ArticlePDF Available

The relationship of in vivo central CB1 receptor occupancy to changes in cortical monoamine release and feeding elicited by CB1 receptor antagonists in rats

Authors:

Abstract and Figures

Cannabinoid type 1 (CB(1)) receptor antagonists are reportedly effective in reducing food intake both preclinically and clinically. This may be due in part to their effects on monoamine release in the brain. The level of central CB(1) receptor occupancy underlying these neurobiological effects is unclear. We explored the relationship between in vivo CB(1) receptor occupancy in the frontal cortex and changes in both monoamine release in the medial prefrontal cortex (mPFC) and feeding behavior in rats in response to two orally administered CB(1) receptor antagonists presently in clinical trials, SR141716A (rimonabant) and SLV319. CB(1) receptor occupancy was measured using [(3)H] SR141716A, and these occupancies were related to potencies to mediate increases in dopamine (DA) and norepinephrine (NE) release measured with microdialysis and decreases in consumption of a highly palatable diet (HP). High receptor occupancy levels (>65%) were required to detect increases in monoamine release that were achieved with 3 and 10 mg/kg of SR141716A and 10 mg/kg of SLV319 for DA and 10 mg/kg of SR141716A for NE. Decreases in HP consumption were seen at occupancies higher than 65% for SR141716A that were achieved with 3 and 10 mg/kg. In contrast, decreases in HP consumption were seen at relatively low CB(1) receptor occupancies (11%) for SLV319. The occupancy method described here is an effective tool for interrelating central CB(1) receptor occupancy with neurobiological actions of CB(1) receptor antagonists and for furthering our understanding of the role of CB(1) receptors in central nervous system physiology and pathology.
Content may be subject to copyright.
Psychopharmacology (2006) 184: 2635
DOI 10.1007/s00213-005-0234-x
ORIGINAL INVESTIGATION
Anne B. Need .Richard J. Davis .
Jesline T. Alexander-Chacko .Brian Eastwood .
Eyassu Chernet .Lee A. Phebus .Dana K. Sindelar .
George G. Nomikos
The relationship of in vivo central CB1 receptor occupancy
to changes in cortical monoamine release and feeding elicited
by CB1 receptor antagonists in rats
Received: 27 April 2005 / Accepted: 3 October 2005 / Published online: 18 November 2005
#Springer-Verlag 2005
Abstract Rationale: Cannabinoid type 1 (CB
1
) receptor
antagonists are reportedly effective in reducing food intake
both preclinically and clinically. This may be due in part to
their effects on monoamine release in the brain. The level of
central CB
1
receptor occupancy underlying these neurobi-
ological effects is unclear. Objectives: We explored the rela-
tionship between in vivo CB
1
receptor occupancy in the
frontal cortex and changes in both monoamine release in the
medial prefrontal cortex (mPFC) and feeding behavior in
rats in response to two orally administered CB
1
receptor
antagonists presently in clinical trials, SR141716A (rimo-
nabant) and SLV319. Methods: CB
1
receptor occupancy
was measured using [
3
H] SR141716A, and these occupan-
cies were related to potencies to mediate increases in do-
pamine (DA) and norepinephrine (NE) release measured
with microdialysis and decreases in consumption of a highly
palatable diet (HP). Results: High receptor occupancy
levels (>65%) were required to detect increases in mono-
amine release that were achieved with 3 and 10 mg/kg of
SR141716A and 10 mg/kg of SLV319 for DA and 10 mg/kg
of SR141716A for NE. Decreases in HP consumption were
seen at occupancies higher than 65% for SR141716A that
were achieved with 3 and 10 mg/kg. In contrast, decreases in
HP consumption were seen at relatively low CB
1
receptor
occupancies (11%) for SLV319. Conclusions: The occu-
pancy method described here is an effective tool for interrelating
central CB
1
receptor occupancy with neurobiological actions of
CB
1
receptor antagonists and for furthering our understanding
of the role of CB
1
receptors in central nervous system
physiology and pathology.
Keywords SR141716A .SLV319 .[
3
H]SR141716A .
Dopamine .Norepinephrine .Microdialysis .Receptor
occupancy .Feeding .Cannabinoid .In vivo binding
Introduction
Primarily in the form of marijuana, cannabinoids have been
used therapeutically and recreationally for centuries al-
though the precise mechanisms underlying a variety of
their effects have been poorly understood. While much
remains to be discovered, significant progress has been
made in recent decades. The identification of Δ
9
-tetrahy-
drocannabinol (THC) as the major psychoactive compo-
nent of marijuana (Gaoni and Mechoulam 1964) was the
first step. Hindered by the lipophilicity of the known
ligands, it was more than 20 years later that investigators
identified specific binding sites. With the synthesis of [
3
H]
CP-55,940, binding sites for cannabinoids were elucidated
in the brain (Devane et al. 1988). Two receptors were then
cloned, cannabinoid type 1(CB
1
; Matsuda et al. 1990),
primarily located in the central nervous system (CNS), and
type 2 (CB
2
; Munro et al. 1993), found mainly in the
peripheral immune system and reportedly in perivascular
microglial cells in the brain (see, e.g., Nunez et al. 2004).
Several endogenous compounds have been found to
activate the CB
1
receptor, including arachidonylethanol-
amide (anandamide) (Devane et al. 1992) and 2-arachidon-
oyl glycerol (2-AG) (Mechoulam et al. 1995). Along with
the synthesis of selective antagonists to the CB
1
receptor,
such as SR141716A (N- (piperidin-1-yl)-5-(4-chlorophe-
nyl)-1-(2,4- dichlorophenyl)-4- methyl-1H-pyrazole-3-car-
boxamide-hydrochloride; rimonabant) (Rinaldi-Carmona
et al. 1994), these fairly recent discoveries have led to an
explosion of research aimed at elucidating the many func-
tions of the endocannabinoid systems in CNS physiology
and pathology.
Evidence is now well established for a role of the CB
1
receptor in feeding. One of the best-known effects of
marijuana use is to increase appetite, and this has been
A. B. Need .R. J. Davis .J. T. Alexander-Chacko .
B. Eastwood .E. Chernet .L. A. Phebus .
D. K. Sindelar .G. G. Nomikos (*)
Lilly Research Laboratories,
Lilly Corporate Center,
Indianapolis, IN 46285-0510, USA
e-mail: nomikos_george@lilly.com
Tel.: +1-317-4332541
Fax: +1-317-2765546
confirmed in controlled settings (see, e.g., Abel 1971).
Cannabinoid agonists have been prescribed for the treat-
ment of nausea during chemotherapy and to improve ap-
petite of patients suffering from AIDS (Plasse et al. 1991).
Genetically obese mice have elevated anandamide and 2-
AG levels in the hypothalamus, and intravenous adminis-
tration of leptin, a key regulator of food intake, suppresses
hypothalamic levels of endocannabinoids in these mice, as
well as in normal rats (Di Marzo et al. 2001). SR141716A
administration to rodents decreases sucrose consumption
(Arnone et al. 1997) and reduces the intake of a highly
palatable diet in marmosets (Simiand et al. 1998). It was also
shown in nondeprived rats to decrease the intake of all food,
regardless of macronutrient composition (Verty et al. 2004).
Moreover, CB
1
receptor knockout mice display decreased
neuropeptide Y (NPY)-induced overeating (Poncelet et al.
2003) and are resistant to diet-induced obesity (Ravinet
Trillou et al. 2004).
SR141716A also decreases self-administration of various
drugs, such as ethanol (Arnone et al. 1997), nicotine (Cohen
et al. 2002), and heroin (Navarro et al. 2001) and reduces
responding for rewarding brain stimulation (Deroche-
Gamonet et al. 2001). Cohen et al. (2002) also found that
SR141716A reduces the dopamine-releasing properties of
both nicotine and alcohol in the nucleus accumbens. CB
1
receptor knockout mice have been shown to exhibit reduced
ethanol and sucrose consumption (Poncelet et al. 2003).
These and other studies indicate a role for the cannabinoid
system in reward function in the brain. There is also ev-
idence for a role of the cannabinoid system in learning and
memory (Lichtman 2000; reviewed in Robinson and Riedel
2004), sleep/arousal (Mechoulam et al. 1997; Santucci et al.
1996), analgesia (reviewed in Walker and Huang 2002),
movement disorders (reviewed by De Fonseca et al. 1998),
neuroprotection (reviewed in Grundy 2002), and anxiety
(Haller et al. 2004). Some of the aforementioned physio-
logical, psychotropic, and potentially therapeutic actions of
SR141716A have been attributed to its selective effects on
monoamine release in the medial prefrontal cortex (mPFC;
Tzavara et al. 2003), although the level of CB
1
receptor
occupancy in the brain that is required to detect these effects
in animal models and in humans is presently not known.
Although others have measured in vivo CB
1
receptor
occupancy with a derivative of SR141716A, [
131
I]AM281,
relating this occupancy to changes in locomotor activity
(Cosenza et al. 2000), we sought to evaluate central CB
1
receptor occupancy using [
3
H]SR141716A in relation to
physiological and neurochemical changes elicited by leading
CB
1
receptor antagonists that are directly relevant to their
therapeutic potential in the clinic. Specifically, doseoccu-
pancy curves were generated for two orally administered
CB
1
receptor antagonists, SR141716A and SLV319 ((4S)-
(-)-3- (4-chlorophenyl)-N-methyl-N'-[(4-chlorophenyl)sulfo-
nyl] -4-phenyl- 4,5-dihydro -1H-pyrazole-1- carboxamidine)
(Lange et al. 2004). Preclinical studies have shown that these
two compounds exhibit similar in vitro binding affinity for
CB
1
receptors and in vivo CB
1
receptor antagonistic activity
(Lange et al. 2004). Importantly, SR141716A is in Phase III
clinical trials for obesity (see Van Gaal et al. 2005)and
smoking cessation, and SLV319 is in Phase I clinical testing
for obesity and metabolic disorder (Smith and Fathi 2005).
With this in mind, we compared the CB
1
receptor occupancy
in rats required to generate release of the monoamines
dopamine (DA) and norepinephrine (NE) in the mPFC and
changes in feeding behavior (consumption of a highly
palatable diet).
Materials and methods
Drugs
SR141716A and SLV319 were synthesized at Eli Lilly and
Company, Indianapolis, IN, USA. These were administered
by oral gavage in a vehicle comprised of 1% sodium car-
boxymethylcellulose (Hercules, Wilmington, DE, USA),
0.5% sodium lauryl sulfate (Sigma Chemical Co. St. Louis,
MO, USA), 0.085% Povidone K30 (International Specialty
Products, Wayne, NJ, USA), 0.05% Antifoam 1510-US
(Dow Corning, Midland, MI, USA) (all w/w). Both drugs
were prepared fresh and administered as an even and fine
suspension. [
3
H]SR141716A, used as a tracer to determine
in vivo CB
1
receptor occupancy, was purchased from
Amersham Biosciences (Piscataway, NJ, USA) at a specific
activity of 103 mCi/mg.
Animals
Male Wistar rats (240300 g; Harlan SpragueDawley,
Indianapolis, IN, USA, for feeding studies and Charles
Rivers Laboratories, Wilmington, MA, USA, for occupancy
and microdialysis studies) had ad libitum access to normal
rat chow and water unless indicated below. Two different
vendors were used as a source for animals, as each method
followed in the present study was conducted according to
conditions previously established (see below), i.e., for
reasons of continuity and comparison. All studies were
performed in accordance with National Institutes of Health
guidelines under protocols approved by the Animal Care
and Use Committee of Eli Lilly and Company.
Receptor occupancy determinations
Vehicle or CB
1
receptor antagonist test compound was
administered orally (p.o.) at various doses ranging from 0.01
to 30 mg/kg to groups of four rats. Either 1 or 3 h later,
10 μCi [
3
H]SR141716A was administered intravenously
(i.v.) in the lateral tail vein. After an additional hour, rats
were killed by cervical dislocation, and a portion of the
frontal cortex was removed and placed on ice. The frontal
cortex was chosen due to the density of CB
1
receptors in this
area, as well as its relevance to the psychotropic effects of
27
CB
1
receptor antagonists and potentially their therapeutic
applicability (see, e.g., Tzavara et al. 2003; Tataranni et al.
1999). In one doseoccupancy experiment, the brain stem
was also collected in order to examine specific binding in
this brain structure. The tissues were solubilized with 2 ml
Soluene 350 (Perkin Elmer, Wellesley, MA, USA), and
samples were agitated slowly on a shaker table. After ap-
proximately 24 h, 5 ml of scintillation fluid (Ready Solv HP,
Beckman Coulter, Fullerton, CA, USA) was added, and the
samples were shaken for an additional 24 h. Samples were
counted using liquid scintillation spectroscopy, and the
results were expressed as disintegrations per minute per
milligram tissue. Tracer levels in the frontal cortex re-
presented total binding, the sum of specific and nonspecific
binding sites. In order to calculate receptor occupancy, the
level of tracer that represented nonspecific binding needed
to be determined. To accomplish this, we used an intrave-
nous dose of 10 mg/kg unlabeled SR141716A that we had
previously demonstrated, through the generation of i.v. dose
occupancy curves, was well on the doseoccupancy curve
plateau, with higher doses not further suppressing tracer
binding. In each experiment, we included an additional
group of four rats dosed with SR141716A at 10 mg/kg, i.v.,
followed by the tracer 15 min later. The level of tracer in the
cortex of vehicle-pretreated animals represents the sum of
nonspecific and specific binding and is assigned the value of
0% occupancy (all receptors available to the tracer). The
lower level of tracer in animals pretreated with the very high
intravenous dose of SR141716A, the positive control group,
represents the nonspecific binding and is assigned the value
of 100% occupancy (no receptors available to the tracer).
Levels of tracer in the cortex following oral CB
1
receptor
antagonist treatment were interpolated linearly between
these two extremes in order to determine the percent CB
1
receptor occupancy. Data were expressed as means±SEM
(N=4/group) calculated using Prism version 3.0 (GraphPad
Software Inc., San Diego, CA, USA). The ED
50
values were
obtained by fitting the data to a sigmoidal curve using
nonlinear regression. To compare the occupancy curves
generated with the cortex and brain stem, the relative
potency was estimated by simultaneously fitting a four-
parameter logistic equation to the cortex and brain stem
occupancy dose-responses assuming common top, bottom,
and slope parameters using SAS/STAT version 8 (SAS
Institute, Cary, NC, USA). To account for the correlation
between occupancies measured on the same animal, the
multivariate nonlinear regression methods outlined in
Gallant (1987) were used to estimate the correlation and
refit the results with the data transformed to eliminate the
correlation.
Microdialysis studies
Two weeks prior to the microdialysis experiments, rats were
anaesthetized with a mixture of chloral hydrate and
pentobarbital (170 and 36 mg/kg in 30% propylene glycol
and 14% ethanol) and implanted with a guide cannula
(BAS) aimed at the mPFC (AP +3.2, ML +0.6, DV -2.2)
according to Paxinos and Watson (1998). Twenty-four
hours before testing, a 4-mm concentric microdialysis probe
was inserted in the mPFC (BAS, BR-4). Microdialysis ex-
periments were conducted in freely moving animals es-
sentially as described by Tzavara et al. (2003) and briefly
outlined below. The appropriate location of the probes was
verified histologically at the end of the experiment.
On the day of the experiment, a modified Ringers so-
lution (150 mM NaCl, 3 mM KCl, 1.7 mM CaCl
2
, 0.9 mM
MgCl
2
) was perfused at a rate of 1.0 μl/min. Samples were
collected in the presence of an antioxidant (3 mM L-cysteine,
10 mM EDTA) every 30 min into a refrigerated fraction
collector and analyzed the same day with high performance
liquid chromatography (HPLC) coupled to electrochemical
detection as previously described (Perry and Fuller, 1997).
Briefly, catecholamines were separated on a 250×2-mm C-
18 carbon polymer column (BDS Hypersil, ThermoHy-
persil, USA) (mobile phase: 75 mM NaH
2
PO
4
,0.5mM
EDTA, 1.6 mM SOS, 8% CH
3
CN, 75% THF) and detected
on dual carbon electrodes with the potentials set at +680
and + 100 mV. After about 3 h of baseline sampling, vehicle
or drugs were administered p.o., and samples were col-
lected for an additional 4-h period. In this manner, con-
centrations of the catecholamines DA and NE and their
metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC)
and homovanillic acid (HVA), were quantitated in the
dialysate from the mPFC, reflecting their extracellular
concentrations as a result of release or efflux.
Data obtained in response to vehicle and two doses of
each of the compounds were expressed as mean multifold
changes over baseline (±SEM, N=58 rats/group), which is
the average of the three basal values collected immediately
before treatment, and were analyzed with a two-way
ANOVA (dose/treatment × time) followed by a Newman
Keuls multiple comparisons analysis using Statistica soft-
ware (StatSoft, Inc., Tulsa, OK, USA). Data were also
presented as overall (average) mean (±SEM) multifold
changes over baseline and analyzed with one-way ANOVA
followed by the NewmanKeuls test.
Feeding studies
Animals were exposed to a highly palatable, high-fat diet
(HP, 40% fat, 41.3% carbohydrate, 4.25 kcal/g; Teklad,
Madison, WI, USA) at the same time each day for 1 h over a
period of 4 days. Normal rodent chow (Purina 5001, PMI
Nutrition International, Brentwood, MO, USA) was avail-
able at all times. One or three hours prior to HP diet pre-
sentation, rats were dosed p.o. with vehicle on days 1 to 3
and either vehicle, 1, 3, or 10 mg/kg of a CB
1
receptor
antagonist on day 4. The studies were conducted in the light
phase to minimize the impact of nocturnal feeding bouts.
Data were expressed as means (±SEM; N=7 rats/group).
Comparisons between vehicle- and drug-treated groups
28
were performed using one-way ANOVAs followed by
Tukeys test using Prism version 3.0 (GraphPad Software
Inc.), with a pvalue of<0.05 considered significant.
Results
Receptor occupancy determinations
Doseoccupancy curves were generated for the two CB
1
receptor antagonists with either 1- or 3-h pretreatment.
SR141716A administration generated similar curves at both
time points, with absolute ED
50
values of 1.0 mg/kg at 1 h
and 1.1 mg/kg at 3 h (Fig. 1). The doseoccupancy curves
generated for SLV319 were also similar, with absolute ED
50
values of 5.7 and 7.8 mg/kg at 1 and 3 h, respectively
(Fig. 2). The SLV319 occupancy curve generated in the rat
brain stem confirmed the presence of specific binding sites
in this brain structure. A statistical comparison of the curves
obtained simultaneously from cortical and brain stem tissue
demonstrated no significant difference (relative potency of
0.64, 95% CI=0.381.05, p=0.078) (Fig. 3).
Microdialysis studies
Basal dialysate concentrations of either DA or NE in the
various pretreatment groups were pooled, as they were not
statistically different. The overall mean (±SEM) values of
DA and NE were 0.54±0.05 and 1.00±0.10 pmol/ml, re-
spectively. Both CB
1
receptor antagonists increased extra-
cellular DA levels in the mPFC. SR141716A appeared to be
more potent, causing a significant increase following the 3-
and 10-mg/kg doses, while SLV319 only caused a signif-
icant increase at 10 mg/kg (Fig. 4). The time course of the
effects of the two compounds at the 10 mg/kg dose appeared
to be similar, although a more pronounced biphasic effect on
DA release was seen with SLV319, with peak effects at 30
and 180 min (Fig. 4); this may be due to the pharmacokinetic
characteristics of this compound that influence plasma and
brain concentrations, accordingly. Specifically, significant
dose (F
2,14
=12.96, p<0.001), time (F
9,126
=8.30, p<0.001),
and interaction (F
18,126
=2.57, p<0.01) effects were found in
response to SR141716A, and multiple comparisons re-
vealed significant (p<0.05) differences in the effects of each
0.01 0.1
1 3 10 30
-40
-20
0
20
40
60
80
100
120
0.3
SR141716A (mg/kg, p.o.)
% occupancy
Fig. 1 In vivo CB1 receptor
doseoccupancy curves in the
frontal cortex of Wistar rats by
orally administered
SR141716A, as measured with
[3H]SR141716A tracer.
SR141716A was dosed 1 h
(squares)or3h(triangles) prior
to intravenous tracer adminis-
tration. Animals were killed 1 h
after tracer
0.01 0.1 110
-20
0
20
40
60
80
100
120
3300.30.03
SLV319 (mg/kg, p.o.)
% occupancy
Fig. 2 In vivo CB1 receptor
doseoccupancy curves in the
frontal cortex of Wistar rats by
orally administered SLV319, as
measured with [3H]SR141716A
tracer. SLV319 was dosed 1 h
(squares)or3h(triangles) prior
to intravenous tracer adminis-
tration. Animals were killed 1 h
after tracer
29
dose of SR141716A from the effects of vehicle at several
time points (Fig. 4a). In addition, when the data were
expressed as overall effects, a significant (F
2,16
=15.09,
p<0.001) treatment effect was found, as well as significant
(p<0.05) differences of the effect of each dose compared to
the effects of vehicle (Fig. 4c). Similarly, significant dose
(F
2,9
=8.71, p<0.01), time (F
9,81
=3.83, p<0.001), and inter-
action (F
18,81
=3.45, p<0.001) effects were found after
administration of SLV319, and multiple comparisons
revealed significant (p<0.05) differences of each dose of
SVL319 compared to vehicle at specific time points
(Fig. 4b). In addition, when the data were expressed as
0.01 0.1 1 10
-40
-20
0
20
40
60
80
100
3300.30.03
SLV319 (mg/kg, p.o., 1 hr)
% occupancy
Fig. 3 In vivo CB1 receptor
doseoccupancy curves in the
frontal cortex (squares) and
brain stem (triangles) of Wistar
rats by orally administered
SLV319, as measured with [3H]
SR141716A tracer. One hour
after SLV319 administration,
intravenous tracer was injected.
Animals were killed 1 h after
tracer
-90 -60 -30 0 30 60 90 120 150 180 210 240
0.5
1.0
1.5
2.0
vehicle
SR141716A (10 mg/kg)
*
SR141716A (3 mg/kg)
p.o.
*
*
*
*
*
*
*
*
*
Time (min)
DA efflux (fold over baseline)
vehicle
SLV319 (10 mg/kg)
p.o.
*
*
***
*
SLV319 (3 mg/kg)
*
0.50
0.75
1.00
1.25
1.50
1.75
*
*
vehicle SR141716A
(
10 m
g
/k
g)
SR141716A
(
3 m
g
/k
g)
67-74%
88-107%
Overall DA efflux (fold over baseline)
*
11-30%
69%
a
-90 -60 -30 0 30 60 90 120 150 180 210 240
0.5
1.0
1.5
2.0
Time (min)
DA efflux (fold over baseline)
b
c
0.50
0.75
1.00
1.25
1.50
1.75
vehicle
(
10 m
g
/k
g)
SLV319 SLV319
(
3 m
g
/k
g)
Overall DA efflux (fold over baseline)
d
Fig. 4 The CB
1
receptor antagonists SR141716A and SLV319 dose-
dependently increased extracellular DA concentrations (efflux) in the
medial prefrontal cortex of Wistar rats. aand bTime course of DA
changes in the dialysate expressed as multifold change from baseline.
cand dThe average changes from baseline over the entire experiment
(overall effects). SR141716A (aand c) or SLV319 (band d) were
administered orally (p.o., indicated by arrows). Percentages in boxes
represent mean CB
1
receptor occupancy at each dose as determined in
in vivo occupancy experiments. *p<0.05 compared to vehicle
30
overall effects, a significant (F
2,13
=7.69, p<0.01) treatment
effect was found that was due to a significant (p<0.05)
difference of the effect of the 10-mg/kg dose compared to
the effect of vehicle (Fig. 4d). The 10-mg/kg dose of
SR141716A also increased extracellular NE levels in this
brain area, while neither dose of SLV319 caused a sig-
nificant NE increase (Fig. 5). Specifically, significant dose
(F
2,15
=5.76, p<0.05) and time (F
9,135
=3.41, p<0.001)
effects were only found in response to SR141716A; for
this reason, one-way ANOVAs were performed at each time
point, and multiple comparisons thereafter revealed signif-
icant (p<0.05) differences in the effects of the 10-mg/kg
dose of SR141716A from the effects of vehicle at several
time points (Fig. 5a). In addition, when the data were
expressed as overall effects, a significant (F
2,16
=6.20,
p<0.05) treatment effect was found that was due to sig-
nificant (p<0.05) differences of the effect of the 10-mg/kg
dose compared to the effects of vehicle (Fig. 5c). When the
NE data in response to SLV319 were analyzed, only a
significant time effect reached statistical significance
(F
9,90
=2.44, p<0.05), whereas neither dose affected NE
levels (Fig. 5b,d). As previously shown with intraperitoneal
administration (Tzavara et al., 2003), SR141716A adminis-
tered orally significantly increased extracellular concentra-
tions of DOPAC and HVA after both doses, while SLV319
administration only caused a significant increase in these
metabolites after the 10-mg/kg dose (data not shown).
Feeding studies
As can be seen in Fig. 6, prior to antagonist treatment (days 1
to 3), the rats consumed an increasing amount of the HP diet
during their 1 h of exposure. Then on the treatment day
(day 4), there was a significant (p<0.05) treatment effect at
both time points with either SR141716A (F
3,27
=9.08,
pretreatment of 1 h; F
3,27
=8.66, 3 h) or SLV319
(F
3,27
=12.41, 1 h; F
3,27
=6.32, 3 h) resulting in a decrease
in HP diet intake compared to vehicle-treated animals.
Significant (p<0.05) decreases were seen with both CB
1
receptor antagonists at the 3- and 10-mg/kg doses at 1 h
pretreatment, as well as at 3 h pretreatment. As this dif-
ference was not compensated for by an increase in normal
chow consumption, there was a significant (p<0.05) dose
effect resulting in a decrease in total kilocalories per
kilogram (caloric intake) consumed for the 3- and 10-mg/kg
vehicle
SR141716A (10 mg/kg)
SR141716A (3 mg/kg)
p.o.
*
*
*
**
vehicle
SLV319 (10 mg/kg)
p.o.
SLV319 (3 mg/kg)
*
67-74%
88-107%
11-30%
69%
-90 -60 -30 0 30 60 90 120 150 180 210 240
0.5
1.0
1.5
2.5
2.0
Time (min)
NE efflux (fold over baseline)
0.5
1.0
1.5
2.0
vehicle SR141716A
(
10 m
g
/k
g)
SR141716A
(
3 m
g
/k
g)
Overall NE efflux (fold over baseline)
a
-90 -60 -30 0 30 60 90 120 150 180 210 240
0.5
1.0
1.5
2.5
2.0
Time (min)
NE efflux (fold over baseline)
b
c
0.5
1.0
1.5
2.0
vehicle
(
10 m
g
/k
g)
SLV319 SLV319
(
3 m
g
/k
g)
Overall NE efflux (fold over baseline)
d
Fig. 5 SR141716A (10 mg/kg, p.o.) increased extracellular NE
efflux in the medial prefrontal cortex of Wistar rats. aand bTime
course of NE changes in dialysate expressed as multifold change
from baseline. cand dThe average changes from baseline over the
entire experiment (overall effects). Rats were orally administered
(indicated by arrows) SR141716A (aand c) or SLV319 (band d).
Percentages in boxes represent mean CB
1
receptor occupancy at
each dose as determined in in vivo occupancy experiments. *p<0.05
compared to vehicle
31
groups with both compounds at both time points
[SR141716A: 1 h 227±4, 194±7, 137±8 (F
3,27
=42.52) and
3 h 231±7, 203±8, 165±12 (F
3,27
=11.36); SLV319: 1 h 215±
4, 200±3, 171±3 (F
3,27
=29.51) and 3 h 219±8, 194±5, 187±
10 (F
3,27
=6.33) kcal/kg for the vehicle, 3- and 10-mg/kg
groups, respectively].
Discussion
The CB
1
receptor antagonists SR141716A and SLV319
dose-dependently occupied CB
1
receptors in the frontal
cortex of rats after oral administration. There was little
difference in ED
50
values between the 1- and 3-h post-
injection time intervals for either compound, showing that
occupancy was relatively stable within that time frame. One
hour after SLV319 treatment, the only point where brain
stem binding was examined, there was no statistically
significant difference in the receptor occupancy curves gen-
erated simultaneously with the brain stem and the cortex,
despite differences in CB
1
receptor densities between these
two areas (see, e.g., Tsou et al. 1998). The microdialysis data
demonstrated that orally administered SR141716A (at 3 and
10 mg/kg) and SLV319 (at 10 mg/kg) increased extracel-
lular DA concentrations in the mPFC. SR141716A also
increased NE efflux in the mPFC at 10 mg/kg, while none of
the tested doses of SLV319 affected NE concentrations.
Both compounds, administered at 3 and 10 mg/kg, caused a
significant decrease in HP diet consumption at both the 1- and
3-h postinjection intervals. Although it appeared that
SR14716A was more potent than SLV319 in occupying
cortical CB
1
receptors and in increasing cortical release of
monoamines, but not in reducing HP food intake, any claim
for actual differences in in vivo potency of these compounds
would require additional work, taking into consideration
solubility, exposure, and other pharmacokinetic variables.
The occupancy method described here offers certain
methodological advances compared to the study by
Cosenza et al. (2000). Specifically, in the latter study, the
nonspecific binding was estimated using an area thought to
contain very low or no CB
1
receptor density, the brain
stem. We found that the use of the brain stem as a surrogate
0 1 2 3 4
0.0
2.5
5.0
7.5
10.0
12.5
vehicle
SR141716A (1 mg/kg)
SR141716A (3 mg/kg)
SR141716A (10 mg/kg)
*
*
51%
67%
88%
days
grams consumed
vehicle
SR141716A (1 mg/kg)
SR141716A (3 mg/kg)
SR141716A (10 mg/kg)
*
*
48%
74%
107%
vehicle
SLV319 (1 mg/kg)
SLV319 (3 mg/kg)
SLV319 (10 mg/kg)
*
*
12%
30%
69%
vehicle
SLV319 (1 mg/kg)
SLV319 (3 mg/kg)
SLV319 (10 mg/kg)
*
*
3%
11%
69%
a
01234
0.0
2.5
5.0
7.5
10.0
12.5
days
grams consumed
b
01234
0.0
2.5
5.0
7.5
10.0
12.5
da
y
s
grams consumed
c
01234
0.0
2.5
5.0
7.5
10.0
12.5
da
y
s
grams consumed
d
Fig. 6 SR141716A and SLV319 dose-dependently reduced intake
by Wistar rats of highly palatable chow (HP) during 1 h of daily
exposure. Animals were orally administered vehicle prior to HP
presentation on days 13. On day 4, they were dosed with the CB
1
receptor antagonist. aand cData after 1 h pretreatment with
SR141716A (a) or SLV319 (c). band dData after 3 h pretreatment
with SR141716A (b) or SLV319 (d). Percentages in boxes represent
mean CB
1
receptor occupancy at each dose as determined in in vivo
occupancy experiments. *p<0.05 vs vehicle
32
region for nonspecific binding is not appropriate in rats.
Instead, a high dose of a receptor-blocking drug was used
for this purpose (Goeders and Kuhar 1985). This method
of estimating nonspecific binding has the advantage in
that both total and nonspecific binding are determined in
the same brain area, obviating the need for assuming uni-
form nonspecific binding throughout the brain. Employing
this positive control method to determine nonspecific bind-
ing, we generated dose occupancy curves for the CB1
antagonists, SR141716A and SLV319.
As can be seen in Fig. 4,CB
1
receptor occupancy of
approximately 65% was sufficient to increase DA efflux in
the mPFC. This was attained by the 3- and 10-mg/kg doses
of SR141716A and the 10-mg/kg dose of SLV319. When
looking at the NE efflux data (Fig. 5), 65% occupancy was
not sufficient to cause an increase with either antagonist.
Only the 10-mg/kg dose of SR141716A, with occupancy
of 85%, caused an increase in cortical NE release, while no
doses of SLV319 used in the microdialysis experiments
met this level. The reasons why a higher level of CB
1
receptor occupancy is required to elicit changes in NE than
for DA are not clear. It should be noted that the SR
141716A (administered i.p.) mediated neurotransmitter
changes appear to be brain-region-specific (Tzavara et al.
2003), i.e., marked increases were only found in the mPFC
and not in a subcortical dopaminergic area, the nucleus
accumbens, and even if the CB
1
receptor occupancy is
similar in different parts of the brain, depending on the
circuitry involved, the overall functional consequences
may well differ regionally.
The increases in extracellular DA and NE measured in
the mPFC after administration of SR141716A may be
relevant to its apparent effectiveness in regulating food-
and drug-induced motivation and reward and, in turn, to its
therapeutic actions in obesity and smoking cessation. In
this regard, it is noteworthy that neuronal activity within
the prefrontal cortex appears to play an important role in
the regulation of eating behavior and satiety control
(Tataranni et al. 1999). Similar to SR141716A, bupropion,
an atypical antidepressant, increased extracellular DA and
NE in the prefrontal cortex (Li et al. 2002) and was the first
FDA-approved non-nicotine-based pharmacotherapy for
smoking cessation. Its effectiveness has been linked to
decreases in craving (Durcan et al. 2002).
The feeding data showed little difference between the
1- and 3-h time intervals for either CB
1
receptor anta-
gonist, as would be expected from the occupancy data.
There was no difference in potency and efficacy between
the compounds with respect to the HP feeding effect, with
both antagonists reaching significant decreases at 3 mg/kg,
in spite of an apparent difference in cortical CB
1
receptor
occupancy. There are many possible explanations for this.
For example, occupancy in other CB
1
-receptor-containing
areas such as the nucleus accumbens, hippocampus, endo-
penduncular nucleus, and the hypothalamus that are in-
volved in motivational, hedonic, and metabolic aspects of
eating may be different than in the frontal cortex and may
be more directly involved in the feeding effect. However,
CB
1
receptor occupancy curves generated for at least
SLV319 using frontal cortex, an area with relatively high
CB
1
receptor density, and brain stem, with much lower
density, dissected from the same rats were very similar
(see Fig. 3).
The apparent discrepancy could also be due to the
possibility that these antagonists have effects in the
periphery as well. Several lines of evidence for a peripheral
role of CB
1
receptors in feeding exist, including increased
anandamide in small intestine but not brain of rats with
food deprivation and loss of hyperphagic and hypophagic
responses of anandamide and SR141716A, respectively,
after deafferentation by capsaicin (Gomez et al. 2002). It
was recently shown that SR141716A reduced levels of
plasma ghrelin (Cani et al. 2004). As ghrelin stimulates
appetite, CB
1
receptor antagonists may work in part by
decreasing ghrelin production in the periphery. We mea-
sured central CB
1
receptor occupancy, which is dependent
on brain penetration. SLV319 may have higher peripheral
occupancy, while SR141716A may have higher brain
penetration and therefore higher central occupancy.
Additionally, this incongruity between central CB
1
recep-
tor occupancy and potency in the feeding studies may be
due to other actions of SLV319 not shared with
SR141716A. The regulation of feeding is extremely
complex, with multiple signals and pathways involved.
SLV319 may hypothetically interact with another regula-
tory pathway, in addition to its actions on the cannabinoid
system, leading to a larger decrease in consumption than
that anticipated by its CB
1
receptor occupancy alone.
Nevertheless, it should be emphasized that, if anything,
there is evidence in support of the notion that SR141716A
has off-target, non-CB
1
-receptor-related activities (see,
e.g., Breivogel et al. 2001; Monory et al. 2002), although
the significance of these findings in food intake regulation
is not readily apparent.
While occupancy measurements have previously been
reported using [
131
I]AM281 (Cosenza et al. 2000), we have
described here a more advantageous in vivo central CB
1
receptor occupancy method, this one using commercially
available [
3
H]SR141716A. Employing this method, we
demonstrated that SR141716A and SLV319 occupy cor-
tical CB
1
receptors consistently between 1 and 3 h after
dosing. They are both able to increase extracellular DA
levels in the frontal cortex at CB
1
receptor occupancies
exceeding 65%. SR141716A also increases extracellular
NE levels in this brain region when occupancy exceeds
85%. Both compounds are effective at decreasing intake of
a highly palatable diet, although the effectiveness of
SLV319 at such low CB
1
receptor occupancies may sug-
gest it exerts at least some of its effects through mech-
anisms other than central CB
1
receptor occupancy.
References
Abel E (1971) Effects of marihuana on the solution of anagrams,
memory, and appetite. Nature 231:260261
33
Arnone M, Maruani J, Chaperon F, Thiebot M, Poncelet MH,
Soubrie P, Le Fur G (1997) Selective inhibition of sucrose and
ethanol intake by SR141716, an antagonist of central canna-
binoid (CB1) receptors. Psychopharmacology (Berl) 132:104
106
Breivogel CS, Griffin G, Di Marzo V, Martin BR (2001) Evidence
for a new G protein-coupled cannabinoid receptor in mouse
brain. Mol Pharmacol 60:155163
Cani P, Montoya M, Neyrinck A, Delzenne N, Lambert D (2004)
Potential modulation of plasma ghrelin and glucagon-like
peptide-1 by anorexigenic cannabinoid compounds, SR14171
6A (rimonabant) and oleoylethanolamide. Br J Nutr 92:757
761
Cohen C, Perrault G, Voltz C, Steinberg R, Soubrie P (2002)
SR141716, a central cannabinoid (CB1) receptor antagonist,
blocks the motivational and dopamine-releasing effects of
nicotine in rats. Behav Pharmacol 13:451463
Cosenza M, Gifford A, Gatley S, Pyatt B, Liu Q, Makriyannis A,
Volkow N (2000) Locomotor activity and occupancy of brain
cannabinoid CB1 receptors by the antagonist/inverse agonist
AM281. Synapse 38:477482
De Fronseca F, Del Arco I, Martin-Calderon J, Gorriti M, Navarro
M (1998) Role of the endogenous cannabinoid system in the
regulation of motor activity. Neurobiol Dis 5:483501
Deroche-Gamonet V, Le Moal M, Piazza P, Soubrie P (2001)
SR141716, a CB1 receptor antagonist, decreases the sensitivity
to the reinforcing effects of electrical brain stimulation in rats.
Psychopharmacology (Berl) 157:254259
Devane W, Dysarc F, Johnson M, Mevin L, Howlett A (1988)
Determination and characterization of a cannabinoid receptor in
rat brain. Mol Pharmacol 34:605613
Devane W, Hanus L, Breuer A, Pertwee R, Stevenson L, Griffin G,
Gibson D, Mandelbaum A, Etinger A, Mechoulam R (1992)
Isolation and structure of a brain constituent that binds to the
cannabinoid receptor. Science 258:19461949
Di Marzo V, Goparaju S, Wang L, Liu J, Batkai S, Jarai Z, Fezza F,
Miura G, Palmiter R, Sugiura T, Kunos G (2001) Leptin-
regulated endocannabinoids are involved in maintaining food
intake. Nature 410:822825
Durcan M, Denner G, White J, Johnston J, Gonzales D, Niaura R,
Rigotti N, Sachs D (2002) The effect of bupropion sustained-
release on cigarette craving after smoking cessation. Clin Ther
24:540551
Gallant A (1987) Nonlinear statistical models. Wiley, New York
Gaoni Y, Mechoulam R (1964) Isolation, structure and partial
synthesis of an active component of hashish. J Am Chem Soc
86:16461647
Goeders N, Kuhar M (1985) Benzodiazepine receptor binding in
vivo with [3H]-Ro 15-1788. Life Sci 37:345355
Gomez R, Navarro M, Ferrer B, Trigo J, Bilbao A, Del Arco I,
Cippitelli A, Nava F, Piomelli D, Rodriguez de Fonseca F
(2002) A peripheral mechanism for CB1 cannabinoid receptor-
dependent modulation of feeding. J Neurosci 22:96129617
Grundy R (2002) The therapeutic potential of the cannabinoids in
neuroprotection. Expert Opin Investig Drugs 11:13651374
Haller J, Varga B, Ledent C, Freund T (2004) CB1 cannabinoid
receptors mediate anxiolytic effects: convergent genetic and
pharmacological evidence with CB1-specific agents. Behav
Pharmacol 15:299304
Lange J, Coolen H, van Stuivenberg H, Dijksman J, Herremans A,
Ronken E, Keizer H, Hiskias G, Tipker K, McCreary A,
Veerman W, Wals H, Stork B, Verveer P, den Hartog A, de Jong
N, Adolfs T, Hoogendoorn J, Kruse C (2004) Synthesis,
biological properties, and molecular modeling investigations of
novel 3,4-diarylpyrazolines as potent and selective CB1
cannabinoid receptor antagonists. J Med Chem 47:627643
Li S, Perry K, Wong D (2002) Influence of fluoxetine on the ability
of bupropion to modulate extracellular dopamine and norepi-
nephrine concentrations in three mesocorticolimbic areas of
rats. Neuropharmacology 42:181190
Lichtman A (2000) SR141716A enhances spatial memory as as-
sessed in a radial-arm maze task in rats. Eur J Pharmacol
404:175179
Matsuda L, Lolait S, Brownstein M, Young A, Bonner T (1990)
Structure of cannabinoid receptor and functional expression of
the cloned cDNA. Nature 346:561564
Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski N
et al (1995) Identification of an endogenous 2-monoglyceride,
present in canine gut, that binds to cannabinoid receptors.
Biochem Pharmacol 50:8390
Mechoulam R, Fride E, Hanus L, Sheskin T, Bisogno T, Di Marzo
V, Bayewitch M, Vogel Z (1997) Anandamide may mediate
sleep induction. Nature 389:2526
Monory K, Tzavara ET, Lexime J, Ledent C, Parmentier M, Borsodi
A, Hanoune J (2002) Novel, not adenylyl cyclase-coupled
cannabinoid binding site in cerebellum of mice. Biochem
Biophys Res Commun 292:231235
Munro S, Thomas K, Abu-Shaar M (1993) Molecular characteriza-
tion of a peripheral receptor for cannabinoids. Nature 365:61
65
Navarro M, Carrera M, Fratta W, Valverde O, Cossu G, Fattore L,
Chowen J, Gomez R, Del Arco I, Villanua M, Maldonado R,
Koub G, Rodriguez de Foncesca F (2001) Functional interac-
tion between opioid and cannabinoid receptors in drug self-
administration. J Neurosci 21:53445350
Nunez E, Benito C, Pazos M, Barbachano A, Fajardo O, Gonzalez
S, Tolon R, Romero J (2004) Cannabinoid CB2 receptors are
expressed by perivascular microglial cells in the human brain:
an immunohistochemical study. Synapse 53:208213
Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates.
Academic, San Diego
Perry K, Fuller R (1997) Fluoxetine increases norepinephrine
release in rat hypothalamus as measured by tissue levels of
MHPG-SO4 and microdialysis in conscious rats. J Neural
Transm 104:953966
Plasse T, Gorter R, Krasnow S, Lane M, Shepard K, Wadleigh R
(1991) Recent clinical experience with dronabinol. Pharmacol
Biochem Behav 40:695700
Poncelet M, Maruani J, Calassi R, Soubrie P (2003) Overeating,
alcohol and sucrose consumption decrease in CB1 receptor
deleted mice. Neurosci Lett 343:216218
Ravinet Trillou C, Delgorge C, Menet C, Arnone M, Soubrie P
(2004) CB1 cannabinoid receptor knockout in mice leads to
leanness, resistance to diet-induced obesity and enhanced leptin
sensitivity. Int J Obes 28:640648
Rinaldi-Carmona M, Barth F, Heaulme M, Shire D, Calandra B et al
(1994) SR141716A, a potent and selective antagonist of the
brain cannabinoid receptor. FEBS Lett 350:240244
Robinson L, Riedel G (2004) Cannabinoid function in spatial
learning: an update. Curr Neuropharmacol 2:125143
Santucci V, Storme J, Soubrie P, Le Fur G (1996) Arousal-enhancing
properties of the CB1 cannabinoid receptor antagonist SR
141716A in rats as assessed by electroencephalographic spectral
and sleep-waking cycle analysis. Life Sci 58:PL103110
Simiand J, Keane M, Keane PE, Soubrie P (1998) SR141716, a CB1
cannabinoid receptor antagonist, selectively reduces sweet food
intake in marmoset. Behav Pharmacol 9:179181
Smith R, Fathi Z (2005) Recent advances in the research and
development of CB(1) antagonists. IDrugs 8:5366
Tataranni P, Gautier JF, Chen K, Uecker A, Bandy D, Salbe A,
Pratley R, Lawson M, Reiman E, Ravussin E (1999) Neuro-
anatomical correlates of hunger and satiation in humans using
positron emission tomography. Proc Natl Acad Sci U S A 96:
45694574
34
Tsou K, Brown S, Sanudo-Pena M, Mackie K, Walker J (1998)
Immunohistochemical distribution of cannabinoid CB1 recep-
tors in the rat central nervous system. Neuroscience 83:393
411
Tzavara E, Davis R, Perry K, Li X, Salhoff C, Bymaster F, Witkin J,
Nomikos GG (2003) The CB1 receptor antagonist SR141716A
selectively increases monoaminergic neurotransmission in the
medial prefrontal cortex: implications for therapeutic actions.
Br J Pharmacol 138:544553
Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rössner S (2005)
Effects of the cannabinoid-1 receptor blocker rimonabant on
weight reduction and cardiovascular risk factors in overweight
patients: 1-year experience from the RIO-Europe study. Lancet
365:13891397
Verty A, McGregor I, Mallet P (2004) Consumption of high
carbohydrate, high fat, and normal chow is equally suppressed
by a cannabinoid receptor antagonist in non-deprived rats.
Neurosci Lett 354:217220
Walker M, Huang S (2002) Cannabinoid analgesia. Pharmacol Ther
95:127135
35
... In an effort to eliminate these adverse effects, a peripherally restricted CB1R inverse agonist, JD5037, was developed [8]. JD5037 was effective in decreasing body weight, indicating that JD5037 functions via peripheral CB1Rs, rather than having central action [16]. In this study, we showed that in Mdr2 −/− mice, JD5037 decreased body weight gains with unclear underlying mechanisms but that could be a result of reduced food intake or increased energy expenditure, as shown in diet-induced obesity mice [8]. ...
Article
Full-text available
Previous research highlighted the involvement of the cannabinoid CB1 receptor in regulating the physiology of hepatocytes and hepatic stellate cells. The inhibition of the CB1 receptor via peripherally restricted CB1 receptor inverse agonist JD5037 has shown promise in inhibiting liver fibrosis in mice treated with CCl4. However, its efficacy in phospholipid transporter-deficiency-induced liver fibrosis remains uncertain. In this study, we investigated the effectiveness of JD5037 in Mdr2−/− mice. Mdr2 (Abcb4) is a mouse ortholog of the human MDR3 (ABCB4) gene encoding for the canalicular phospholipid transporter. Genetic disruption of the Mdr2 gene in mice causes a complete absence of phosphatidylcholine from bile, leading to liver injury and fibrosis. Mdr2−/− mice develop spontaneous fibrosis during growth. JD5037 was orally administered to the mice for four weeks starting at eight weeks of age. Liver fibrosis, bile acid levels, inflammation, and injury were assessed. Additionally, JD5037 was administered to three-week-old mice to evaluate its preventive effects on fibrosis development. Our findings corroborate previous observations regarding global CB1 receptor inverse agonists. Four weeks of JD5037 treatment in eight-week-old Mdr2−/− mice with established fibrosis led to reduced body weight gains. However, contrary to expectations, JD5037 significantly exacerbated liver injury, evidenced by elevated serum ALT and ALP levels and exacerbated liver histology. Notably, JD5037-treated Mdr2−/− mice exhibited significantly heightened serum bile acid levels. Furthermore, JD5037 treatment intensified liver fibrosis, increased fibrogenic gene expression, stimulated ductular reaction, and upregulated hepatic proinflammatory cytokines. Importantly, JD5037 failed to prevent liver fibrosis formation in three-week-old Mdr2−/− mice. In summary, our study reveals the exacerbating effect of JD5037 on liver fibrosis in genetically MDR2-deficient mice. These findings underscore the need for caution in the use of peripherally restricted CB1R inverse agonists for liver fibrosis treatment, particularly in cases of dysfunctional hepatic phospholipid transporter.
... Among the most common adverse reactions in the taranabant-treated groups compared with placebo-treated groups were gastrointestinal disorders (diarrhea, nausea, and vomiting), nervous system disorders (dizziness and headache), psychiatric disorders (including depression and anxiety), and irritability [221][222][223]225]. Ibipinabant is another first-generation antagonist/inverse agonist of CB1Rs; it appears to be as effective as rimonabant in decreasing the intake of a highly palatable diet in Wistar rats, but with a lower degree of occupancy of brain CB1Rs [228], suggesting that part of the anti-obesity effect of CB1R antagonists/inverse agonists is due to their action on peripheral receptors [229]. Ibipinabant has been included in Phase II (NCT00388609) and Phase III (NCT00541567) clinical trials to study its efficacy in obese or overweight patients with comorbidities and obese or overweight T2DM patients, respectively. ...
Article
Full-text available
Recently, research has greatly expanded the knowledge of the endocannabinoid system (ECS) and its involvement in several therapeutic applications. Cannabinoid receptors (CBRs) are present in nearly every mammalian tissue, performing a vital role in different physiological processes (neuronal development, immune modulation, energy homeostasis). The ECS has an essential role in metabolic control and lipid signaling, making it a potential target for managing conditions such as obesity and diabetes. Its malfunction is closely linked to these pathological conditions. Additionally, the immunomodulatory function of the ECS presents a promising avenue for developing new treatments for various types of acute and chronic inflammatory conditions. Preclinical investigations using peripherally restricted CBR antagonists that do not cross the BBB have shown promise for the treatment of obesity and metabolic diseases, highlighting the importance of continuing efforts to discover novel molecules with superior safety profiles. The purpose of this review is to examine the roles of CB1R and CB2Rs, as well as their antagonists, in relation to the above-mentioned disorders.
... Rimonabant alone had only modest effects on responding across the dose range tested, with similar effects in both young and aged rats. We selected this range based on evidence that the highest dose of rimonabant tested occupies over 65% of CB1 receptors, which is sufficient to affect feeding-related behavior, but mitigates the likelihood nonselective effects at higher doses of rimonabant (Need et al., 2006). There was some evidence that higher doses of rimonabant began to produce some disruption. ...
Article
Cannabinoid use has increased among aging individuals. However, little information on age-related differences in the behavioral effects of these agents is available. To explore potential differences in the behavioral effects of cannabinoids, we determined effects of Δ9-tetrahydrocannabinol (THC, 1-10 mg/kg) or rimonabant (0.3-3.2 mg/kg) on operant fixed-ratio responding (FR10) for food in young adult (6 months) and aged (29 months) rats. THC dose-dependently decreased responding for food. Rimonabant alone had little or no effect on responding up to 1.0 mg/kg, but disrupted responding following a 3.2 mg/kg dose. Rimonabant (1.0 mg/kg) partially antagonized response disruption by THC. These effects were similar in young adult and aged rats. However, aging has been reported to change the neurobiology of cannabinoid CB1 receptors. To confirm our rats exhibited such differences, we assessed CB1 receptor binding sites and function in six subcortical (caudate, nucleus accumbens CA1, and CA2/CA3), and three cortical regions (medial prefrontal, temporal, entorhinal) in young adult (6 months) or aged (26 months) male Lewis rats using quantitative autoradiography. CB1 receptor binding sites were reduced in cortical, but not subcortical brain regions of aged rats. CB1 receptor function, at the level of receptor-G protein interaction, was not different in any region studied. Results indicate that down-regulation of CB1 receptor binding sites observed in cortical regions of aged rats was not accompanied by a commensurate decrease in CB1 receptor-stimulated [35S]GTPγS binding, suggesting a compensatory increase in receptor function in cortical areas. Together, our results provide additional evidence of age-related changes in central CB1 receptor populations. However, the functional compensation for decreased CB1 receptor binding may mitigate changes in behavioral effects of cannabinoids. With the rising use of cannabinoid-based therapeutics among aging populations, further evaluation of age-related changes in the cannabinoid system and the impact of these changes on effects of this class of drugs is warranted.
... MRI-1891 Is a β-Arrestin-2-Biased Peripheral CB 1 R Antagonist. We modified the structure of the brain penetrant CB 1 R antagonist/inverse agonist ibipinabant 16 in order to reduce its ability to cross the blood/brain barrier. The structurally modified compound (S)-MRI-1891 (referred to as MRI-1891) has increased total polar surface area and hydrogen bonding capacity relative to ibipinabant, predicting reduced Figure 1. ...
Article
Full-text available
Seven-transmembrane receptors signal via G-protein- and β-arrestin-dependent pathways. We describe a peripheral CB1R antagonist (MRI-1891) highly biased toward inhibiting CB1R-induced β-arrestin-2 (βArr2) recruitment over G-protein activation. In obese wild-type and βArr2-knockout (KO) mice, MRI-1891 treatment reduces food intake and body weight without eliciting anxiety even at a high dose causing partial brain CB1R occupancy. By contrast, the unbiased global CB1R antagonist rimonabant elicits anxiety in both strains, indicating no βArr2 involvement. Interestingly, obesity-induced muscle insulin resistance is improved by MRI-1891 in wild-type but not in βArr2-KO mice. In C2C12 myoblasts, CB1R activation suppresses insulin-induced akt-2 phosphorylation, preventable by MRI-1891, βArr2 knockdown or overexpression of CB1R-interacting protein. MRI-1891, but not rimonabant, interacts with nonpolar residues on the N-terminal loop, including F108, and on transmembrane helix-1, including S123, a combination that facilitates βArr2 bias. Thus, CB1R promotes muscle insulin resistance via βArr2 signaling, selectively mitigated by a biased CB1R antagonist at reduced risk of central nervous system (CNS) side effects.
... While this effect of WIN55-212,2 was reversed by rimonabant, NA efflux was unaffected by a lower dose of this CB1R antagonist when given alone (Oropeza et al., 2005;Page et al., 2008). However, other studies have shown that higher doses of rimonabant also increase prefrontal NA release (Need et al., 2006;Tzavara et al., 2003). There are several possible explanations for why both CB1R agonism and antagonism increase NA release in the mPFC. ...
Article
Full-text available
Anxiety and trauma‐related disorders, such as post‐traumatic stress disorder (PTSD), are debilitating mental illnesses with great personal and socioeconomic costs. Examining memory formation and relevant behavioural responding associated with aversive stimuli may improve our understanding of the neurobiology underlying fear memory processing and PTSD treatment. The neurocircuitry underpinning learned fear and its inhibition through extinction is complex, involving synergistic interactions between different neurotransmitter systems in inter‐connected brain areas. Endocannabinoid and noradrenergic transmission have both been implicated separately in fear memory processing and PTSD but potential interactions between these systems in relation to fear extinction have received little attention to date. Their receptors are expressed together in brain areas crucial for fear extinction, which is enhanced by both cannabinoid and noradrenergic receptor activation in these areas. Moreover, cannabinoid signalling modulates the activity of locus coeruleus noradrenaline neurons and the release of noradrenaline in the medial prefrontal cortex, a brain area that is crucial for fear extinction. Interestingly, endocannabinoid‐noradrenergic system interactions have been shown to regulate the encoding and retrieval of fear memory. Thus noradrenergic regulation of fear extinction may also be driven indirectly in part via cannabinoid receptor signalling. In this perspective paper, we collate the available relevant literature and propose a synergistic role for the endocannabinoid and noradrenergic systems in regulating fear extinction, the study of which may further our understanding of the neurobiological substrates of PTSD and its treatment.
... Ibipinabant is a potent and highly selective CB1 antagonist [64] that was found to significantly decrease feeding in animal models [65]. One study noted a significant side effect of myotoxicity in beagle dogs [66]. ...
... However, CB1 receptors also regulate the release of these neurotransmitters (Fig. 3). For example, rimonabant enhances the concentration of serotonin, norepinephrine and dopamine in the PFC (Tzavara et al. 2003;Need et al. 2006), while fluoxetine reduces CB1 receptor expression in rodents (Oliva et al. 2005). Increased levels of serotonin exerted by fluoxetine appear to have an impact on CB1 receptor expression, signifying a probable role of communication involving serotonin and the ECS in the regulation of mood and cognition. ...
Article
Full-text available
Depression is a common mental disorder and is the leading cause of suicide globally. Because of the significant diversity in mental disorders, accurate diagnosis is difficult. Hence, the investigation of novel biomarkers is a key research perspective in psychotherapy to enable an individually tailored treatment approach. The prefrontal cortex (PFC) is a vital cortical region whose circuitry has been implicated in the development of depressive disorder. The endocannabinoid system (ECS) has garnered increasing attention because of its involvement in several diverse physiological brain processes including regulation of emotional, motivational and cognitive functions. The current review article explores the function of the key elements of the ECS as a biomarker in depressive disorder. The activity of endocannabinoids is thought to be moderated by the CB1 receptors in the central nervous system (CNS). Variations in the concentration of endocannabinoids and the binding affinity of CB1 receptors and their density have been identified in the PFC of persons with depression. Such discoveries support our theory that alteration in endocannabinoid function leads to the pathophysiological features of depressive disorders. Moreover, evidence from animal and human studies has revealed that dysfunction in endocannabinoid signalling can produce depression-like behaviours; therefore, improvement of endocannabinoid signalling may represent a new therapeutic approach for the management of depressive disorders. Graphical abstract
... Because SLV319 is 3-4 times less potent than rimonabant (Lange et al. 2004), it was administered I.P. at a dose of 15 mg kg −1 . Following the systemic administration of this dose, SLV319 has been shown to occupy >80% of brain CB1 receptors (Need et al. 2006). At an ambient temperature of 22 • C, rats received the I.P. pretreatment with SLV319 or its vehicle and 90 min later were injected I.V. with LPS or saline. ...
Article
Hypothermia occurs in the most severe cases of systemic inflammation, but the mechanisms involved are poorly understood. This study evaluated whether the hypothermic response to bacterial lipopolysaccharide (LPS) is modulated by the endocannabinoid anandamide(AEA) and its receptors: cannabinoid-1 (CB1), cannabinoid-2 (CB2) and transient receptor potential vanilloid-1 (TRPV1). In rats exposed to an ambient temperature of 22◦C, a moderate dose of LPS (25 - 100 μg kg−1 I.V.) induced a fall in body temperature with a nadir at ∼100 minpostinjection. This response was not affected by desensitization of intra-abdominal TRPV1 receptors with resiniferatoxin (20 μg kg - 1 I.P.), by systemic TRPV1 antagonism with capsazepine(40mg kg−1 I.P.), or by systemic CB2 receptor antagonism with SR144528 (1.4 mg kg−1 I.P.).However, CB1 receptor antagonism by rimonabant (4.6mg kg−1 I.P.) or SLV319 (15mg kg−1 I.P.)blocked LPS hypothermia. The effect of rimonabant was further studied. Rimonabant blocked LPS hypothermia when administered I.C.V. at a dose (4.6 μg) that was too low to produce systemic effects. The blockade of LPS hypothermia by I.C.V. rimonabant was associated with suppression of the circulating level of tumour necrosis factor-α. In contrast to rimonabant,the I.C.V. administration of AEA (50 μg) enhanced LPS hypothermia. Importantly, I.C.V. AEAdid not evoke hypothermia in rats not treated with LPS, thus indicating that AEA modulates LPS-activated pathways in the brain rather than thermo effector pathways. In conclusion, the present study reveals a novel, critical role of brain CB1 receptors in LPS hypothermia. Brain CB1 receptors may constitute a new therapeutic target in systemic inflammation and sepsis.
Article
Monoamine oxidase (MAO) enzymes, type A and B metabolise the amine neurotransmitters of the body. Selective inhibition of either enzyme is an approach for treating neurodegenerative and stress-induced disorders, and inhibition of an enzyme is proportional to the binding of the MAO inhibitor. Conventionally, the binding of test compounds to enzymes is assessed by radiolabelled ligands in ex vivo and in vivo occupancy assays. Regulatory restrictions and turnaround time are the limitations of the methods that use radiolabelled ligands. But the use of non-radiolabelled tracers and sensitive mass spectrometry (LC-MS/MS) based assays accelerated the determination of target occupancy in pre-clinical species. A report on use of non-radiolabelled ligand in in vivo MAO occupancy assay is not available. The objectives of the present study were to optimise non-radiolabelled harmine and deprenyl as selective tracers in MAO-A and MAO-B occupancy assays and evaluate MAO occupancy of test compounds in rat brain. Tracer optimisation resulted in a detectable, stable, and low ratio (< 3.0) of tracer concentrations between any two brain tissues. In occupancy assay, tracer was intravenously administered (10 μg/kg, harmine or 60 μg/kg, L-deprenyl) after the treatment with test compound (clorgyline or tranylcypromine or pargyline or phenelzine or thioperamide). Specific brain tissues were isolated at a defined interval and tracer concentrations were quantified using LC-MS/MS method. Pre-treatment with MAO inhibitors resulted in a decrease (maximum, 80-85%) in harmine or an increase (maximum, 85-300%) in L-deprenyl concentrations. But we considered the change in tracer concentration, relative to the vehicle and positive control groups to calculate MAO occupancy. The observed selectivity and ratio of occupancies (ED50) of test compound towards MAO-A and MAO-B are comparable with the results from in vitro radiolabelled ligand-based inhibition assay. The results demonstrated the application of these non-radiolabelled tracers as suitable pre-clinical tools to determine MAO occupancy.
Article
Full-text available
Endocannabinoids acting via CB1 receptors (CB1R) play a critical role in regulating energy homeostasis, which was the rationale for the pharmaceutical development of CB1R antagonists for the treatment of obesity. Although the first-in-class CB1R antagonist rimonabant proved to be effective in mitigating obesity and its multiple cardiometabolic complications, it was withdrawn from clinical use due to CNS-mediated neuropsychiatric side effects, which halted the further therapeutic development of the whole class of these compounds. Compared to the brain, CB1Rs are expressed at low yet functional levels in peripheral organs involved in regulating energy homeostasis, including liver, skeletal muscle, adipose tissue and endocrine pancreas. In recent preclinical studies, selective targeting of these receptors by 'second generation' peripherally restricted CB1R antagonists replicated the metabolic benefits of rimonabant in rodent models of obesity and diabetes without causing CNS-mediated side effects. Increased CB1R activity also contributes to complex, multifactorial disorders such as various forms of tissue fibrosis, treatment of which may benefit from simultaneous engagement of more than one therapeutic target. Accordingly, novel 'third generation' hybrid inhibitors of peripheral CB1R and inducible NO synthase were tested in mouse models of liver and pulmonary fibrosis where their antifibrotic efficacy was found to exceed the efficacy of drugs that inhibit only one of these targets. In this review, we will discuss the challenges and opportunities offered by second and third generation CB1R antagonists and their potential therapeutic uses.
Article
Full-text available
The central role of the hypothalamus in the origination and/or processing of feeding-related stimuli may be modulated by the activity of other functional areas of the brain including the insular cortex (involved in enteroceptive monitoring) and the prefrontal cortex (involved in the inhibition of inappropriate response tendencies). Regional cerebral blood flow (rCBF), a marker of neuronal activity, was measured in 11 healthy, normal-weight men by using positron emission tomography in a state of hunger (after 36-h fast) and a state of satiation (after a liquid meal). Hunger was associated with significantly increased rCBF in the vicinity of the hypothalamus and insular cortex and in additional paralimbic and limbic areas (orbitofrontal cortex, anterior cingulate cortex, and parahippocampal and hippocampal formation), thalamus, caudate, precuneus, putamen, and cerebellum. Satiation was associated with increased rCBF in the vicinity of the ventromedial prefrontal cortex, dorsolateral prefrontal cortex, and inferior parietal lobule. Changes in plasma insulin concentrations in response to the meal were negatively correlated with changes in rCBF in the insular and orbitofrontal cortex. Changes in plasma free fatty acid concentrations in response to the meal were negatively correlated with changes in rCBF in the anterior cingulate and positively correlated with changes in rCBF in the dorsolateral prefrontal cortex. In conclusion, these findings raise the possibility that several regions of the brain participate in the regulation of hunger and satiation and that insulin and free fatty acids may be metabolic modulators of postprandial brain neuronal events. Although exploratory, the present study provides a foundation for investigating the human brain regions and cognitive operations that respond to nutritional stimuli.
Article
Immunohistochemical distribution of cannabinoid receptors in the adult rat brain was studied using specific purified antibodies against the amino-terminus of the CB1 receptor. Our results generally agree well with the previous studies using CB1 receptor autoradiography and messenger RNA in situ hybridization. However, because of its greater resolution, immunohistochemistry allowed identification of particular neuronal cells and fibers that possess cannabinoid receptors. CB1-like immunoreactivity was found in axons, cell bodies and dendrites, where it appeared as puncta in somata and processes. Both intensely and moderately or lightly stained neurons were observed. The intensely stained neurons were dispersed and only occur in cortical structures including hippocampal formation and olfactory bulb. Moderately or lightly stained neurons were found in caudate-putamen and amygdala. In the hippocampal formation only intensely stained neurons were observed. The cell bodies of pyramidal neurons in CA1 and CA3 fields appeared to be unstained but surrounded by a dense plexus of immunoreactive fibers. The granule cells in the dentate area were also immunonegative. Many intensely stained neurons were located at the base of the granule cell layer. CB1-like immunoreactive neurons and fibers were also found in the somatosensory, cingulate, perirhinal, entorhinal and piriform cortices, in claustrum, amygdaloid nuclei, nucleus accumbens and septum. Beaded immunoreactive fibers were detected in periaqueductal gray, nucleus tractus solitarius, spinal trigeminal tract and nucleus, dorsal horn and lamina X of the spinal cord. A triangular cap-like mass of immunoreactivity was found to surround the basal part of the Purkinje cell body in the cerebellum. Only small, lightly stained cells were found in the molecular layer in the cerebellum close to the Purkinje cell layer. The CB1 receptor is widely distributed in the forebrain and has a more restricted distribution in the hindbrain and the spinal cord. It appears to be expressed on cell bodies, dendrites and axons. According to the location and morphology, many, but not all, CB1-like immunoreactive neurons appear to be GABAergic. Therefore, cannabinoids and cannabinoid receptors may play a role in modulating GABAergic neurons.
Article
It has long been known that cannabinoids can alter perception and cognitive function. In this review article, we first discuss the current knowledge of both plant-derived and synthetic cannabinoid effects on learning and memory formation in animals with a particular emphasis on spatial mapping. It appears that stimulation of the cannabinoid receptors in the brain is detrimental to spatial learning, and recent advances suggest that working memory is particularly vunerable and in some cases the involvement of the cannabinoid system in memory consolidation. Most of these effects are mediated by CB1 receptors, and blockade of these receptors can, under certain conditions enhance learning. In the second part, we investigate whether cannabinoid effects can be explained in terms of modulation of other transmitter systems. Since CB1 is G-protein-coupled and predominantly presynaptically localised, the primary effect may lie in a down / up-regulation of other transmitters, resulting in memory deficits. Such an analysis proved useful as it revealed that modulation of the noradrenergic and cholinergic system is readily able to explain deficits reported in memory consolidation, while cholinergic, dopaminergic, and GABAergic regulation may explain working memory deficits.
Article
The goals of this study were to examine the relationship between intravenous doses of the cannabinoid CB1 receptor antagonist AM281 (N-(morpholin-4-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide) and the degree of occupancy of this receptor, and to relate occupancy to the ability of this compound to antagonize the sedative effects of the cannabinoid receptor agonist WIN 55,212-2. Occupancy was determined by measuring the ability of intravenous doses of AM281 to inhibit in vivo binding of [131I]AM281 in brain areas, and locomotor activity was assessed by measuring the rate of beam crossings in a photocell apparatus. As previously documented, WIN 55,212-2 (1 mg/kg, i.v.) significantly reduced locomotor activity at early times after administration. Co-injection of AM281 (0.3 mg/kg i/v) and WIN 55,212-2 restored the rate of beam crossings to that seen on injection of vehicle. In addition, AM281 (0.3 mg/kg i/v) approximately doubled locomotor activity between 60–120 min when injected alone. The IC50 value for displacement of [131I]AM281 by AM281 was 0.45 mg/kg. These observations confirm earlier indications that AM281 is a CB1 receptor antagonist or inverse agonist and suggest the existence of an endogenous cannabinoid tone that moderates exploratory locomotor activity. Synapse 38:477–482, 2000. © 2000 Wiley-Liss, Inc.