorexigenic peptide, with potent abilities to reduce food intake and body weight. The physiological complexity of GLP-1 entails glucose homeostasis, gastrointestinal motility, and the release of insulin and glucagon. As reviewed in this study, acute or repeated treatment with GLP-1 receptor (GLP-1R) agonists decreases alcohol consumption in rodents. Moreover, the abilities of alcohol to promote hyperlocomotion, dopamine release in the nucleus accumbens, and reward in the conditioned place preference paradigm are all suppressed by GLP-1R ligands. Moreover, activation of GLP-1R suppresses the motivation to consume alcohol, alcohol-seeking behaviors, and relapse drinking in male rodents. Similarly, abstinence symptoms experienced during alcohol withdrawal are attenuated by activation of the GLP-1 pathway. On a similar note, the activation of GLP-1 receptors within areas of the brain that are processing reward modulates these alcohol-related responses. Another area that is crucial for this ability is the nucleus of the solitary tract, which is where GLP-1 is produced and from which GLP-1-containing neurons project to areas of reward. These findings may have clinical relevance as AUD is associated with polymorphisms in GLP-1-related genes. Although a GLP-1R agonist does not alter alcohol intake in AUD patients, it reduces this consumption in a sub-population of obese AUD individuals. Given the uncertainty of this outcome, additional clinical studies of obese AUD patients should explore the effects of the GLP-1R agonists on alcohol intake and body weight. Furthermore, GLP-1 receptors modulate the behavioral and neurochemical responses to addictive drugs. Taken together, these preclinical and clinical findings imply that the GLP-1 pathway plays a role in the complex mechanisms regulating alcohol and drug consumption patterns, unveiling a novel aspect of addiction medicine.
... GLP-1, PYY, leptin, and PP are anorexigenic hormones while ghrelin is orexigenic. In addition, amylin, which is released by pancreatic β-cells, may also act in the brain, producing satiety-like effects [34]. Consequently, all these hormones are involved in the highly complex process of appetite/satiety regulation [35]. ...
Article
Full-text available
Cereal β-glucans are beneficial health ingredients that reduce cholesterolemia and postprandial glycaemia. However, their impact on digestive hormones and gut microbiota is not yet fully established. Two randomized, double-blind, controlled studies were conducted. In the first study, 14 subjects ingested a breakfast with or without β-glucan from oats (5.2 g). Compared to the control, β-glucan increased orocecal transit time (p = 0.028) and decreased mean appetite score (p = 0.014) and postprandial plasma ghrelin (p = 0.030), C-peptide (p = 0.001), insulin (p = 0.06), and glucose (p = 0.0006). β-glucan increased plasma GIP (p = 0.035) and PP (p = 0.018) without affecting leptin, GLP-1, PYY, glucagon, amylin, or 7α-hydroxy-4-cholesten-3-one, a biomarker of bile acid synthesis. In the second study, 32 subjects were distributed into 2 groups to ingest daily foods with (3 g/day) or without β-glucan for 3 weeks; stools were collected before/after treatment. No changes in fecal microbiota composition/diversity (deep sequencing) were detected with β-glucans. These results indicate that acute intake of 5 g β-glucan slows transit time and decreases hunger sensation and postprandial glycaemia without affecting bile-acid synthesis, these changes being associated with decreased plasma insulin, C-peptide, and ghrelin, and increased plasma GIP and PP. However, regular daily intake of 3 g β-glucan is not sufficient to have an effect on fecal microbiota composition.
... More recently, amylin and amylin analogs like pramlintide (PRAM) have been linked to several CNS processes and diseases, most notably with cognition and AD pathogenesis [9][10][11][12][13][14][15][16][17][18][19][20][21][22][23]. These findings are supported by the fact that amylin receptor (AMYR) components are expressed throughout the CNS, more specifically, in areas relevant to cognition and AD pathogenesis [6,[24][25][26][27][28][29][30][31][32][33]. Our group, as well as others, have shown that plasma amylin levels positively correlate with cognition and negatively correlate with AD pathologies [10,13,34]. ...
Article
Background: Amylin, a pancreatic amyloid peptide involved in energy homeostasis, is increasingly studied in the context of Alzheimer's disease (AD) etiology. To date, conflicting pathogenic and neuroprotective roles for this peptide and its analogs for AD pathogenesis have been described. Objective: Whether the benefits of amylin are associated with peripheral improvement of metabolic tone/function or directly through the activation of central amylin receptors is also unknown and downstream signaling mechanisms of amylin receptors are major objectives of this study. Methods: To address these questions more directly we delivered the amylin analog pramlintide systemically (IP), at previously identified therapeutic doses, while centrally (ICV) inhibiting the receptor using an amylin receptor antagonist (AC187), at doses known to impact CNS function. Results: Here we show that pramlintide improved cognitive function independently of CNS receptor activation and provide transcriptomic data that highlights potential mechanisms. Furthermore, we show than inhibition of the amylin receptor increased amyloid-beta pathology in female APP/PS1 mice, an effect than was mitigated by peripheral delivery of pramlintide. Through transcriptomic analysis of pramlintide therapy in AD-modeled mice we found sexual dimorphic modulation of neuroprotective mechanisms: oxidative stress protection in females and membrane stability and reduced neuronal excitability markers in males. Conclusion: These data suggest an uncoupling of functional and pathology-related events and highlighting a more complex receptor system and pharmacological relationship that must be carefully studied to clarify the role of amylin in CNS function and AD.
... Also, insulin receptors in the NAc directly respond to glucose (137), which is relevant to the systemic circulation and food intake drive, since insulin does get past the blood-brain barrier (138). Amylin, which also crosses the blood-brain barrier (139), activates amylin receptors in the NAcSh to suppress feeding (140). PYY and GLP-1 also influence activity in the NAc, but most likely via dopaminergic inputs originating from receptors for these hormones in the VTA (141). ...
Article
Full-text available
This review focuses on summarizing current knowledge on how time-restricted feeding (TRF) and continuous caloric restriction (CR) affect central neuro-endocrine systems involved in regulating satiety. Several interconnected regions of the hypothalamus, brainstem, and cortical areas of the brain are involved in the regulation of satiety. Following CR and TRF, the increase in hunger and reduction of satiety signals of the melanocortin system (NPY, POMC, and AgRP) appear similar between CR and TRF protocols, as do the dopaminergic responses in the mesocorticolimbic circuit. However, ghrelin and leptin signaling via the melanocortin system appears to improve energy balance signals and reduce hyperphagia following TRF, which has not been reported in CR. In addition to satiety systems, CR and TRF also influence circadian rhythms. CR influences the SCN (suprachiasmatic nucleus) or the primary circadian clock as seen by increased clock gene expression. In contrast, TRF appears to affect both the SCN and the peripheral clocks, as seen by phasic changes in the non-SCN (potentially the elusive food entrainable oscillator) and metabolic clocks. The peripheral clocks are influenced by the primary circadian clock but are also entrained by food timing, sleep timing, and other lifestyle parameters, which can supersede the metabolic processes that are regulated by the primary circadian clock. Taken together, TRF influences hunger/satiety, energy balances systems, and circadian rhythms, suggesting a role for adherence to CR in the long run if implemented using the TRF approach. However, these suggestions are based on only a few studies, and future investigations that use standardized protocols for the evaluation of the effect of these diet patterns (time, duration, meal composition, sufficiently powered) are necessary to verify these preliminary observations.
... Although these data would seem to indicate that amylin signaling in the accumbens does not play a role in drinking behavior, the potential role of NAcC/NAcSh amylin signaling in modulating water intake as a result of different types of dehydration is not well-studied. This is an important consideration because accumbens amylin signaling has been shown to modulate the feeding effects of the opioid system [159]. Therefore, it seems important to consider the possibility that amylin, although not producing observable effects on its own, may interact with other signals to produce changes in fluid intake. ...
Article
Maintaining fluid balance is critical for life. The central components that control fluid intake are only partly understood. This contribution to the collection of papers highlighting work by members of the Society for the Study of Ingestive Behavior focuses on the role that dopamine has on fluid intake and describes the roles that various bioregulators can have on thirst and sodium appetite by influencing dopamine systems in the brain. The goal of the review is to highlight areas in need of more research and to propose a framework to guide that research. We hope that this framework will inspire researchers in the field to investigate these interesting questions in order to form a more complete understanding of how fluid intake is controlled.
Article
Full-text available
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol’s rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Article
A plethora of studies to date has examined the roles of feeding-related peptides in the control of food intake. However, the influence of these peptides on the intake of particular macronutrient constituents of food – carbohydrate, fat, and protein – has not been as extensively addressed in the literature. Here, the roles of several feeding-related peptides in controlling macronutrient intake are reviewed. Next, the relationship between macronutrient intake and diseases including diabetes mellitus, obesity, and eating disorders are examined. Finally, some key considerations in macronutrient intake research are discussed. We hope that this review will shed light onto this underappreciated topic in ingestive behavior research and will help to guide further scientific investigation in this area.
Article
The peptide hormone amylin reduces food intake and body weight, and is an attractive candidate target for novel pharmacotherapies to treat obesity. However, the short half-life of native amylin and amylin analogs like pramlintide limits these compounds' potential utility in promoting sustained negative energy balance. Here, we evaluate the ability of the novel long-acting amylin/calcitonin receptor agonist ZP5461 to reduce feeding and body weight in rats, and also test the role of calcitonin receptors (CTRs) in the dorsal vagal complex (DVC) of the hindbrain in the energy balance effects of chronic ZP5461 administration. Acute dose-response studies indicate that systemic ZP5461 (0.5-3 nmol/kg) robustly suppresses energy intake and body weight gain in chow- and high-fat diet (HFD)-fed rats. When HFD-fed rats received chronic systemic administration of ZP5461 (1-2 nmol/kg), the compound initially produced reductions in energy intake and weight gain, but failed to produce sustained suppression of intake and body weight. Using virally-mediated knockdown of DVC CTRs, the ability of chronic systemic ZP5461 to promote early reductions in intake and body weight gain was determined to be mediated in part by activation of DVC CTRs, implicating the DVC as a central site of action for ZP5461. Future studies should address other dosing regimens of ZP5461 to determine whether an alternative dose/frequency of administration would produce more sustained body weight suppression.
Article
Full-text available
Alcohol causes stimulatory behavioral responses by activating reward-processing brain areas including the laterodorsal (LDTg) and ventral tegmental areas (VTA) and the nucleus accumbens (NAc). Systemic administration of the amylin and calcitonin receptor agonist salmon calcitonin (sCT) attenuates alcohol-mediated behaviors, but the brain sites involved in this process remain unknown. Firstly, to identify potential sCT sites of action in the brain, we used immunohistochemistry after systemic administration of fluorescent-labeled sCT. We then performed behavioral experiments to explore how infused sCT into the aforementioned reward-processing brain areas affects acute alcohol-induced behaviors in mice and chronic alcohol consumption in rats. We show that peripheral sCT crosses the blood brain barrier and is detected in all the brain areas studied herein. sCT infused into the LDTg attenuates alcohol-evoked dopamine release in the NAc shell in mice and reduces alcohol intake in rats. sCT into the VTA blocks alcohol-induced locomotor stimulation and dopamine release in the NAc shell in mice and decreases alcohol intake in rats. Lastly, sCT into the NAc shell prevents alcohol-induced locomotor activity in mice. Our data suggest that central sCT modulates the ability of alcohol to activate reward-processing brain regions.
Article
Full-text available
We have demonstrated previously that injections of 6,7-dinitroquinoxaline-2,3-dione into the nucleus accumbens shell (AcbSh) elicits pronounced feeding in satiated rats. This glutamate antagonist blocks AMPA and kainate receptors and most likely increases food intake by disrupting a tonic excitatory input to the AcbSh, thus decreasing the firing rate of a population of local neurons. Because the application of GABA agonists also decreases neuronal activity, we hypothesized that administration of GABA agonists into the AcbSh would stimulate feeding in satiated rats. We found that acute inhibition of cells in the AcbSh via administration of the GABA A receptor agonist muscimol or the GABA B receptor agonist baclofen elicited intense, dose-related feeding without altering water intake. Muscimol-induced feeding was blocked by coadministration of the selective GABA A receptor blocker bicuculline, but not by the GABA B receptor blocker saclofen. Conversely, baclofen-induced feeding was blocked by coadministration of saclofen, but was not affected by bicuculline. Furthermore, we found that increasing local levels of GABA by administration of a selective GABA-transaminase inhibitor, γ-vinyl-GABA, elicited robust feeding in satiated rats, suggesting a physiological role for endogenous AcbSh GABA in the control of feeding. A mapping study showed that although some feeding can be elicited by muscimol injections near the lateral ventricles, the ventromedial AcbSh is the most sensitive site for eliciting feeding. These findings demonstrate that manipulation of GABA-sensitive cells in the AcbSh can have a pronounced, but specific, effect on feeding behavior in rats. They also constitute the initial description of a novel and potentially important component of the central mechanisms controlling food intake.
Article
Full-text available
Amylin is a calcitonin-related peptide co-secreted with insulin, which produces satiety through brainstem-localized receptors; however, its effects in forebrain are poorly understood. The nucleus accumbens shell (AcbSh) exhibits among the densest concentrations of high-affinity amylin binding; nevertheless, these receptors have not been explored beyond one study showing dopamine antagonist-like effects of intra-Acb amylin on feeding and associated behavior (Baldo and Kelley, 2001). Here, we investigated whether intra-Acb amylin signaling modulates prepulse inhibition (PPI), a measure of sensorimotor gating deficient in several illnesses including schizophrenia. First, in situ hybridization revealed marked anatomical gradients for both receptor activity-modifying protein-1 (RAMP-1) and calcitonin receptor gene (CT-R) expression in striatum [coexpression of these genes yields a high-affinity amylin-1 receptor (AMY1-R)], with highest overlap in the medial AcbSh. Intra-AcbSh amylin infusions in rats (0, 30, and 100 ng) reversed amphetamine (AMPH)-induced PPI disruption without affecting baseline startle; dorsal striatal amylin infusions had no effect. Coinfusion of AC187 (20 μg), an antagonist for AMY1-R, blocked the ability of amylin to normalize AMPH-induced PPI disruption, showing the specificity of AcbSh amylin effects to the AMY1-R. Intra-AcbSh AC187 on its own disrupted PPI in a haloperidol-reversible manner (0.1 mg/kg). Thus, AMY1-R may be a potential target for the development of putative antipsychotics or adjunct treatments that oppose metabolic side effects of current medications. Moreover, AMY1-Rs may represent a novel way to modulate activity preferentially in ventral versus dorsal striatum.
Article
Full-text available
Morphine and related µ-opioid receptor (MOR) agonists remain among the most effective drugs known for acute relief of severe pain. A major problem in treating painful conditions is that tolerance limits the long-term utility of opioid agonists. Considerable effort has been expended on developing an understanding of the molecular and cellular processes that underlie acute MOR signaling, short-term receptor regulation, and the progression of events that lead to tolerance for different MOR agonists. Although great progress has been made in the past decade, many points of contention and controversy cloud the realization of this progress. This review attempts to clarify some confusion by clearly defining terms, such as desensitization and tolerance, and addressing optimal pharmacological analyses for discerning relative importance of these cellular mechanisms. Cellular and molecular mechanisms regulating MOR function by phosphorylation relative to receptor desensitization and endocytosis are comprehensively reviewed, with an emphasis on agonist-biased regulation and areas where knowledge is lacking or controversial. The implications of these mechanisms for understanding the substantial contribution of MOR signaling to opioid tolerance are then considered in detail. While some functional MOR regulatory mechanisms contributing to tolerance are clearly understood, there are large gaps in understanding the molecular processes responsible for loss of MOR function after chronic exposure to opioids. Further elucidation of the cellular mechanisms that are regulated by opioids will be necessary for the successful development of MOR-based approaches to new pain therapeutics that limit the development of tolerance.
Article
Amylin is a 37-amino acid peptide hormone that is co-secreted with insulin by pancreatic B cells in response to a nutrient stimulus (e.g., during meals). To test the hypothesis that amylin acts within the brain to reduce long-term food intake and body weight, we examined the effects of acute and chronic 3rd-ventricular (i3vt) infusion of low doses of amylin on food intake and body weight in rats. In one experiment, separate groups of ad lib-fed male Long Evans rats were given one i3vt infusion (3 μl over 30 s) of synthetic cerebrospinal fluid vehicle or 1 to 100 pmol amylin, and food intake and body weight were monitored for 7 days. Amylin potently and dose-dependently reduced 1-h food intake, with all doses producing significant reductions. The largest dose (100 pmol) significantly reduced 24-h intake by over 30%. The effect was persistent in that both 7-day cumulative food intake and body weight change were significantly decreased over the 7 days following a single injection of 100 pmol of amylin. Other groups of rats received continuous i3vt infusion (0.5 μl/h volume) of saline or 2.0 pmol/h amylin via osmotic minipumps over 10 days. Food intake over the 10-day infusion was significantly suppressed in amylin-treated rats as compared to that of controls. Consequently, by the 4th day of infusion, amylin rats weighed significantly less than baseline relative to saline controls, and this difference persisted throughout the remainder of the infusion period. At sacrifice (Day 10), the percent of body weight from retroperitoneal fat depots was significantly lower in the amylin-treated rats, indicative of a reduction of total body adiposity. In summary, the results support the hypothesis that amylin acts as a signal to the brain contributing to the maintenance of long-term energy balance.
Article
The ability of amylin, a pancreatic β-cell-derived neuropeptide, to promote negative energy balance has been ascribed to neural activation at the area postrema. However, despite amylin binding throughout the brain, the possible role of amylin signaling at other nuclei in the control of food intake has been largely neglected. We show that mRNA for all components of the amylin receptor complex is expressed in the ventral tegmental area (VTA), a mesolimbic structure mediating food intake and reward. Direct activation of VTA amylin receptors reduces intake of chow and palatable sucrose solution in rats. This effect is mediated by reductions in meal size and is not due to nausea/malaise or prolonged suppression of locomotor activity. VTA amylin receptor activation also reduces sucrose self-administration on a progressive ratio schedule. Finally, antagonist studies provide novel evidence that VTA amylin receptor blockade increases food intake and attenuates the intake-suppressive effects of a peripherally administered amylin analog, suggesting that amylin receptor signaling in the VTA is physiologically relevant for food intake control and potentially clinically relevant for the treatment of obesity.Neuropsychopharmacology accepted article preview online, 8 March 2013; doi:10.1038/npp.2013.66.
Article
Compulsive overconsumption of reward characterizes dis-orders ranging from binge eating to drug addiction. Here, we provide evidence that enkephalin surges in an anterome-dial quadrant of dorsal neostriatum contribute to generating intense consumption of palatable food. In ventral striatum, mu opioid circuitry contributes an important component of motivation to consume reward [1–4]. In dorsal neostriatum, mu opioid receptors are concentrated within striosomes that receive inputs from limbic regions of prefrontal cortex [5–13]. We employed advanced opioid microdialysis tech-niques that allow detection of extracellular enkephalin levels. Endogenous >150% enkephalin surges in anterior dorsomedial neostriatum were triggered as rats began to consume palatable chocolates. In contrast, dynorphin levels remained unchanged. Furthermore, a causal role for mu opioid stimulation in overconsumption was demonstrated by observations that microinjection in the same anterior dorsomedial quadrant of a mu receptor agonist ([D-Ala2, N-MePhe4, Gly-ol]-enkephalin; DAMGO) generated intense >250% increases in intake of palatable sweet food (without altering hedonic impact of sweet tastes). Mapping by ''Fos plume'' methods confirmed the hyperphagic effect to be anatomically localized to the anteromedial quadrant of the dorsal neostriatum, whereas other quadrants were relatively ineffective. These findings reveal that opioid signals in ante-romedial dorsal neostriatum are able to code and cause motivation to consume sensory reward. Results and Discussion