ArticlePDF Available

Evidence that Behavioral Phenotypes of Morphine in β-arr2−/− Mice Are Due to the Unmasking of JNK Signaling

Authors:

Abstract and Figures

The altered behavioral effects of morphine, but not most other mu agonists, in mice lacking β-arrestin 2, suggest that this scaffolding protein regulates the signaling cascade of this commonly used analgesic. One of the cascades that could be regulated by β-arrestin 2 is cJun-N-terminal kinase (JNK), which binds with β-arrestin 2 and modulates the analgesic effects of morphine. Using neurons lacking β-arrestin 2 (β-arr2-/-) to examine this interaction, we found that β-arr2-/- neurons show altered intracellular distribution of JNK and cJun, and that morphine, but not fentanyl, increased the nuclear localization of the phosphorylated, therefore activated, form of cJun, a JNK target in dorsal root ganglia neurons. This suggests that deleting β-arrestin 2 affects the JNK cascade. We therefore examined whether some of the behavioral phenotypes of mice lacking β-arrestin 2 could be a result of altered JNK signaling. Indeed, two different JNK inhibitors reversed the enhanced analgesic effect of morphine, a known phenotype of β-arr2-/- mice, to +/+ levels. Both the reduced locomotor effect of morphine and the psychomotor sensitization to repeated morphine administration in β-arr2-/- mice were also returned to +/+ levels by inhibiting JNK. In contrast, the behavioral effects of fentanyl were neither genotype-dependent nor affected by JNK inhibition. Furthermore, a PKC inhibitor had a similar effect as inhibiting JNK in reducing the enhanced analgesic effect of morphine in β-arr2-/- mice to +/+ levels. In summary, removing β-arrestin 2 reveals mu receptor activation of the JNK cascade in a ligand-specific manner explaining several behavioral phenotypes of β-arr2-/- mice.
Content may be subject to copyright.
Evidence that Behavioral Phenotypes of Morphine in
b-arr2/Mice Are Due to the Unmasking of JNK Signaling
Nitish Mittal
1,2
, Miao Tan
2
, Onyemachi Egbuta
2
, Nina Desai
2
, Cynthia Crawford
1
, Cui-Wei Xie
2
,
Christopher Evans
2
and Wendy Walwyn*
,2
1
Department of Psychology, California State University, San Bernardino, CA, USA;
2
Department of Psychiatry and Biobehavioral Sciences, Stefan
Hatos Center for Neuropharmacology, Semel Institute, University of California, Los Angeles, CA, USA
The altered behavioral effects of morphine, but not most other mu agonists, in mice lacking b-arrestin 2, suggest that this scaffolding
protein regulates the signaling cascade of this commonly used analgesic. One of the cascades that could be regulated by b-arrestin 2 is
cJun-N-terminal kinase (JNK), which binds with b-arrestin 2 and modulates the analgesic effects of morphine. Using neurons lacking
b-arrestin 2 (b-arr2/) to examine this interaction, we found that b-arr2/neurons show altered intracellular distribution of JNK and
cJun, and that morphine, but not fentanyl, increased the nuclear localization of the phosphorylated, therefore activated, form of cJun, a
JNK target in dorsal root ganglia neurons. This suggests that deleting b-arrestin 2 affects the JNK cascade. We therefore examined
whether some of the behavioral phenotypes of mice lacking b-arrestin 2 could be a result of altered JNK signaling. Indeed, two different
JNK inhibitors reversed the enhanced analgesic effect of morphine, a known phenotype of b-arr2/mice, to + / + levels. Both the
reduced locomotor effect of morphine and the psychomotor sensitization to repeated morphine administration in b-arr2/mice were
also returned to + /+ levels by inhibiting JNK. In contrast, the behavioral effects of fentanyl were neither genotype-dependent nor
affected by JNK inhibition. Furthermore, a PKC inhibitor had a similar effect as inhibiting JNK in reducing the enhanced analgesic effect of
morphine in b-arr2/mice to + / + levels. In summary, removing b-arrestin 2 reveals mu receptor activation of the JNK cascade in a
ligand-specific manner explaining several behavioral phenotypes of b-arr2/mice.
Neuropsychopharmacology (2012) 37, 1953–1962; doi:10.1038/npp.2012.42; published online 11 April 2012
Keywords: mu opioid receptor; JNK; b-arrestin 2; morphine; analgesia; locomotion
INTRODUCTION
One of the many examples that the ubiquitously expressed
arrestins serve as more than terminators of receptor
signaling through initiating receptor internalization (Groer
et al, 2011; Shenoy and Lefkowitz, 2011; Shukla et al, 2011)
is the phenotype of mice lacking b-arrestin 2 (b-arr2/;
Bohn et al, 1999, 2002; Raehal and Bohn, 2011). As
morphine does not induce significant internalization of the
mu opioid receptor, the effects of morphine in b-arr2/
mice cannot be explained by this prototypical role of b-
arrestin 2. However, some progress has recently been made
in revealing several arrestin-dependent, yet internalization-
independent, signaling pathways of the mu opioid receptor
that explain some, but not all, effects of morphine in mice
lacking b-arrestin 2 (Arttamangkul et al, 2008; Dang et al,
2011; Quillinan et al, 2011; Walwyn et al, 2007).
Observations of ligand-specific signaling, or the ability of
different ligands to activate specific effectors, were first
made in the 1980s (Gee and Yamamura, 1983), but only
recently have become an accepted dimension of G-protein-
coupled receptor (GPCR) signaling (Reiter et al, 2012;
Urban et al, 2007). Such functional selectivity is believed to
result from the ability of different agonists of the same
GPCR to induce ligand-specific conformational changes of
the receptor. These distinct conformations recruit different
proteins to the agonist-bound receptor and activate down-
stream signaling cascades in an agonist-specific manner
(Galandrin et al, 2008). The opioid family of GPCRs are no
exception to such diversity; ligands of the mu, delta, and
kappa opioid receptors are known to activate functionally
distinct signaling cascades (Bruchas and Chavkin, 2010;
Melief et al, 2010; Peng et al, 2009; Pradhan et al, 2010).
The enhanced analgesic effect of morphine but not many
other mu agonists in mice lacking b-arrestin 2 suggests that
morphine, in activating a signaling cascade that is regulated
by b-arrestin 2, differs from other agonists (Bohn et al,
1999, 2002). As b-arrestin 2 is a multi-functional scaffolding
Received 13 September 2011; revised 29 February 2012; accepted 5
March 2012
*Correspondence: Dr W Walwyn, Department of Psychiatry and
Biobehavioral Sciences, Stefan Hatos Center for Neuropharmacology,
Semel Institute, University of California, Los Angeles, CA 90095, USA,
Tel: + 1 310 206 3231, Fax: + 1 310 825 7067,
E-mail: wwalwyn@ucla.edu
Neuropsychopharmacology (2012) 37, 1953 1962
&
2012 American College of Neuropsychopharmacology. All rights reserved 0893-133X/12
www.neuropsychopharmacology.org
and signal transduction protein (Xiao et al, 2007, 2010), the
signaling cascade responsible for such ligand specificity
remains unknown. However, the arrestins bind with, and
regulate, many components of the map kinase cascade, one
of which is the stress-activated protein kinase, cJun-N-
terminal kinase (JNK), a kinase that has recently been shown
to play an important role in ligand-directed signaling of the
mu and kappa opioid receptors (Melief et al, 2010).
As JNK binds with b-arrestin 2, this scaffolding protein
may modulate mu receptor activation of the JNK cascade
in a ligand-specific manner. Using mice lacking b-arrestin 2
to determine how this scaffolding protein regulates mu
receptor signaling, we have found that altered JNK activity
explains some of the behavioral phenotypes of mice lacking
b-arrestin 2.
MATERIALS AND METHODS
Animals
b-Arrestin 1 (also known as arrestin 2) or b-arrestin 2 (also
known as arrestin 3) mice, fully back-crossed into the C57Bl/
6 background, were kindly supplied by Dr Lefkowitz (HHMI,
University of North Carolina, Chapel Hill, NC). The
behavioral experiments used equal numbers of adult male
and female b-arrestin (b-arr) 1/2 + / + and /mice that
were 81±10 days old, 22.3±3.7g in weight and bred from
heterozygous matings. For the cellular experiments, early
postnatal b-arr2 + / + and /pups were obtained from
homozygous matings, one generation from heterozygous
breeders. All animal experiments were conducted in accor-
dance with the Guide for the Care and Use of Laboratory
Animals and followed institutionally approved animal care
and use protocols.
SDS–PAGE and Western Blot Analysis of JNK and
Phospho-JNK Proteins Levels
Dorsal root ganglia (DRGs) were collected from all spinal
levels of either b-arr2 + / + or /adult mice, weighed and
homogenized in the following lysis buffer; 20 mM Tris–HCl
pH 7.4, 0.32 M sucrose, 1 mM EDTA, 1 mM EGTA, 50 mM
NaF, 1 mM Na
4
P
2
O
7
,1Halt Protease and Phosphatase
Inhibitor Cocktail (Pierce, Thermo Scientific, Rockford, IL),
and protein content determined (BCA, Pierce). Protein
samples (50 mg/sample) were separated by SDS–PAGE (10%
NuPAGE gel; Invitrogen, Carlsbad, CA) and transferred
to an Immobilon PVDF membrane (0.45 mm; Millipore,
Billerica, MA). After several washes in TBS and a 120 min
blocking step (Casein/0.1% Tween-20, Thermo Scientific),
the membrane was incubated in anti-JNK or anti-phospho-
JNK at 1 : 100 (Thr183/Tyr185), from both Cell Signaling
(Danvers, MA) or glyceraldehyde 3-phosphate dehydrogen-
ase (GAPDH; 2.5 10
2
mg/ml, Thermo Scientific), o/n at
41C. The membrane was then washed in TBS with 0.1%
Tween-20, incubated in Stabilized Peroxidase-conjugated
secondary antibodies, anti-rabbit IgG for JNK (1 : 100) or
anti-mouse IgG for phospho-JNK (1 :100), or GAPDH
(1 : 10 000) for 120 min at RT (Pierce, Rockford, IL). The
labeled protein bands were visualized by chemilumines-
cence (Super signal Chemiluminescent Substrate, Pierce),
and signal intensity (pixels/mm
2
) quantified (ImageJ, NIH).
For each sample, the signal intensity of JNK or phospho-
JNK was normalized to that of GAPDH. Statistical analysis:
The mean of the three experiments for each of the six
samples are expressed as the mean±SEM and the differences
between groups determined by one-way ANOVA (Analyse-it
Software, Leeds, UK) with significance accepted at po0.05.
Cell Culture
DRGs from all spinal levels were harvested from early postnatal
(p0–1) b-arr2+/+ or /pups. After chemical dissociation
in trypsin (Invitrogen) and physical dissociation by trituration,
110
4
cells were plated on poly-D-lysine (Sigma, St Louis,
MO) and laminin (Invitrogen) coated coverslips of MatTek
dishes (MatTek, Ashland, MA) as previously described
(Walwyn et al, 2007). The cells were maintained for 24–48 h
in Neurobasal/B27 media containing NGFs (10 ng/ml) in vitro
after which the cells were treated and fixed for immunocy-
tochemistry or used to determine mu agonist inhibition of
voltage-dependent Ca
2+
channels (VDCCs).
Immunocytochemistry
b-arr2 + / + or /cells remained untreated or were
treated with morphine (NIDA) or fentanyl (Sigma) for
10 min, washed and fixed. After a series of washes, a 0.3%
Triton X100 permeabilization step, and a 10% serum
blocking step, anti-JNK3 (1 : 500, Santa Cruz Biotechnology,
Santa Cruz, CA) or anti-phosphorylated JNK (anti-phos-
pho-JNK: 1 : 100, Cell Signaling) were added. In a second,
matching, set of plates, primary antibodies against cJun
(1 : 500, Santa Cruz Biotechnology) and phosphorylated
cJun (anti-phospho-cJun, 1 : 250, Santa Cruz Biotechnology)
were added. After o/n incubation and further washes,
Alexa-555-conjugated donkey anti-goat/mouse (1 : 1000 for
phospho-cJun and phospho-JNK; Invitrogen) and Alexa-
488-conjugated donkey anti-rabbit/goat (1 : 1000 for cJun and
1 : 400 for JNK; Invitrogen) were added to each plate for
90 min at RT. After a final series of washes, the cells were
mounted in Prolong containing 40,6-diamidino-2-phenylin-
dole (DAPI; Invitrogen). Medium-sized DRGs (20–30 mM)
were visualized by a 60 oil immersion objective on a
Nikon TE2000 microscope and images were captured by a
Retiga 1300Exi CCD camera (QImaging, British Columbia,
Canada). All cells were imaged at the mid-nuclear level
using DAPI as a guide. Pixel intensity per unit area for each
of the three channels (RGB) was quantified by IPLab
(version 3.6.5, BioVision Technologies, Exton, PA). Statis-
tical analysis: Data from untreated cells were expressed as
mean±SEM of the nuclear/cytosolic signal intensity for
4100 cells/sample. The effect of different mu agonists on
cJun and phospho-cJun localization was determined by
comparing the percent change of the nuclear:cytosolic ratio
in the agonist treated vs untreated cells. Differences between
groups were determined using two-way ANOVA between
groups (Analyse-it Software) and significance accepted at
po0.05.
Analgesia
Thermal nociception was measured by the warm-water tail-
immersion assay in which the latency to remove the tail
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1954
Neuropsychopharmacology
from a 49.5 1C water bath was measured. After a basal
response was obtained (average of three to four tests), one
of the JNK or PKC inhibitors, or matching vehicle, was
injected, another basal measurement was obtained 30 (PKC
inhibitor) or 60 (JNK inhibitor) min later and then
morphine or fentanyl was administered. The JNK inhibitors
used were: SP600125 (SP6; Anthra[1,9-cd]pyrazol-6(2H)-
one, 1,9-pyrazoloanthrone, 30 mg/kg i.p., EMD Chemicals,
Merck KGaA, Darmstadt, Germany) or BI78D3 (4-(2,3-
Dihydrobenzo[b][1,4]dioxin-6-yl)-5-(5-nitrothiazol-2-ylthio)
-4H-1,2,4-triazol-3-ol) 25 mg/kg i.p., EMD Chemicals). Both
compounds inhibit the activation of all JNK isoforms by
binding with either the ATP docking site (SP6; Shin et al,
2002) or the D-domain consensus sequence in JNK to inhibit
the docking of upstream kinases (BI78D3; Stebbins et al,
2008). The PKC inhibitor used, Bisindolylmaleimide VIII
(Ro-31-7549, EMD Chemicals), inhibits PKCa,bI, bII, g,e
(Wilkinson et al,1993).Statistical analysis: Data are
expressed as the response in seconds (s), and as the mean±
SEM of each group. Differences between groups were deter-
mined by one- or two-way ANOVA with repeated measures
and factorial analysis at each time interval using Stat View
(v5). Significance was accepted at po0.05.
Locomotion
Locomotor activity was measured in 16.7 12.7 cm
2
boxes by
an infrared video-camera and Ethovision video tracking
software (v7.1; Noldus, Tacoma). The mice were habituated
for 15 min on the first day and, on each of the following 3
days, were given SP6 (30 mg/kg i.p.), or vehicle, 60 min before
morphine (10mg/kg s.c.) or fentanyl (0.2 mg/kg s.c.) and
placed in the locomotor boxes. Locomotion was assessed
over the following 30 min. Statistical analysis: Data are
expressed as mean±SEM and differences between groups
determined by one- or two-way ANOVA with repeated mea-
sures and factorial analysis at each time interval (Stat View).
Electrophysiology
Voltage-dependent Ca
2+
currents were recorded from
small–medium-sized DRG neurons 20–30 mmindiameter
under whole-cell voltage-clamp conditions as previously
described (Tan et al, 2003; Walwyn et al, 2007). Cells were
perfused with an external solution containing 10 mM
CaCl
2
, 130 mM tetraethylammonium chloride, 5 mM
HEPES, 25 mM d-glucose and 0.25 mMtetrodotoxinatpH
7.35. The patch electrode was filled with an internal
solution composed of 105 mM CsCl, 40 mM HEPES, 5 mM
d-glucose, 2.5 mM MgCl
2
, 10 mM EGTA, 2 mM Mg-ATP,
and 0.5 mM GTP at pH 7.2. Ca
2+
currents were evoked
every 10 s by 40-ms voltage steps from 80 to + 10 mV
using an Axopatch 200B patch-clamp amplifier. Capaci-
tance and series resistance were corrected with the
compensation circuitry on the amplifier. Series resistance
was compensated by 80–90%. Leak currents were sub-
tracted using a P/6 protocol. Recorded signals were
acquiredandanalyzedusingAxonpClampv8.0or9.0
software (Axon Instruments, Foster City, CA). Drug
application and desensitization protocols: The JNK in-
hibitor, SP6, was dissolved in DMSO as a 10-mM stock
solution and diluted with the culture medium to a final
concentration of 10 mM for application. The effect of SP6
on mu receptor–VDCC inhibition was assessed by a 4-h
pre-incubation with SP6 (10 mM). For the desensitization
experiments, b-arr2 + / + and /DRG neurons were
pre-incubated with 1 mM morphine, D-Ala
2
-MePhe
4
-Glyol
5
Enkephalin (DAMGO), or fentanyl, in the presence or
absence of 10 mM SP6 for 4 h, the media removed, and the
cells were washed by an B100 solution exchange before
being challenged by the full agonist, DAMGO (1 mM).
Statistical analysis: Mean Ca
2+
current amplitudes were
measured (pCLAMP 9.0; Axon Instruments, CA) between 5
and 10 ms after initiating the depolarizing step. Mean
current amplitudes were then plotted against time. Stable
recordings were fitted by a linear function to compare, by
extrapolation, control current amplitude to the current amplitude
recorded in the presence of opioid receptor agonists or SP6.
Data are expressed as mean±SEM and were compared using
ANOVA with a post hoc Tukey’s test with significance accepted
at po0.05. Recordings that exhibited marked rundown
(430%) were discarded as were significant outliers, five out
of the B300 + recordings (Prism v5.03).
RESULTS
DRG Neurons Lacking b-Arrestin 2 Show Altered JNK
and cJun Localization
JNK and phospho-JNK expression. As b-arrestin 2 facil-
itates GPCR activation of the JNK cascade (Willoughby
and Collins, 2005), genetically deleting b-arrestin 2 may
result in a compensatory adaptation in the expression
of JNK and phospho-JNK. This was examined by
SDS–PAGE and western blotting in b-arr2/and + / +
adult DRGs. No effect of genotype was found. The relative
intensities of the protein of interest normalized to that
of GAPDH were: JNK; 48 kDa: b-arr2 + / + ; 0.39±0.05,
b-arr2/: 0.33±0.05.54 kDa: b-arr2 + / + ; 0.23±0.06,
b-arr2/: 0.16±0.04. phospho-JNK; 54 kDa: b-arr2 + / + ;
0.61±0.13, b-arr2/: 0.64±0.04.
The basal intracellular location of JNK and cJun. As b-
arrestin 2 contains a nuclear export sequence resulting in its
constitutive exclusion from the nucleus (Song et al, 2006),
b-arr2/neurons may show altered nuclear or cytosolic
localization of proteins, such as JNK, that bind with b-
arrestin 2. We therefore examined the effect of deleting b-
arrestin 2 on the relative nuclear:cytosolic ratio of JNK,
phospho-JNK, and the major JNK target, cJun, and
phospho-cJun. b-arr2/DRG neurons showed a basal
increase in the nuclear localization of JNK, phospho-JNK,
and cJun, but not phospho-cJun (Figure 1b).
Ligand-induced changes in phospho-cJun location. We
next examined whether two mu agonists, morphine and
fentanyl, previously shown to activate the JNK cascade
(Melief et al, 2010) could alter the intracellular location of
phospho-cJun in b-arr2 + / + and /neurons. We focused
on phospho-cJun for a number of reasons; the activity of
this terminal kinase of the JNK pathway is finely regulated
by JNK (Hibi et al, 1993) and, importantly, the basal
location of phospho-cJun was unaffected by deleting
b-arrestin 2. We found that morphine increased the relative
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1955
Neuropsychopharmacology
intensity of phospho-cJun in the nucleus of b-arr2/but
had no effect in + / + neurons. Fentanyl did not alter
phospho-cJun localization in + / + or /neurons
(Figure 1c). Neither ligand altered the nuclear:cytosolic
ratio of cJun (data not shown).
JNK Inhibition Reverses Behavioral Phenotypes of
Morphine in b-arr2/Mice
Using DRG neurons as a model, we have found altered basal
and mu agonist-induced changes in the intracellular
location of JNK and/or cJun, suggesting that deleting
b-arrestin 2 alters aspects of the JNK cascade. As this
cascade can modulate the behavioral effects of mu agonists
(Chen et al, 2008; Melief et al, 2010), we next examined
whether the behavioral phenotypes of b-arr2/mice
could be a result of altered JNK activity.
Analgesia. The role of JNK in thermal analgesia was
examined by administering one of two JNK inhibitors, SP6
or BI78D3, to b-arr2/and + / + mice, 60 min before
morphine (5 and 10 mg/kg s.c.) or fentanyl (0.2 mg/kg s.c.).
JNK
1.00
1.05
1.10
1.15
1.20 β-arr2+/+
β-arr2–/–
i cJun p-cJunp-JNK
a
ii iii iv
β-arr2+/+
β-arr2–/–
*
β-arr2+/+
β-arr2–/–
1.00
1.10
1.20
1.30
1.40
1.50
1.60 β-arr2+/+
β-arr2–/–
*
*
b
pcJun
cJun DAPI Merge
β-arr2–/–β-arr2+/+ β-arr2+/+ β-arr2–/– β-arr2–/–β-arr2+/+ β-arr2+/+ β-arr2–/–
Nuclear/Cytosolic
iii
cJun p-cJun
Cytosol 563 506
Nucleus 707 860
1.00
1.05
1.10
1.15
1.20
Nuclear/Cytosolic
Nuclear/Cytosolic
Nuclear/Cytosolic
1.00
1.05
1.10
1.15
1.20
1.25
***
85
90
95
100
105
110
β-Arr2–/–
110
β-Arr2+/+
85
90
95
100
105
Morphine
Fentanyl
i
c
ii
Morphine
Fentanyl
Nuclear/Cytosolic
(% untreated)
p-cJun
Nuclear/Cytosolic
(% untreated)
Figure 1 DRG neurons as a model to examine the JNK cascade. DRG neurons from early postnatal mice were used to examine the effect of deleting b-
arrestin 2 and different mu receptor agonists on the relative nuclear:cytosolic ratio of JNK and cJun. (a) The relative pixel intensity/unit area of the nuclear or
cytosolic labeling for JNK, phospho-JNK, cJun, or phospho-cJun was imaged and quantified in individual neurons. An example of the labeling and quantification
for one cell are shown in (ai, aii), respectively. (b) This method was used to quantify the nuclear:cytosolic ratio of JNK, phospho-JNK, cJun, and phospho-cJun,
in untreated b-arr2 + / + vs /neurons (n4100 neurons/sample). b-arr2/neurons showed a relative increase in the nuclear localization of JNK,
phospho-JNK, and cJun, (*po0.05 vs b-arr2 +/+), but not phospho-cJun (p¼0.75). (c) While morphine treatment (1 mM, 10 min) did not affect the
nuclear:cytosolic ratio of phospho-cJun in b-arr2 + / + neurons, this mu agonist did increase phospho-cJun in the nucleus vs cytosol of b-arr2/neurons.
.***po0.001 vs untreated. Scale bars ¼10 mm.
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1956
Neuropsychopharmacology
Thermal analgesia was assessed by the warm-water tail-
immersion assay 20–30 min later and at regular intervals
thereafter. In b-arr2 + / + mice, neither of the JNK
inhibitors altered the analgesic profile of morphine, which
showed a comparable dose-response curve to previous
publications (Sora et al, 1997; Bohn et al, 2000). In contrast,
both SP6 and BI78D3, reversed the enhanced analgesic
effect of morphine in b-arr2/mice to wild-type levels
(Figure 2a and b). The analgesia induced by fentanyl was
neither genotype-dependent nor influenced by JNK. In
addition, the higher basal analgesia of b-arr2/
(2.19±0.05 s) vs + / + (2.03±0.05 s, po0.05) mice was
not affected by either SP6 or BI78D3. As PKC has been
shown to activate JNK (Lopez-Bergami et al, 2005; Melief
et al, 2010), we next examined whether the effect of
morphine in b-arr2/mice was also PKC-dependent. The
PKC inhibitor, Bisindolylmaleimide VIII, reversed the
enhanced effect of morphine in b-arr2/mice to wild-
type levels (Figure 2c). We also examined whether
b-arrestin 1 may be responsible for the effect of JNK in
b-arr2/mice but found no effect of deleting b-arrestin
1 or inhibiting JNK in the analgesic response to morphine
in these mice (Figure 2e).
Psychomotor effects of morphine
Acute locomotion:Another of the behavioral phenotypes
of b-arr2/mice is the reduced locomotor effect of
morphine (Bohn et al, 2003; Urs et al, 2011). We
hypothesized that, similar to the analgesic effect of
morphine, this may also be due to altered JNK activity.
The JNK inhibitor, SP6, or vehicle, was therefore adminis-
tered 60 min before assessing the locomotor effects of
morphine or fentanyl in b-arr2 + / + and /mice. SP6
had no effect in b-arr2 + / + mice, but SP6-treated /
mice showed an enhanced locomotor response to morphine
matching that of b-arr2 + / + mice (Figure 3a). By contrast,
the locomotor effect of fentanyl was neither genotype nor
SP6-dependent (Figure 3b).
Psychomotor sensitization:As the locomotor effect of a
single morphine injection was influenced by both genotype
and JNK inhibition, locomotor sensitization induced by
repeated morphine administration could similarly be
modulated by JNK in b-arr2/mice. We found that 3
days of repeated morphine administration resulted in less
locomotor sensitization in b-arr2/than + / + mice
(10 mg/kg; Figure 4a). This was reversed by SP6, adminis-
tered 60 min before morphine on each of the 3 test days
(Figure 4b). The locomotor sensitization induced by
repeated fentanyl administration was not influenced by
deleting b-arrestin 2 or inhibiting JNK (Figure 4c and d).
Possible cellular targets of the JNK cascade. Inhibiting JNK
reduced the analgesic efficacy of morphine 30 min after this
drug was administered. This suggests that JNK phosphor-
ylation of cytosolic targets, rather than the transcriptional
activity of cJun, modulates morphine analgesia. We there-
fore examined whether opioid coupling with VDCCs, a
Gbg-mediated second messenger cascade, may be influ-
enced by JNK in a ligand-specific manner. We examined
both the acute effect of inhibiting JNK on mu receptor–
VDCC coupling as well as desensitization of opioid
a
d
b
e
Morphine (10 mg/kg)
i
2
4
6
8
10
12
14
0
–60 0 30 60 90 120
Time (min)
Vehicle
SP6
BI78D3
ii
2
4
6
8
10
12
14
0
–60 0 30 60 90 120
Time (min)
Vehicle
SP6
BI78D3
i
0
2
4
Time (s)
Time (s)
Time (s)
Time (s)
Time (s)
Time (s)
Time (s)
Time (s)
Time (s)
Time (s)
6
8
10
12
–60 0 30 60 90 120
Time (min)
Morphine (5 mg/kg)
β-arrestin 2 +/+
β-arrestin 1+/+ β-arrestin 1–/–
β-arrestin 2–/ –
Vehicle
SP6
ii
0
2
4
6
8
10
12
–60 0 30 60 90 120
Time (min)
Vehicle
SP6
i
0
2
4
6
8
–60 0 20 40 60 80
Time (min)
Fentanyl (0.2 mg/kg)
Vehicle
SP6
ii
0
2
4
6
8
–60 0 20 40 60 80
Time (min)
Vehicle
SP6
i
0
2
4
6
8
10
12
–60 0 30 60 90 120
Time (min)
Morphine (10 mg/kg)
Vehicle
SP6
0
2
4
6
8
10
12
–60 0 30 60 90 120
Time (min)
ii
Vehicle
SP6
cii
2
4
6
8
10
12
14
0
–30 0 30 60 90 120
Time (min)
Vehicle
Bis-8
i
2
4
6
8
10
12
14
0
–30 0 30 60 90 120
Time (min)
Vehicle
Bis-8
Morphine (10 mg/kg)
Figure 2 The enhanced thermal analgesic effect of morphine in b-arr2/
mice is reduced to + / + levels by inhibiting JNK or PKC. The warm-water
(49.5 1C) tail-immersion assay was used to assess the analgesic effects of
different mu agonists in b-Arr2 + / + and /mice and the resultant data are
presented as the response (s; yaxis) over time (min; xaxis). (a, b) b-Arr2/
mice showed a dose-dependent increase in the thermal analgesic effects of
morphine ((a) 5 and (b) 10 mg/kg morphine s.c., 5 mg/kg: p¼0.12 vs b-arr2 + /
+, F
1,5
¼1.773; 10 mg/kg: po0.001 vs b-arr2+/+, F
1,5
13.227), which was
reversed by the JNK inhibitor, SP6 (aii) 5 mg/kg: po0.05 vs vehicle, F
1,5
¼2.422;
(bii) 10 mg/kg: po0.01 vs vehicle, F
1,5
¼4.501). BI78D3 (bii), similarly reversed
the enhanced analgesia seen in morphine-treated (10 mg/kg s.c.) b-arr2/
mice (po0.001 vs vehicle, F
1,5
¼6.142). Neither JNK inhibitor altered the b-arr2
+ / + response ((ai) 5 mg/kg: p¼0.06 vs vehicle, F
1,5
¼10.74; (aii) 10 mg/kg:
SP6:p¼0.94 vs vehicle, F
1,5
¼0.457; BI78D3: p¼0.71 vs vehicle, F
1,5
¼0.575)
(c) The PKC inhibitor Bisindolylmaleimide VIII (10 mg/kg i.p.) also reversed the
enhanced analgesia seen after morphine administration (10 mg/kg s.c.) in b-
arr2/mice (po0.001 vs vehicle, F
1,5
¼7.099), while having no effect in b-
arr2 + / + mice (p¼0.76 vs vehicle, F
1,5
¼0.937). (d) Mice treated with
fentanyl (0.2 mg/kg s.c.) showed no effect of the b-arrestin 2 deletion
(p¼0.84 vs b-arr2 + / + , F
1,5
¼0.403) or JNK inhibition (b-arr2 + / + :
p¼0.59 vs vehicle, F
1,5
¼0.748; b-arr2/:p¼0.96 vs vehicle, F
1,5
¼0.194).
(e) There was also no effect of deleting b-arrestin 1 (p¼0.10 vs b-arr2 + / + ,
F
1,5
¼1.883) or JNK inhibition on the morphine response of b-arr1/or
+/+ mice (SP6:b-arr1 + / + : p¼0.07 vs vehicle, F
1,5
¼2.089; b-arr1/:
p¼0.80 vs vehicle, F
1,5
¼0.224). Injection 1 ¼JNK or PKC inhibitor, or
matching vehicle. Injection 2 ¼morphine or fentanyl.
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1957
Neuropsychopharmacology
receptor–VDCC inhibition following 4 h of opiate pre-
treatment. We found no effect of SP6 pre-treatment on the
acute VDCC inhibition induced by morphine, fentanyl or
DAMGO in either b-arr2 + / + or /neurons (Table 1).
However, desensitization induced by each of the ligands
revealed both genotype and JNK effects, as shown by a
1-mM test dose of DAMGO. In b-arr2 + / + neurons, the
desensitization induced by morphine (4 h, 1 mM) was
prevented by SP6 co-incubation. In b-arr2/neurons,
morphine did not induce significant desensitization, and
this was unaffected by SP6 co-incubation. Pre-incubation
with either DAMGO or fentanyl resulted in significant
desensitization in both b-arr2 + / + and /neurons that
was not altered by SP6.
DISCUSSION
When compared with other mu agonists, morphine induces
less internalization of the mu opioid receptor (Borgland
et al, 2003; McPherson et al, 2010; Zheng et al, 2011). This
suggests that the prototypical role of b-arrestin 2 in
initiating receptor internalization does not explain how
the effects of morphine, but not of other mu agonists, are
altered in mice lacking b-arrestin 2. Here, we demonstrate
that morphine activation of JNK in the absence of b-arrestin
2 explains ligand-specific behavioral phenotypes of b-
arr2/mice. These findings also demonstrate that the
JNK cascade may be activated in a ligand-specific manner,
providing further evidence of the role of this member of the
map kinase pathway in mediating ligand-directed signaling
of the mu receptor.
Compared with other mu agonists such as fentanyl, the
relative inability of morphine to cause significant receptor
phosphorylation, results in the recruitment of PKC to
morphine, but not fentanyl, bound mu receptors (Zheng
et al, 2011). As PKC is known to both scaffold with and
phosphorylate JNK (Lopez-Bergami et al, 2005; Ping, 2003;
Pontrelli et al, 2004), we propose that in wild-type mice,
morphine recruitment of PKC activates JNK but that b-
arrestin 2 recruitment limits further JNK activation. How-
ever, if b-arrestin 2 is absent and is coupled with the
inability of morphine to recruit b-arrestin 1 (Groer et al,
2011), sufficient PKC recruitment results in sustained
activation of the JNK cascade in b-arr2/neurons to
affect analgesia. In contrast, other mu agonists, such as
fentanyl differ from morphine as they are able to
phosphorylate the mu receptor and recruit either b-arrestin
1 or 2 (Groer et al, 2011; Zheng et al, 2011). In the absence
of b-arrestin 2, these agonists recruit b-arrestin 1, PKC
is not recruited and the JNK cascade is not significantly
activated.
Time (min)
Morphine 10 mg/kg
a
0
1000
2000
3000
4000
5000
6000
Total distance traveled (cm)
Vehicle
SP6
Fentanyl 0.2 mg/kg
b
0
500
1000
1500
2000
2500
5 1015202530
Distance traveled (cm)
Time (min)
0
2000
4000
6000
8000
10000
12000
14000
Total distance traveled (cm)
Vehicle
SP6
i
ii
β-arrestin 2 +/+
β-arrestin 2 /
0
200
400
600
800
1000
1200
1400
1600
5 1015202530
Distance traveled (cm)
Vehicle
SP6
ii
0
200
400
600
800
1000
1200
1400
1600
5 1015202530
Distance traveled (cm)
Vehicle
SP6
β-arrestin 2 /
βarr2+/+ βarr2/
0
500
1000
1500
2000
2500
5 1015202530
Distance traveled (cm)
iβ-arrestin 2 +/+
Vehicle
SP6
Vehicle
SP6
βarr2+/+ βarr2/
Time (min)
Time (min)
*
iii
iii
Figure 3 The reduced locomotor effect of morphine in b-arr2/mice is returned to + / + levels by inhibiting JNK. The locomotor effect of different
mu agonists in b-arr2 + / + and /mice is shown over the duration of the 30-min test in the line graphs with distance traveled (cm; yaxis) shown over
time (xaxis). The total distance traveled (cm; yaxis) is shown in the accompanying bar graph. (a) The locomotor effect of morphine was less in b-Arr2/
mice compared with their wild-type littermates (po0.01 vs b-arr2 + / + , F
1,11
¼14.251). SP6 (30 mg/kg i.p.) increased the effect of morphine in b-arr2/
mice (po0.001 vs vehicle, F
1,11
¼19.835) to + /+ levels (p¼0.52 vs b-arr2 + / + , F
1,14
¼0.429) but had no effect in b-arr2 + / + mice (p¼0.46 vs vehicle,
F
1,14
¼0.577). (b) Mice treated with fentanyl (0.2 mg/kg s.c.) showed no effect of genotype (p¼0.81 vs b-arr2 + / + , F
1,9
¼0.061) or JNK inhibition (SP6:b-
arr2 + / + : p¼0.68 vs vehicle, F
1,12
¼0.169; b-arr2/:p¼0.16 vs Vehicle, F
1,11
¼2.194).
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1958
Neuropsychopharmacology
i
a
c
i
b
0
500
1000
1500
2000
2500
5 1015202530
Distance traveled (cm)
Time (min)
Day 1
Day 3
0
500
1000
1500
2000
2500
5 1015202530
Distance traveled (cm)
Time (min)
Day 1
Day 3
0
2000
4000
6000
8000
10000
Total distance traveled (cm)
Day 1
Day 3
0
2000
4000
6000
8000
10000
Total distance traveled (cm)
Day 1
Day 3
ii
0
2000
4000
6000
8000
10000
Total distance traveled (cm)
Day 1
Day 3
0
500
1000
1500
2000
2500
5 1015202530
Distance traveled (cm)
Time (min)
Day 1
Day 3
0
500
1000
1500
2000
2500
5 1015202530
Distance traveled (cm)
Time (min)
Day 1
Day 3
0
2000
4000
6000
8000
10000
Total distance traveled (cm)
Day 1
Day 3
0
1000
2000
3000
4000
5 1015202530
Distance traveled (cm)
Time (min)
0
5000
10000
15000
20000
Total distance traveled (cm)
Day 1
Day 3
0
1000
2000
3000
4000
5 1015202530
Distance traveled (cm)
Time (min)
Day 1
Day 3
0
5000
10000
15000
20000
Total distance traveled (cm)
Day 1
Day 3
0
5000
10000
15000
20000
Total distance traveled (cm)
Day 1
Day 3
d
ii
ii
ii
Vehicle - Morphine
β-arrestin 2 +/+ β-arrestin 2 /
SP6 - Morphine
Vehicle - Fentanyl
SP6 - Fentanyl
0
1000
2000
3000
4000
51015202530
Distance traveled (cm)
Time (min)
iDay 1
Day 3
Day 1
Day 3
1
3
Day
Day
0
5000
10000
15000
20000
Total distance traveled (cm)
0
1000
2000
3000
4000
5 1015202530
Distance traveled (cm)
Time (min)
i
Day 1
Day 3
**
**
***
*
*
Figure 4 The reduced psychomotor effect of morphine in b-arr2/mice is similarly returned to + / + levels by inhibiting JNK. These data are
presented in the same format as in Figure 2 showing both the distance traveled over time in the line graphs and total locomotion in the accompanying bar
graphs (ai) A 3-day protocol of successive morphine (10 mg/kg s.c.) treatment of b-arr2+ /+ mice resulted in an enhanced locomotor response to the same
dose of morphine by day 3 (po0.01 day 1 vs day 3, F
1,5
¼16.972). (aii) Conversely, similarly treated b-Arr2/mice showed no such increase (p¼0.41
day 1 vs day 3, F
1,6
¼0.765). (bi) Administration of SP6 (30 mg/kg i.p.), 60 min before each morphine injection had no effect on the locomotor sensitization in
b-arr2 + / + mice (po0.05 day 1 vs day 3, F
1,9
¼6.866) but reversed the lack of sensitization in the b-arr2/mice (po0.05 day 1 vs day 3, F
1,5
¼6.989) to
wild-type levels. (c, d) Mice treated with fentanyl (0.2 mg/kg s.c.) showed locomotor sensitization (po0.001 day 1 vs day 3, F
1,23
¼63.235), that was
unaffected by b-arrestin 2 deletion or SP6 administration (vehicle; p¼0.93 day genotype, F
1,23
¼0.007, SP6; p¼0.25 day genotype injection,
F
1,23
¼1.382). *po0.05, **po0.01.
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1959
Neuropsychopharmacology
b-Arrestin 2 has been shown to bind with JNK3, but not
JNK1 or 2, to control GPCR activation of the JNK cascade
(McDonald et al, 2000; Miller et al, 2001; Song et al, 2006,
2009; Willoughby and Collins, 2005). Removing b-arrestin 2
may therefore alter both the location and control of JNK3 to
allow sustained JNK activation in b-arr2/mice. If JNK3
were indeed involved, this is a different isoform to the
JNK2-dependent regulation of receptor uncoupling follow-
ing repeated morphine administration (Melief et al, 2010).
In our experiments, acute mu receptor coupling with
VDCCs was unaffected by JNK inhibition, suggesting that
neither the mu receptor nor Gbg-coupled Ca
2+
channels are
downstream targets of the JNK activity unmasked by
deleting b-arrestin 2. In contrast, SP6 modulation of mu
receptor–VDCC desensitization in b-arr2 + / + neurons
shows a JNK dependency (Figure 5) that could be involved
in the JNK2 regulation of mu receptor–effector coupling
and morphine desensitization in vivo (Melief et al, 2010).
This raises an interesting hypothesis that the same isoform,
or possibly different JNK isoforms, may regulate different
signaling cascades of the mu receptor depending on
whether b-arrestin 2 is present or absent and the protocol
of morphine application.
The effects of morphine on locomotion and subsequent
sensitization in rodents are considered a model of the initial
phase of addiction in humans (Robinson and Berridge,
1993; Vanderschuren and Pierce, 2011). This response
requires D1 receptors, the phosphoprotein DARPP32
(Becker et al, 2001; Borgkvist et al, 2007; Urs et al, 2011)
and ERK (Borgkvist et al, 2008; Urs et al, 2011). We, and
others, have shown that the locomotor effect of morphine is
blunted in mice lacking b-arrestin 2 (Bohn et al, 2003; Urs
et al, 2011). As morphine induces the formation of a
b-arrestin 2/ERK complex in D1 neurons, it has been
suggested that this complex mediates the hyperlocomotor
effect of morphine (Urs et al, 2011). In using pharma-
cological tools to inhibit JNK in b-arrestin 2/mice, we
have shown that activation of the JNK cascade in the
absence of b-arrestin 2 explains the reduced locomotor
response of morphine in these mice. Taken together, these
data and those of Caron and colleagues (Urs et al, 2011)
suggest that b-arrestin 2 plays complementary roles in
this paradigm, one prevents JNK activation whereas the
other allows a b-arrestin/ERK complex to form in D1
neurons. We propose that both roles synergize, and
are necessary for the characteristic locomotor effect of
morphine to occur.
Although others have found that sensitization to morphine
is not b-arrestin 2-dependent (Bohn et al, 2003), we have
found that this context-specific response to multiple doses of
morphine is reduced in mice lacking b-arrestin 2. However,
our assay differed in several important ways; we examined
the first 30 min after morphine (30 vs 120 min), after 3 rather
than 9 days of repeated morphine, and our sensitization
protocol was context-associated. The initial loss of such
‘short-term’ sensitization in b-arr2/mice may therefore
not reflect a lack of, but rather an attenuation of the onset of
sensitization that may not be detectable in a context-
independent manner after an extended protocol of morphine
administration. Furthermore, as JNK inhibition recovered
the locomotor response at similar relative proportions for
each day of the test, this suggests that the JNK cascade does
not block morphine sensitization. However, inhibiting JNK
allows an initial locomotor response to morphine thus
permitting sensitization to develop.
Table 1 JNK Inhibition by SP6 has no Effect on Mu Receptor–VDCC Inhibition in b-arr2/or +/+ Neurons
Agonist Pre-treatment Morphine Fentanyl DAMGO
SP6 SP6 SP6
b-arr2+/+ 32.5±4.1 37.8±5.2 36.3±2.4 36.5±2.5 44.4±2.9 56.0±7.2
b-arr2/22.0±3.6* 23.6±2.8* 26.4±3.1* 24.4±4.4* 26.9±3.0** 23.1±4.4**
The effect of SP6 incubation (10 mM, 4 h) on Ca
2+
channel inhibition (%) by the mu opioid receptor agonists, morphine, fentantyl, and DAMGO (1 mM each) was
assessed in early postnatal dorsal root ganglia neurons from b-arr2+/+ and /mice.
*, **po0.05, 0.01 vs b-arr2+/+, same treatment.
10
0
10
20
30
DAMGO (1μM) inhibition (%)
40
50
60
β-Arr2+/+
β-Arr2/
Baseline
DAMGO
Wash
β-Arr2+/+
β-Arr2/
Pre-treatment M M + SP6 F F + SP6 D D + SP6
a
b
c
M M + SP6 F F + SP6
Figure 5 Mu receptor-Ca
2+
channel inhibition as a possible cytosolic
JNK target. Mu receptor inhibition of voltage-dependent Ca
2+
channels
(VDCC) was examined in b-arr2 + / + and /DRG neurons. Exemplar
currents are shown above the bar graph, which summarizes the effect of
mu agonist pre-treatment coupled with JNK inhibition on mu receptor–
VDCC inhibition, n¼6–30 neurons/sample. Although JNK inhibition had
no effect on mu receptor–VDCC inhibition in b-arr2/or + / + neurons
(Table 1), the desensitization induced by a 4-h pre-treatment of b-arr2 + / +
neurons with morphine (1 mM) was prevented by SP6 co-incubation
(10 mM, 4 h). In contrast, b-arr2/neurons showed no desensitization to
morphine and no effect of SP6. Both fentanyl and DAMGO pre-treatment
desensitized mu receptor–VDCC inhibition in b-ar2 + / + and /neurons
but this was not affected by SP6 co-incubation. M ¼morphine; F ¼fentanyl;
D¼DAMGO. a: b-arr2 + / + neurons, po0.05 vs M, po0.001 vs F, F +
SP6, D, D + SP6. b: b-arr2/neurons; po0.05 vs D, D + SP6, po0.001
vs F, F + SP6, c: b-arr2 + / + neurons; po0.05 vs M + SP. Scale
bars ¼40 ms on the xaxis and 0.4 nA on the yaxis.
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1960
Neuropsychopharmacology
Our findings, and those of several others, show that b-
arrestin 2 regulates mu receptor function through several
different pathways. For example, the enhanced basal
analgesia seen in b-arr2/mice results from an
enhanced level of constitutively active mu receptors
(Lam et al, 2011; Walwyn et al, 2007). Such constitutive
activity also explains the reduced VDCC inhibition by mu
agonists in b-arr2/neurons (Table 1; Figure 5; Lam
et al, 2011; Walwyn et al,2007).Thewell-known
attenuated morphine desensitization and tolerance of the
mu receptor in b-arr2/mice may result from b-arrestin
2 inhibition of mu receptor resensitization, independent of
receptor internalization (Dang et al, 2011; Quillinan et al,
2011). This could explain the lack of desensitization of
DAMGO-VDCC inhibition following chronic morphine
treatments in b-arr2/neurons seen in Figure 5. Our
data add yet another mechanism to b-arrestin 2 regulation
of mu receptor function. We show that the enhanced
morphine-induced analgesia and decreased locomotor
response in b-arr2/mice is JNK-mediated, demonstrat-
ing how b-arrestin 2 normally controls this pathway. It
would be of further interest to examine whether this role
of b-arrestin 2 in masking JNK activation may underlie
the enhanced reward, attenuated tolerance, or indeed
other pertinent phenotypes of morphine in mice lacking
b-arrestin 2.
ACKNOWLEDGEMENTS
This work was supported by NIH Grants DA05010 and
R24-DA025319. O Egbuta, N Desai, N Mittal, and WM
Walwyn are supported in part by Hatos Scholarships. N
Mittal was also supported by the Gates Millennium Scholars
program. Thanks to the laboratory of Robert Lefkowitz for
the b-arrestin-1 and 2 knockout mice.
DISCLOSURE
The authors declare no conflict of interest.
REFERENCES
Arttamangkul S, Quillinan N, Low MJ, von Zastrow M, Pintar J,
Williams JT (2008). Differential activation and trafficking
of micro-opioid receptors in brain slices. Mol Pharmacol 74:
972–979.
Becker A, Grecksch G, Kraus J, Peters B, Schroeder H, Schulz S
et al. (2001). Loss of locomotor sensitisation in response to
morphine in D1 receptor deficient mice. Naunyn Schmiedebergs
Arch Pharmacol 363: 562–568.
Bohn LM, Gainetdinov RR, Sotnikova TD, Medvedev IO, Lefkowitz
RJ, Dykstra LA et al. (2003). Enhanced rewarding properties of
morphine, but not cocaine, in beta(arrestin)-2 knock-out mice.
J Neurosci 23: 10265–10273.
Bohn LM, Lefkowitz RJ, Caron MG (2002). Differential mechan-
isms of morphine antinociceptive tolerance revealed in (beta)ar-
restin-2 knock-out mice. J Neurosci 22: 10494–10500.
Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG, Lin
FT (1999). Enhanced morphine analgesia in mice lacking beta-
arrestin 2. Science 286: 2495–2498.
Bohn LM, Xu F, Gainetdinov RR, Caron MG (2000). Potentiated
opioid analgesia in norepinephrine transporter knock-out mice.
J Neurosci 20: 9040–9045.
Borgkvist A, Usiello A, Greengard P, Fisone G (2007). Activation of
the cAMP/PKA/DARPP-32 signaling pathway is required for
morphine psychomotor stimulation but not for morphine
reward. Neuropsychopharmacology 32: 1995–2003.
Borgkvist A, Valjent E, Santini E, Herve D, Girault JA, Fisone G
(2008). Delayed, context- and dopamine D1 receptor-dependent
activation of ERK in morphine-sensitized mice. Neuropharma-
cology 55: 230–237.
Borgland SL, Connor M, Osborne PB, Furness JB, Christie MJ
(2003). Opioid agonists have different efficacy profiles for G
protein activation, rapid desensitization, and endocytosis of mu-
opioid receptors. J Biol Chem 278: 18776–18784.
Bruchas M, Chavkin C (2010). Kinase cascades and ligand-directed
signaling at the kappa opioid receptor. Psychopharmacology 210:
137–147.
Chen Y, Geis C, Sommer C (2008). Activation of TRPV1 contributes
to morphine tolerance: involvement of the mitogen-activated
protein kinase signaling pathway. J Neurosci 28: 5836–5845.
Dang VC, Chieng B, Azriel Y, Christie MJ (2011). Cellular
morphine tolerance produced by betaarrestin-2-dependent
impairment of mu-opioid receptor resensitization. J Neurosci
31: 7122–7130.
Galandrin SGN, Oligny-Longpre
´GV, Bonin HLN, Ogawa K, Gale
´s
CL, Bouvier M (2008). Conformational rearrangements and
signaling cascades involved in ligand-biased mitogen-activated
protein kinase signaling through the b1-adrenergic receptor. Mol
Pharmacol 74: 162–172.
Gee KW, Yamamura HI (1983). Selective anxiolytics: are the
actions related to partial 00agonist00 activity or a preferential
affinity for benzodiazepine receptor subtypes? Adv Biochem
Psychopharmacol 38: 1–9.
Groer CE, Schmid CL, Jaeger AM, Bohn LM (2011). Agonist-
directed interactions with specific {beta}arrestins determine MU
opioid receptor trafficking, ubiquitination, and dephosphoryla-
tion. J Biol Chem 286: 31731–31741.
Hibi M, Lin A, Smeal T, Minden A, Karin M (1993). Identification
of an oncoprotein- and UV-responsive protein kinase that
binds and potentiates the c-Jun activation domain. Genes Dev 7:
2135–2148.
Lam H, Maga M, Pradhan A, Evans CJ, Maidment NT, Hales TG
et al. (2011). Analgesic tone conferred by constitutively active mu
opioid receptors in mice lacking beta-arrestin 2. Mol Pain 7: 24.
Lopez-Bergami P, Habelhah H, Bhoumik A, Zhang W, Wang LH,
Ronai Z (2005). RACK1 mediates activation of JNK by protein
kinase C [corrected]. Mol Cell 19: 309–320.
McDonald PH, Chow CW, Miller WE, Laporte SA, Field ME, Lin FT
et al. (2000). Beta-arrestin 2: a receptor-regulated MAPK scaffold
for the activation of JNK3. Science 290: 1574–1577.
McPherson J, Rivero G, Baptist M, Llorente J, Al-Sabah S, Krasel C
et al. (2010). m-Opioid receptors: correlation of agonist efficacy
for signalling with ability to activate internalization. Mol
Pharmacol 78: 756–766.
Melief EJ, Miyatake M, Bruchas MR, Chavkin C (2010). Ligand-
directed c-Jun N-terminal kinase activation disrupts opioid
receptor signaling. Proc Natl Acad Sci USA 107: 11608–11613.
Miller WE, McDonald PH, Cai SF, Field ME, Davis RJ, Lefkowitz RJ
(2001). Identification of a motif in the carboxyl terminus of beta-
arrestin2 responsible for activation of JNK3. J Biol Chem 276:
27770–27777.
Peng Y, Zhang Q, Arora S, Keenan SM, Kortagere S, Wannemacher
KM et al. (2009). Novel delta opioid receptor agonists exhibit
differential stimulation of signaling pathways. Bioorg Med Chem
17: 6442–6450.
Ping P (2003). Identification of novel signaling complexes by
functional proteomics. Circ Res 93: 595–603.
Pontrelli P, Ranieri E, Ursi M, Ghosh-Choudhury G, Gesualdo L,
Paolo Schena F et al. (2004). jun-N-terminal kinase regulates
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1961
Neuropsychopharmacology
thrombin-induced PAI-1 gene expression in proximal tubular
epithelial cells. Kidney Int 65: 2249–2261.
Pradhan AA, Walwyn W, Nozaki C, Filliol D, Erbs E, Matifas A
et al. (2010). Ligand-directed trafficking of the delta-opioid
receptor in vivo: two paths toward analgesic tolerance. J Neurosci
30: 16459–16468.
Quillinan N, Lau EK, Virk M, von Zastrow M, Williams JT (2011).
Recovery from mu-opioid receptor desensitization after
chronic treatment with morphine and methadone. J Neurosci
31: 4434–4443.
Raehal KM, Bohn LM (2011). The role of beta-arrestin2 in the
severity of antinociceptive tolerance and physical dependence
induced by different opioid pain therapeutics. Neuropharmacol-
ogy 60: 58–65.
Reiter E, Ahn S, Shukla AK, Lefkowitz RJ (2012). Molecular
mechanism of beta-arrestin-biased agonism at seven-transmem-
brane receptors. Annu Rev Pharmacol Toxicol 52: 179–197.
Robinson TE, Berridge KC (1993). The neural basis of drug
craving: an incentive-sensitization theory of addiction. Brain Res
Brain Res Rev 18: 247–291.
Shenoy SK, Lefkowitz RJ (2011). beta-arrestin-mediated receptor
trafficking and signal transduction. Trends Pharmacol Sci 32:
521–533.
Shin M, Yan C, Boyd D (2002). An inhibitor of c-jun aminoter-
minal kinase (SP600125) represses c-Jun activation, DNA-
binding and PMA-inducible 92-kDa type IV collagenase
expression. Biochim Biophys Acta 1589: 311–316.
Shukla AK, Xiao K, Lefkowitz RJ (2011). Emerging paradigms of
beta-arrestin-dependent seven transmembrane receptor signal-
ing. Trends Biochem Sci 36: 457–469.
Song X, Coffa S, Fu H, Gurevich VV (2009). How does arrestin
assemble MAPKs into a signaling complex? J Biol Chem 284:
685–695.
Song X, Raman D, Gurevich EV, Vishnivetskiy SA, Gurevich
VV (2006). Visual and both non-visual arrestins in their
00inactive00 conformation bind JNK3 and Mdm2 and relocalize
them from the nucleus to the cytoplasm. J Biol Chem 281:
21491–21499.
Sora I, Takahashi N, Funada M, Ujike H, Revay RS, Donovan DM
et al. (1997). Opiate receptor knockout mice define mu
receptor roles in endogenous nociceptive responses and
morphine-induced analgesia. Proc Natl Acad Sci USA 94:
1544–1549.
Stebbins JL, De SK, Machleidt T, Becattini B, Vazquez J, Kuntzen C
et al. (2008). Identification of a new JNK inhibitor targeting
the JNK-JIP interaction site. Proc Natl Acad Sci USA 105:
16809–16813.
Tan M, Groszer M, Tan AM, Pandya A, Liu X, Xie CW (2003).
Phosphoinositide 3-kinase cascade facilitates mu-opioid desen-
sitization in sensory neurons by altering G-protein-effector
interactions. J Neurosci 23: 10292–10301.
Urban JD, Clarke WP, von Zastrow M, Nichols DE, Kobilka B,
Weinstein H et al. (2007). Functional selectivity and classical
concepts of quantitative pharmacology. J Pharmacol Exp Ther
320: 1–13.
Urs NM, Daigle TL, Caron MG (2011). A dopamine D1 receptor-
dependent [beta]-arrestin signaling complex potentially regu-
lates morphine-induced psychomotor activation but not reward
in mice. Neuropsychopharmacology 36: 551–558.
Vanderschuren LJ, Pierce RC (2011). Sensitization processes in
drug addiction. Curr Top Behav Neurosci 3: 179–195.
Walwyn W, Evans CJ, Hales TG (2007). Beta-arrestin2 and c-Src
regulate the constitutive activity and recycling of mu opioid
receptors in dorsal root ganglion neurons. JNeurosci27: 5092–5104.
Wilkinson SE, Parker PJ, Nixon JS (1993). Isoenzyme specificity of
bisindolylmaleimides, selective inhibitors of protein kinase C.
Biochem J 294(Part 2): 335–337.
Willoughby EA, Collins MK (2005). Dynamic interaction between
the dual specificity phosphatase MKP7 and the JNK3 scaffold
protein beta-arrestin 2. J Biol Chem 280: 25651–25658.
Xiao K, McClatchy DB, Shukla AK, Zhao Y, Chen M, Shenoy SK
et al. (2007). Functional specialization of beta-arrestin interac-
tions revealed by proteomic analysis. Proc Natl Acad Sci USA
104: 12011–12016.
Xiao K, Sun J, Kim J, Rajagopal S, Zhai B, Villen J et al. (2010).
Global phosphorylation analysis of beta-arrestin-mediated
signaling downstream of a seven transmembrane receptor
(7TMR). Proc Natl Acad Sci USA 107: 15299–15304.
Zheng H, Chu J, Zhang Y, Loh HH, Law PY (2011). Modulating
micro-opioid receptor phosphorylation switches agonist-depen-
dent signaling as reflected in PKCepsilon activation and
dendritic spine stability. J Biol Chem 286: 12724–12733.
Arrestin and JNK regulation of mu receptor function
N Mittal et al
1962
Neuropsychopharmacology
... Previous work using βarrestin2-knockout (βarrestin2-KO) mice showed that morphine induced greater antinociception in the absence of βarrestin2, with less development of tolerance [6,7]. The βarrestin2-KO mice also displayed less morphine-induced hyperactivity [8,9]. Recently, we described a series of structurally related MOR-selective agonists that displayed a preference for inducing MOR-G protein signaling over βarrestin2 recruitment [10]. ...
Article
Full-text available
Opioid analgesics such as morphine and fentanyl induce mu-opioid receptor (MOR)-mediated hyperactivity in mice. Herein, we show that morphine, fentanyl, SR-17018, and oliceridine have submaximal intrinsic efficacy in the mouse striatum using 35S-GTPγS binding assays. While all of the agonists act as partial agonists for stimulating G protein coupling in striatum, morphine, fentanyl, and oliceridine are fully efficacious in stimulating locomotor activity; meanwhile, the noncompetitive biased agonists SR-17018 and SR-15099 produce submaximal hyperactivity. Moreover, the combination of SR-17018 and morphine attenuates hyperactivity while antinociceptive efficacy is increased. The combination of oliceridine with morphine increases hyperactivity, which is maintained over time. These findings provide evidence that noncompetitive agonists at MOR can be used to suppress morphine-induced hyperactivity while enhancing antinociceptive efficacy; moreover, they demonstrate that intrinsic efficacy measured at the receptor level is not directly proportional to drug efficacy in the locomotor activity assay.
... Previous work using arrestin2-knockout (arrestin2-KO) mice showed that morphine induced greater antinociception in the absence of arrestin2 with less development of tolerance [6,7]. The arrestin2-KO mice also display less morphine-induced hyperactivity [8,9]. We described a series of structurally related MOR-selective agonists that display a preference for inducing MOR-G protein signaling over arrestin2 recruitment (Schmid et al., 2017). ...
Preprint
Opioid analgesics like morphine and fentanyl induce mu-opioid receptor (MOR)-mediated hyperactivity in mice. Here we show that morphine, fentanyl, SR-17018, and oliceridine have submaximal intrinsic efficacy in the mouse striatum using 35S-GTPγS binding assays. While all of the agonists act as partial agonists for stimulating G protein coupling in striatum, morphine, fentanyl and oliceridine are fully efficacious in stimulating locomotor activity; meanwhile, the noncompetitive biased agonists, SR-17018 and SR-15099 produce submaximal hyperactivity. Moreover, the combination of SR-17018 and morphine attenuates hyperactivity while antinociceptive efficacy is increased. The combination of oliceridine with morphine increases hyperactivity which is maintained over time. These findings provide evidence that noncompetitive agonists at MOR can be used to suppress morphine-induced hyperactivity while enhancing antinociceptive efficacy; moreover, they demonstrate that intrinsic efficacy measured at the receptor level is not directly proportional to drug efficacy in the locomotor activity assay.
... No phenotype differences were detected between βarr2(+/+) and βarr2(−/−) mice with respect to their sensitivity to formalin and motor performance on the rotarod. This confirms previous studies showing no differences in terms of nociception between βarr2(+/+) and βarr2(−/−) mice (Bohn et al., 1999;Mittal et al., 2012). Additionally, no differences were reported in the literature between the basal locomotor activity of βarr2(+/+) and βarr2(−/−) mice (Beaulieu et al., 2005;Urs et al., 2011;Del'Guidice and Beaulieu, 2015;Toth et al., 2018). ...
Article
Full-text available
Nociceptin/orphanin FQ controls several functions, including pain transmission, via stimulation of the N/OFQ peptide (NOP) receptor. Here we tested the hypothesis that NOP biased agonism may be instrumental for identifying innovative analgesics. In vitro experiments were performed with the dynamic mass redistribution label free assay and the NOP non-peptide agonists Ro 65-6570, AT-403 and MCOPPB. In vivo studies were performed in wild type and β-arrestin 2 knockout mice using the formalin, rotarod and locomotor activity tests. In vitro all compounds mimicked the effects of N/OFQ behaving as potent NOP full agonists. In vivo Ro 65-6570 demonstrated a slightly higher therapeutic index (antinociceptive vs. motor impairment effects) in knockout mice. However, all NOP agonists displayed very similar therapeutic index in normal mice despite significant differences in G protein biased agonism. In conclusion the different ability of inducing G protein vs. β-arrestin 2 recruitment of a NOP agonist cannot be applied to predict its antinociceptive vs. motor impairment properties.
... β-Arrestin2, as a scaffolding protein, regulates the signaling cascade of commonly used analgesics, including the JNK pathway; JNK binds to β-arrestin2 and modulates the analgesic effects of morphine. 39 Morphine can activate and bind to Gα i -coupled μ receptors, and its binding to these receptors leads to the dissociation of Gα i and Gβγ and induces β-arrestin2 recruitment. 40 MOR is phosphorylated by the action of G protein-coupled receptor kinases (GRKs) after binding with morphine. ...
Article
Full-text available
Purpose: Morphine is often used for the treatment of moderate and severe cancer pain, but long-term use can lead to morphine tolerance. Methods for effectively inhibiting morphine tolerance and the related mechanism of action are of great significance for the treatment of cancer pain. Previous studies have shown that electroacupuncture (EA) can inhibit the occurrence of morphine tolerance, but the mechanism is not yet clear. The aim of the present study was to explore the signaling pathway by which EA attenuates the development of bone cancer pain (BCP)-morphine tolerance (MT). Materials and methods: Changes in the paw withdrawal threshold (PWT) of rats with bone cancer pain-morphine tolerance were observed in a study of EA combined with intrathecal injection of a PI3K inhibitor (LY294002) or agonist (insulin-like growth factor-1 [IGF-1]). We also tested the protein expression of phosphorylated phosphatidylinositol 3-kinase (p-PI3K), phosphorylated protein kinase B (p-Akt), phosphorylated c-Jun NH2-terminal kinase 1/2 (p-JNK1/2), and β-arrestin2 in the L4-6 spinal dorsal horn of rats. Results: The protein expression of p-PI3K, p-Akt, p-JNK1/2, and β-arrestin2 was upregulated in the L4-6 spinal dorsal horn of rats with bone cancer pain and bone cancer pain-morphine tolerance. EA delayed the occurrence of morphine tolerance in rats with bone cancer pain and downregulated the protein expression of p-PI3K, p-Akt, p-JNK1/2, and β-arrestin2 in the L4-6 spinal dorsal horn of rats with bone cancer pain-morphine tolerance. Intrathecal injection of LY294002 attenuated the development of morphine tolerance and downregulated the protein expression of p-Akt, p-JNK1/2, and β-arrestin2 in the spinal dorsal horn of rats with bone cancer pain-morphine tolerance. In addition, the inhibitory effect of EA on morphine tolerance was reversed by IGF-1. Conclusion: The mechanism underlying the ability of EA to attenuate morphine tolerance may be associated with inhibition of the PI3K/Akt/JNK1/2 signaling pathway.
... One component of acute desensitization was dependent on PKC following chronic morphine treatment (Bailey et al., 2009;Levitt and Williams, 2012). In addition, morphine-induced acute MOR desensitization in the dorsal root ganglion (Mittal et al., 2012), spinally-mediated acute analgesic tolerance induced by morphine administration (Melief et al., 2010) and centrally-mediated tolerance to morphine all involved JNK (Kuhar et al., 2015). In the present study, when the combination of staurosporine and CMP101 was used, the results were indistinguishable from those using the combination of selective inhibitors of GRK2/3, PKC, and JNK. ...
Article
Based on studies using mutations of the m-opioid receptor (MOR), phosphorylation of multiple sites on the C-terminus has been recognized as a critical step underlying acute desensitization and the development of cellular tolerance. The aim of this study is to explore which kinases mediate desensitization of MOR in brain slices from drug-naïve and morphine-treated animals. Whole-cell recordings from locus coeruleus neurons were made, and the agonist-induced increase in potassium conductance was measured. In slices from naïve animals, pharmacological inhibition of G-protein receptor kinase (GRK2/3) with compound 101 blocked acute desensitization. Following chronic treatment withmorphine, compound 101 was less effective at blocking acute desensitization. Compound 101 blocked receptor internalization in tissue from both naïve and morphine-treated animals, suggesting that GRK2/3 remained active. Kinase inhibitors aimed at blocking protein kinase C and c-Jun N-terminal kinase had no effect on desensitization in tissue taken from naïve animals. However, in slices taken from morphine-treated animals, the combination of these blockers along with compound 101 was required to block acute desensitization. Acute desensitization of the potassium conductance induced by the somatostatin receptor was also blocked by compound 101 in slices from naïve but not morphinetreated animals. As was observed with MOR, it was necessary to use the combination of kinase inhibitors to block desensitization of the somatostatin receptor in slices from morphine-treated animals. The results show that chronic treatment with morphine results in a surprising and heterologous adaptation in kinasedependent desensitization. © 2020 by The American Society for Pharmacology and Experimental Therapeutics.
Article
Full-text available
According to the World Health Organization (WHO), around 11 million people suffer from burns every year, and 180,000 die from them. A burn is a condition in which heat, chemical substances, an electrical current or other factors cause tissue damage. Burns mainly affect the skin, but can also affect deeper tissues such as bones or muscles. When burned, the skin loses its main functions, such as protection from the external environment, pathogens, evaporation and heat loss. Depending on the stage of the burn, the patient’s condition and the cause of the burn, we need to choose the most appropriate treatment. Personalization and multidisciplinary collaboration are key to the successful management of burn patients. In this comprehensive review, we have collected and discussed the available treatment options, focusing on recent advances in topical treatments, wound cleansing, dressings, skin grafting, nutrition, pain and scar tissue management.
Article
Opioid receptors are G protein-coupled receptors (GPCRs) that regulate activity within peripheral, subcortical and cortical circuits involved in pain, reward, and aversion processing. These receptors are expressed in both presynaptic terminals where they inhibit neurotransmitter release and postsynaptic locations where they act to hyperpolarize neurons and reduce activity. Agonist activation of postsynaptic receptors at the plasma membrane signal via ion channels or cytoplasmic second messengers. Agonist binding initiates regulatory processes that include phosphorylation by G protein receptor kinases (GRKs) and recruitment of beta-arrestins that desensitize and internalize the receptors. Opioid receptors also couple to effectors from endosomes activating intracellular enzymes and kinases. In contrast to postsynaptic opioid receptors, receptors localized to presynaptic terminals are resistant to desensitization such that there is no loss of signaling over the same time scale. Thus, the balance of opioid signaling in circuits expressing pre- and postsynaptic opioid receptors is shifted toward inhibition of presynaptic neurotransmitter release. The functional implication of this shift is not often acknowledged in behavioral studies. This review covers what is currently understood about regulation of opioid/nociceptin receptors, with an emphasis on opioid receptor signaling in pain and reward circuits. Importantly, the review covers regulation of presynaptic receptors and the critical gaps in understanding this area, as well as the opportunities to further understand opioid signaling in brain circuits.
Article
Background: The prescription of methadone in advanced cancer poses multiple challenges due to the considerable interpatient variation seen in effective dose and toxicity. Previous reports have suggested that ARRB2 influences the response to methadone in opioid substitution therapy. Associations with opioid response for pain management in advanced cancer are conflicting, with no studies including methadone as the primary intervention. Methods: In a prospective, multicenter, open-label dose individualization study, we investigated whether polymorphisms in ARRB2 were associated with methadone dose requirements and pain severity. Results: Significant associations were found for rs3786047, rs1045280, rs2036657 and pain score. Conclusion: While studies are few and the sample size small, ARRB2 genotyping may assist in individualized management of the most feared symptom in advanced cancer.
Chapter
Opioid analgesics have been used for centuries for the treatment of acute and chronic pain conditions. In the acute surgical or injury pain setting, their efficacy is a mainstay; however, for the management of chronic pain, their use may be limited due to a myriad of unwanted side effects. Attempts have been made to reduce these side effects by modifying opioid drugs to impart diverse pharmacological properties ranging from varying efficacy and selectivity to diversifying downstream receptor signaling. In this review we discuss how recently developed compounds compare to conventional agonists such as morphine and fentanyl and discuss strategies that could contribute to improve opioid utility while limiting side adverse events.
Article
Morphine is a potent opioid analgesic with high propensity for the development of antinociceptive tolerance. Morphine antinociception and tolerance are partially regulated by the midbrain ventrolateral periaqueductal gray (vlPAG). However, the majority of research evaluating mu‐opioid receptor signaling has focused on males. Here, we investigate kinase activation and localization patterns in the vlPAG following acute and chronic morphine treatment in both sexes. Male and female mice developed rapid antinociceptive tolerance to morphine (10 mg/kg i.p.) on the hot plate assay, but tolerance did not develop in males on the tail flick assay. Quantitative fluorescence immunohistochemistry was used to map and evaluate the activation of extracellular signal‐regulated kinase 1/2 (ERK 1/2), protein kinase‐C (PKC), and protein kinase‐A (PKA). We observed significantly greater phosphorylated ERK 1/2 in the vlPAG of chronic morphine‐treated animals which co‐localized with the endosomal marker, Eea1. We note that pPKC is significantly elevated in the vlPAG of both sexes following chronic morphine treatment. We also observed that although PKA activity is elevated following chronic morphine treatment in both sexes, there is a significant reduction in the nuclear translocation of its phosphorylated substrate. Taken together, this study demonstrates increased activation of ERK 1/2, PKC, and PKA in response to repeated morphine treatment. The study opens avenues to explore the impact of chronic morphine treatment on G‐protein signaling and kinase nuclear transport. image
Article
Full-text available
Signaling via G-protein-coupled receptors undergoes desensitization after prolonged agonist exposure. Here we investigated the role of phosphoinositide 3-kinase (PI3K) and its downstream pathways in desensitization of mu-opioid inhibition of neuronal Ca2+ channels. In cultured mouse dorsal root ganglion neurons, two mechanistically different forms of desensitization were observed after acute or chronic treatment with the mu agonist [D-Ala(2), N-MePhe(4), Gly-ol(5)]-enkephalin (DAMGO). Chronic DAMGO desensitization was heterologous in nature and significantly attenuated by blocking the activity of PI3K or mitogen-activated protein kinase (MAPK). A combined application of PI3K and MAPK inhibitors showed no additive effect, suggesting that these two kinases act in a common pathway to facilitate chronic desensitization. Acute DAMGO desensitization, however, was not affected by the inhibitors. Furthermore, upregulation of the PI3K-Akt pathway in mutant mice lacking phosphatase and tensin homolog, a lipid phosphatase counteracting PI3K, selectively enhanced chronic desensitization in a PI3K- and MAPK-dependent manner. Using the prepulse facilitation (PPF) test, we further examined changes in the voltage-dependent component of DAMGO action that requires direct interactions between betagamma subunits of G-proteins and Ca2+ channels. DAMGO-induced PPF was diminished after chronic treatment, suggesting disruption of G-protein-channel interactions. Such disruption could occur at the postreceptor level, because chronic DAMGO also reduced GTPgammaS-induced PPF that was independent of receptor activation. Again, inhibition of PI3K or MAPK reduced desensitization of PPF. Our data suggest that the PI3K cascade involving MAPK and Akt enhances mu-opioid desensitization via postreceptor modifications that interfere with G-protein-effector interactions.
Article
Full-text available
The concept of intrinsic efficacy has been enshrined in pharmacology for half of a century, yet recent data have revealed that many ligands can differentially activate signaling pathways mediated via a single G protein-coupled receptor in a manner that challenges the traditional definition of intrinsic efficacy. Some terms for this phenomenon include functional selectivity, agonist-directed trafficking, and biased agonism. At the extreme, functionally selective ligands may be both agonists and antagonists at different functions mediated by the same receptor. Data illustrating this phenomenon are presented from serotonin, opioid, dopamine, vasopressin, and adrenergic receptor systems. A variety of mechanisms may influence this apparently ubiquitous phenomenon. It may be initiated by differences in ligand-induced intermediate conformational states, as shown for the beta(2)-adrenergic receptor. Subsequent mechanisms that may play a role include diversity of G proteins, scaffolding and signaling partners, and receptor oligomers. Clearly, expanded research is needed to elucidate the proximal (e.g., how functionally selective ligands cause conformational changes that initiate differential signaling), intermediate (mechanisms that translate conformation changes into differential signaling), and distal mechanisms (differential effects on target tissue or organism). Besides the heuristically interesting nature of functional selectivity, there is a clear impact on drug discovery, because this mechanism raises the possibility of selecting or designing novel ligands that differentially activate only a subset of functions of a single receptor, thereby optimizing therapeutic action. It also may be timely to revise classic concepts in quantitative pharmacology and relevant pharmacological conventions to incorporate these new concepts.
Article
Full-text available
The concept of biased agonism has recently come to the fore with the realization that seven-transmembrane receptors (7TMRs, also known as G protein-coupled receptors, or GPCRs) activate complex signaling networks and can adopt multiple active conformations upon agonist binding. As a consequence, the "efficacy" of receptors, which was classically considered linear, is now recognized as pluridimensional. Biased agonists selectively stabilize only a subset of receptor conformations induced by the natural "unbiased" ligand, thus preferentially activating certain signaling mechanisms. Such agonists thus reveal the intriguing possibility that one can direct cellular signaling with unprecedented precision and specificity and support the notion that biased agonists may identify new classes of therapeutic agents that have fewer side effects. This review focuses on one particular class of biased ligands that has the ability to alter the balance between G protein-dependent and β-arrestin-dependent signal transduction.
Article
Full-text available
Morphine and other opiates mediate their effects through activation of the μ-opioid receptor (MOR), and regulation of the MOR has been shown to critically affect receptor responsiveness. Activation of the MOR results in receptor phosphorylation, β-arrestin recruitment, and internalization. This classical regulatory process can differ, depending on the ligand occupying the receptor. There are two forms of β-arrestin, β-arrestin1 and β-arrestin2 (also known as arrestin2 and arrestin3, respectively); however, most studies have focused on the consequences of recruiting β-arrestin2 specifically. In this study, we examine the different contributions of β-arrestin1- and β-arrestin2-mediated regulation of the MOR by comparing MOR agonists in cells that lack expression of individual or both β-arrestins. Here we show that morphine only recruits β-arrestin2, whereas the MOR-selective enkephalin [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), recruits either β-arrestin. We show that β-arrestins are required for receptor internalization and that only β-arrestin2 can rescue morphine-induced MOR internalization, whereas either β-arrestin can rescue DAMGO-induced MOR internalization. DAMGO activation of the receptor promotes MOR ubiquitination over time. Interestingly, β-arrestin1 proves to be critical for MOR ubiquitination as modification does not occur in the absence of β-arrestin1 nor when morphine occupies the receptor. Moreover, the selective interactions between the MOR and β-arrestin1 facilitate receptor dephosphorylation, which may play a role in the resensitization of the MOR and thereby contribute to overall development of opioid tolerance.
Article
This paper presents a biopsychological theory of drug addiction, the 'Incentive-Sensitization Theory'. The theory addresses three fundamental questions. The first is: why do addicts crave drugs? That is, what is the psychological and neurobiological basis of drug craving? The second is: why does drug craving persist even after long periods of abstinence? The third is whether 'wanting' drugs (drug craving) is attributable to 'liking' drugs (to the subjective pleasurable effects of drugs)? The theory posits the following. (1) Addictive drugs share the ability to enhance mesotelencephalic dopamine neurotransmission. (2) One psychological function of this neural system is to attribute 'incentive salience' to the perception and mental representation of events associated with activation of the system. Incentive salience is a psychological process that transforms the perception of stimuli, imbuing them with salience, making them attractive, 'wanted', incentive stimuli. (3) In some individuals the repeated use of addictive drugs produces incremental neuroadaptations in this neural system, rendering it increasingly and perhaps permanently, hypersensitive ('sensitized') to drugs and drug-associated stimuli. The sensitization of dopamine systems is gated by associative learning, which causes excessive incentive salience to be attributed to the act of drug taking and to stimuli associated with drug taking. It is specifically the sensitization of incentive salience, therefore, that transforms ordinary 'wanting' into excessive drug craving. (4) It is further proposed that sensitization of the neural systems responsible for incentive salience ('for wanting') can occur independently of changes in neural systems that mediate the subjective pleasurable effects of drugs (drug 'liking') and of neural systems that mediate withdrawal. Thus, sensitization of incentive salience can produce addictive behavior (compulsive drug seeking and drug taking) even if the expectation of drug pleasure or the aversive properties of withdrawal are diminished and even in the face of strong disincentives, including the loss of reputation, job, home and family. We review evidence for this view of addiction and discuss its implications for understanding the psychology and neurobiology of addiction.
Article
Mice lacking D1 receptors were used to study the role of these receptors in morphine-induced antinociception and locomotor sensitisation. In the hot-plate test D1 receptor deficient (-/-) and wild-type (+/+) mice showed similar reaction times under basal conditions. A single injection of 1.25 mg/kg and 2.5 mg/kg morphine resulted in a stronger antinociceptive response in D1 receptor deficient mice than in wild-type animals. Tolerance to the analgesic effect did not develop in both groups of animals when 12.5 mg/kg morphine was chronically applied twice daily for 13 days. There was no change in basal locomotor activity between saline-injected wild-type and D1 receptor deficient mice. After chronic treatment wild-type mice showed a continuous increase in locomotor activity, indicating the development of sensitisation. In contrast, a subchronic administration of morphine did not change locomotor activity in mutant mice. The lack of the development of locomotor sensitisation in D1 deficient mice was associated with reduced levels of immunoreactive opioid receptors in dorsal striatal patches as compared to wild-type mice. In contrast, no change in the distribution of immunoreactive receptors could be detected in areas related to pain pathways such as the spinal cord. Taken together, these results suggest an involvement of D1 receptors in morphine-induced locomotor activity and analgesia.
Article
This paper presents a biopsychological theory of drug addiction, the ‘Incentive-Sensitization Theory’. The theory addresses three fundamental questions. The first is: why do addicts crave drugs? That is, what is the psychological and neurobiological basis of drug craving? The second is: why does drug craving persist even after long periods of abstinence? The third is whether ‘wanting’ drugs (drug craving) is attributable to ‘liking’ drugs (to the subjective pleasurable effects of drugs)? The theory posits the following.
Article
β-Arrestins, originally discovered to desensitize activated seven transmembrane receptors (7TMRs; also known as G-protein-coupled receptors, GPCRs), are now well established mediators of receptor endocytosis, ubiquitylation and G protein-independent signaling. Recent global analyses of β-arrestin interactions and β-arrestin-dependent phosphorylation events have uncovered several previously unanticipated roles of β-arrestins in a range of cellular signaling events. These findings strongly suggest that the functional roles of β-arrestins are much broader than currently understood. Biophysical studies aimed at understanding multiple active conformations of the 7TMRs and the β-arrestins have begun to unravel the mechanistic basis for the diverse functional capabilities of β-arrestins in cellular signaling.