Sunteți pe pagina 1din 9

ARCHIVAL REPORT

Presynaptic Leptin Action Suppresses Excitatory


Synaptic Transmission onto Ventral Tegmental Area
Dopamine Neurons
Jennifer L. Thompson and Stephanie L. Borgland
Background: Leptin is an adipocyte-derived cytokine that can act in the brain to suppress feeding and maintain energy homeostasis.
Additionally, leptin activates its receptors in the ventral tegmental area (VTA), a critical site for neuroadaptations to rewarding stimuli, to
modulate reward-seeking behaviors. Although leptin can decrease intrinsic excitability of dopamine neurons in the VTA, it is unknown
whether leptin can modulate excitatory synaptic transmission in this brain region. Because plasticity of glutamatergic synapses onto VTA
neurons can encode predictive information about reward, we hypothesized that leptin can decrease excitatory synaptic transmission
onto dopamine neurons.

Methods: Using whole-cell patch clamp electrophysiology in mouse midbrain slices, we tested the effects of leptin on evoked a-amino-
3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) or N-methyl-D-aspartate receptor (NMDAR)-mediated excitatory postsynaptic
currents (EPSCs) onto VTA dopamine neurons.
Results: Leptin depressed both AMPAR and NMDAR EPSCs in VTA dopamine neurons and reduced frequency but not amplitude of
mini EPSCs. Bath application of the MEK1/2 inhibitor U0126 did not alter leptin-induced suppression of AMPAR EPSCs. However,
external, but not internal, application of the phosphoinositol 3-kinase (PI3K) or Janus kinase 2 (Jak2) tyrosine kinase inhibitors abolished
leptin-induced synaptic depression.

Conclusions: This study demonstrates that leptin causes a presynaptic inhibition of the probability of glutamate release onto VTA
dopamine neurons. This synaptic inhibition requires Jak2 and PI3K activation. Leptin-induced weakening of synaptic strength onto
dopamine cells may underlie its inhibitory effects on appetitive behavior for rewarding stimuli.

So far, there is little understanding of the mechanism behind


Key Words: AMPA, dopamine, leptin, presynaptic, synaptic leptin modulation of these effects. Supraphysiologic doses of
transmission, VTA systemic leptin decrease the firing rate of putative VTA dopamine
neurons in anesthetized rats (5). Furthermore, bath application
of leptin to midbrain slices decreases firing rate of putative

L
eptin is a cytokine released from adipocytes that circulates dopamine neurons by approximately 20% (5) in an extracellular
throughout the central nervous system (CNS) to convey the signal-regulated kinase (ERK)-dependent manner (13). Leptin-
status of body energy stores and to control feeding and induced decrease in neuronal firing may be due to a change in
energy expenditure. Leptin readily crosses the blood–brain intrinsic neuronal properties of dopamine neurons or a result of
barrier through a saturable transport system (1–3) and binds to altered pre- or postsynaptic effects of glutamate or gamma-
its long-form signaling receptor, LepRb, in many brain regions aminobutyric acid (GABA) onto dopamine neurons. However, it
(2,4) including the ventral tegmental area (VTA) (5–8). VTA is unknown how leptin modulates excitatory or inhibitory
dopamine neurons and their widespread neural targets in the synaptic transmission onto VTA dopamine neurons. Here, we
striatum, amygdala, prefrontal cortex, and elsewhere mediate the test the hypothesis that leptin modulates excitatory synaptic
incentive salience of food and other rewards (9). Leptin signaling transmission onto dopamine neurons.
in the VTA may be one mechanism used to alter the motivation Glutamatergic synapses onto VTA dopamine neurons can
to consume food or drugs of abuse. Indeed, intra-VTA leptin undergo both long-term potentiation (LTP) and long-term
decreases food consumption (5) and LepRb knockdown in the depression (LTD) to regulate their synaptic strength and conse-
VTA increases effort to obtain palatable food reinforcers (10). quent dopaminergic output (14–18). Enhanced synaptic efficacy
Systemic leptin reduces the rewarding value of sucrose that is onto dopamine neurons has been linked not only to exposure
dependent on dopamine neuron activation (11). Taken together, to drugs of abuse but to feeding-related peptides, such as
a wealth of evidence suggests that leptin signaling in the hypocretin/orexin (19,20) and ghrelin (21). Potentiation of synap-
mesolimbic dopamine circuit can modulate natural and drug tic strength onto dopamine neurons by feeding-promoting
reward-seeking behaviors (12). peptides may underlie motivation to obtain food (20,22,23).
Therefore, modulation of excitatory inputs by anorectic feeding
peptides such as leptin could be particularly important in
From the Department of Anesthesiology, Pharmacology and Therapeutics, decreasing dopamine neuron firing activity and ultimately dopa-
University of British Columbia, Vancouver, Canada. mine release.
Address correspondence to Stephanie L. Borgland, Ph.D., Department of Leptin modulation of glutamatergic synaptic transmission in
Anesthesiology, Pharmacology and Therapeutics, University of British other brain regions has been demonstrated in the hippocampus
Columbia, 212-2176 Health Sciences Mall, Vancouver BC V6T 1Z3, (24–26), hypothalamus (27), and cerebellum (28). Thus, we
Canada; E-mail: Borgland@mail.ubc.ca. hypothesized that leptin action in the VTA can modulate
Received Jul 14, 2012; revised and accepted Oct 31, 2012. glutamatergic synaptic transmission onto VTA dopamine neurons.

0006-3223/$36.00 BIOL PSYCHIATRY 2013;73:860–868


http://dx.doi.org/10.1016/j.biopsych.2012.10.026 & 2013 Society of Biological Psychiatry
J.L. Thompson and S.L. Borgland BIOL PSYCHIATRY 2013;73:860–868 861

Here, we used whole-cell patch clamp electrophysiology in VTA stimulate local presynaptic terminals, a bipolar stimulating electrode
brain slices to determine the effect of leptin on excitatory synaptic was placed 100 to 300 mm rostral to the recording electrode and was
transmission. used to stimulate excitatory afferents at .1 Hz. Neurons were voltage-
clamped at –70 mV and ⫹40 mV to record AMPAR- and N-methyl-D-
aspartate receptor (NMDAR)-mediated evoked excitatory-
Methods and Materials
postsynaptic currents (EPSCs), respectively. EPSCs were filtered at 2
Animals kHz, digitized at 5 to 10 kHz, and collected online using pClamp10
All protocols were in accordance with the ethical guidelines software (Molecular Devices). Series resistance (6–30 MO) and input
established by the Canadian Council for Animal care and were resistance were monitored online with a 5-mV depolarizing step (50
approved by the University of British Columbia Animal Care msec) given just after every afferent stimulus. Paired-pulse ratio was
Committee. Male C57BL/6 mice (p21–25; University of British elicited by evoking 2 EPSCs with an interval of 50 msec. mEPSCs
Columbia) were housed in groups of 3 to 5. For some experi- were recorded in the presence of tetrodotoxin (TTX; 500 nmol/L) to
ments, mice expressing pituitary homeobox 3 tagged with green block action potentials driven by spontaneous events as well as
fluorescent protein (Pitx3-GFP) knock-in mice (p21–25; bred picrotoxin (100 mmol/L) and (2R)-amino-5-phosphonovaleric acid AP-
inhouse) were used to easily identify VTA dopaminergic neurons 5 (50 mmol/L). For these experiments, mEPSCs were recorded before
because Pitx3 is a transcription factor necessary for the develop- and 15 min after a 15-min application of leptin (100 nmol/L). mEPSCs
ment of midbrain dopamine neurons (29). Data from C57BL/6 were selected on the basis of their amplitude (⬎10 pA), decay time
mice and Pitx3-GFP mice were not significantly different and (⬍3 msec), and rise time (⬍3 msec) using the MiniAnalysis60
therefore were grouped together. Mice were maintained on a 12- program (Synaptosoft, Decateur, Georgia). Average noise levels were
hour light:dark schedule (lights on at 7:00 AM) and given food and 4 ⫾ .2 pA. Example traces for evoked EPSCs were constructed from
water ad libitum. All experiments were performed during the the averaged trace of 12 sweeps (2 min).
animals light cycle.
Drugs
Slice Preparation Recombinant mouse leptin (R&D Systems, Minneapolis,
Briefly, animals were deeply anaesthetized with isoflurane and Minnesota), was reconstituted in phosphate-buffered saline
decapitated, and brains were rapidly extracted into ice-cold (pH 8.0–8.2) and stored at –801C. Leptin was bath applied at
sucrose solution containing (in mmol/L: 75 sucrose, 87 NaCl, 2.5 100-nmol/L concentrations for 15 min in all experiments. Picrotoxin
KCl, 1.25 NaH2PO4, 25 NaHCO3, 7 MgCl2, .95 CaCl2, 1 to 1.5 (Sigma, St. Louis, Missouri) was dissolved in H20 and was present in
ascorbic acid. Horizontal brain sections containing the VTA (30) the external solution throughout all recordings. U0126 (Promega,
were cut at 250 mm on a vibratome (Leica, Nussloch, Germany). Madison, Wisconsin), wortmannin (Tocris, Ellisville, Missouri), or
Slices were transferred to 250-mL bicarbonate-buffered solution AG490 (Calbiochem, Billerica, Massachusetts) were dissolved in
(artificial cerebrospinal fluid [aCSF]) containing (in mmol/L) 126 dimethyl sulfoxide and used at 1/1000 working concentrations.
NaCl, 1.6 KCl, 1.1 NaH2PO4, 1.4 MgCl2, 26 NaHCO3, 11 glucose, 2.4 AG490 was protected from light throughout experiments. Signal
CaCl2 and incubated for a minimum of 45 min at 31.41 to 331C transduction inhibitors were preincubated with slices for a mini-
before recording. All solutions were continuously saturated with mum of 20 min before recording and were also present throughout
95% O2/5% CO2. the experiment. In some experiments, wortmannin (100 nmol/L) or
AG490 (50 mmol/L) was included in the patch pipette.
Electrophysiology
Slices were placed in the recording chamber and perfused Statistical Analysis
with aCSF with the addition of picrotoxin (100 mmol/L) to block All data are expressed as mean percent change in baseline
GABAA receptor-mediated inhibitory postsynaptic currents. Cells levels ⫾ SEM. To determine statistical significance, a section of
were visualized on an upright microscope using “Dodt-type” the baseline (average of 12 sweeps at minutes 6 through 7) was
gradient contrast infrared optics (31). Whole-cell voltage clamp compared with the maximal effect of drug treatment (average of
recordings were made using a Multiclamp 700B amplifier (Molecular 12 sweeps) using a paired t test. N refers to the number of cells
Devices, Union City, California). Recording electrodes (3–5 MO) recorded from a minimum of 3 rats.
were filled with (in mmol/L): 120 cesium methanesulfonate, 20 4-
(2-hydroxyethyl)-1-piperazineethanesulfonic acid, .4 ethylene glycol TH Immunocytochemistry
tetraacetic acid, 5 tetraethylammonium chloride, 2 MgCl2, 2.5 Brain slices from patch-clamp recording were fixed overnight
MgATP, .3 NaGTP, with a pH of 7.2 to 7.3 and 273 to 285 mOsm. in cold 4% paraformaldehyde, rinsed in PBS, blocked in 10%
Putative dopaminergic VTA neurons were identified by the presence normal donkey serum, incubated with monoclonal mouse anti-
of a large hyperpolarization-activated, cyclic nucleotide-regulated TH antibody (Sigma, 1:1000) for 48 hours at 41C. DyLight 594
cation (Ih) current (32–36). A subset of C57BL/6 neurons was streptavidin (Jackson ImmunoResearch Laboratories, West Grove,
identified by biocytin (.2%) filling and post hoc staining for tyrosine Pennsylvania; 1:200) was applied overnight at 41C. Secondary
hydroxylase (TH; discussed below). Of the 11 biocytin-filled neurons donkey anti-mouse fluorescein isothiocyanate antibody (1:50)
expressing Ih, only 2 did not express TH and were excluded from the was applied for 2 hours at 41C. Slices were mounted using
analysis. Dopamine neurons expressing Ih from Pitx3-GFP mice were Fluoromount (Sigma).
identified by GFP fluorescence. Of 15 VTA dopamine neurons
recorded from Pitx3-GFP mice, 3 cells were used for the experiments Results
with leptin on a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
receptor (AMPAR) EPSCs, 2 cells with wortmannin in the internal Leptin Suppresses Excitatory Synaptic Transmission onto
solution, 2 cells were used with wortmannin in the external solution, VTA Dopamine Neurons
1 cell for the paired pulse ratio, 3 cells for the miniature EPSCs AMPAR EPSCs were recorded from VTA dopamine neurons of
(mEPSCs) with leptin, and 3 cells for the mEPSCs with vehicle. To mouse midbrain slices voltage-clamped at –70 mV. Bath

www.sobp.org/journal
862 BIOL PSYCHIATRY 2013;73:860–868 J.L. Thompson and S.L. Borgland

Figure 1. Leptin depresses a-amino-


3-hydroxy-5-methyl-4-isoxazolepropionic
acid receptor (AMPAR)- and N-methyl-
D-aspartate receptor (NMDAR)-mediated
150 -200
synaptic transmission onto ventral teg-
AM PAR EPSCs (% Baseline)

mental area (VTA) dopamine neurons.

AM PAR EPSCs (pA)


125 -150 Evoked AMPAR (–70 mV) or NMDAR
(⫹40 mV) excitatory postsynaptic currents
(EPSCs) were recorded before, during,
100 -100 and after bath application of leptin
(100 nmol/mL). (A) Fifteen-minute bath
application of leptin caused a long-lasting
75 -50
decrease of AMPAR-mediated EPSCs.
100 nM leptin
(B) Example recording of leptin on evoked
50 0 AMPAR EPSCs from a VTA dopamine
0 10 20 30 40 0 10 20 30 40 neuron. Filled bar indicates presence of
Time (min) Time (min) leptin in the bath. Inset, representative
traces of AMPAR EPSCs before (black) and
after leptin (gray). Scale bars, 10 msec,
100 pA. (C) Fifteen minute bath applica-
150 200 tion of leptin caused a lasting decrease in
NMDAR EPSCs (% baseline)

NMDAR-mediated EPSCs of VTA dopamine


neurons. (D) Example recording of leptin
NM DAR EPSCs (pA)

125 150 on evoked NMDAR EPSCs from a dopa-


mine neuron in the VTA. Filled bar indi-
cates presence of leptin in the bath. Inset,
100 100
representative traces of NMDAR EPSCs
before (black) and after leptin (gray). Scale
75 50 bars: 20 msec, 100 pA. Stimulus artifacts
have been removed for clarity. Error bars
100 nM leptin indicate SEM.
50 0
0 10 20 30 40 0 10 20 30 40
Time (min) Time (min)

application of leptin for 15 min caused a long-lasting depression A reduction of mEPSCs frequency concomitant with unchanged
of AMPAR EPSCs (Figure 1A,B; baseline: 98% ⫾ 1% vs. leptin: amplitude likely reflects a decrease in the probability of presynaptic
77% ⫾ 6%; n ¼ 7, p ⬍ .05). Leptin depressed AMPAR EPSCs in all neurotransmitter release (38). To further verify whether leptin-
four neurons that were post hoc labeled for TH. To measure mediated depression of AMPARs was mediated pre- or postsynap-
NMDAR EPSCs, neurons were voltage clamped at ⫹40 mV and tically, we examined the effects of leptin on the probability of
measurements were taken 20 ms after the stimulus artifact, transmitter release, comparing the response to paired pulses, a
a time point at which the glutamatergic EPSC is mediated measure that changes in a highly predictable fashion with release
primarily by NMDARs (14). Leptin caused a long-lasting depres- probability. We recorded AMPAR EPSCs at –70 mV using a paired-
sion of NMDAR EPSCs (Figure 1C,D; baseline: 100% ⫾ 2% vs. pulse stimulation protocol with a 50 msec interval and observed
leptin: 77% ⫾ 7%; n ¼ 6, p ⬍ .05). The maximum effect of leptin significant paired pulse facilitation after leptin bath application
on AMPAR or NMDAR was not significantly different (p ⬎ .05). (Figure 2F,G; n ¼ 6, p ⬍ .05). Taken together, these data suggest
Furthermore, of the 9 biocytin-labeled neurons included in the that leptin-induced synaptic depression is consistent with a reduc-
analysis, leptin depressed AMPAR EPSCs in five neurons and tion in the probability of presynaptic glutamate release.
NMDAR in three neurons expressing TH, suggesting that leptin
inhibition of glutamatergic synaptic transmission can occur onto Leptin-Induced Synaptic Depression Occurs by Activation
VTA dopamine neurons. of the Jak2–PI3K Pathway
Because leptin suppressed AMPAR or NMDARs with equal LepRb activation triggers three main signal transduction
magnitude, leptin may be decreasing glutamate release. Therefore, cascades upon association with Janus kinase 2 (Jak2) tyrosine
to investigate whether leptin altered the number or function of kinase. First, Tyr(985) of LepRb recruits SH2-containing tyrosine
postsynaptic AMPARs or caused a presynaptic inhibition of gluta- phosphatase (SHP-2) resulting ERK activation. Second, Tyr(1138)
mate release, we measured AMPAR-mediated miniature EPSCs of LepRb recruits signal transducer and activator of transcription
(mEPSCs), a standard method for determining the locus of synaptic 3 (STAT3). Third, tyrosine phosphorylation sites on the receptor-
change (37). Fifteen to 18 minutes after bath application of leptin associated Jak2 leads to activation of phosphatidylinositol 3-
(100 nmol/L, 15 min), mEPSCs frequency was significantly reduced kinase (PI3K) (3).
(1.2 ⫾ .3 Hz) compared with baseline mEPSCs (Figure 2A–D; To determine whether Jak2 signaling was required for leptin-
1.9 ⫾ .4 Hz; n ¼ 12, p ⬍ .05). This effect was not due to rundown induced synaptic depression, we bath applied the Jak2 inhibitor,
over time because there was no significant difference between the AG-490 (50 mmol/L) (39) to midbrain slices. Bath application
baseline and vehicle treatment (Figure 2A–2D; baseline: 1.7 ⫾ .3 Hz of AG-490 significantly inhibited leptin-induced depression of
vs. vehicle: 1.7 ⫾ .3 Hz, n ¼ 12, p ⬎ .05). Leptin did not alter mEPSC AMPAR EPSCs (Figure 3A,B; leptin: 100% ⫾ 6%, n ¼ 7, compared
amplitude (Figure 2B,C,E; baseline: 16.6 ⫾ .9 pA vs. leptin: 16.0 ⫾ .9 with maximum leptin response from Figure 1A (77% ⫾ 6%),
pA, n ¼ 12, p ⬎ .05) nor were there changes in mEPSC amplitude p ⬍ .05).
after vehicle application (baseline: 18.6 ⫾ 1.1 pA vs. vehicle: Leptin-induced synaptic depression may be due to activation
17.9 ⫾ .9 pA, n ¼ 12, p ⬎ .05). of postsynaptic leptin receptors on dopamine neurons, leading to

www.sobp.org/journal
J.L. Thompson and S.L. Borgland BIOL PSYCHIATRY 2013;73:860–868 863

Figure 2. Leptin-induced synaptic inhibi-


3 30 tion is mediated presynaptically. a-amino-

mEPSC peak amplitude (pA)


* 3-hydroxy-5-methyl-4-isoxazolepropionic
mEPSC fre que ncy (Hz)

acid receptor (AMPAR) miniature excita-


2 20 tory postsynaptic currents (mEPSCs) were
recorded at –70 mV in the presence of
picrotoxin, TTX and AP-5 before and after
a 15 min bath application of leptin
1 10 (100 nmol/L). (A) Frequency of mEPSCs
events was significantly decreased after
leptin application (filled bars) compared
with baseline (open bars, n ¼ 8, p ⬍ .05).
0 0
Leptin Vehicle Leptin Vehicle
Vehicle treatment (filled bar) did not alter
mEPSC frequency compared to baseline
(p ⬎ .05). (B) AMPAR mEPSCs amplitude
Baseline Baseline was not significantly different before
(open bar) or after (filled bars) bath appli-
cation of leptin (right panel) or vehicle (left
Leptin Vehicle panel; p ⬎ .05). (C) Example recordings of
AMPAR mEPSCs before or after leptin (left
side) or vehicle (right side) application.
Scale bars: 100 msec, 20 pA. (D) Cumula-
1.0
tive probability plots for interevent interval
1.0
for before (black) and after (gray) leptin
Cumulative Proba bility

application. A significant right-shift in the


Cumulative Proba bility

cumulative probability of mEPSC fre-


quency was detected after leptin applica-
0.5
tion compared to the baseline (p ⬍ .001,
0.5
Baseline Kolmogorov–Smirnov test). (E) Cumulative
Leptin probability plots for amplitude for before
Baseline (VEH) (black) and after (gray) leptin application.
Vehicle No significant difference was detected in
cumulative probability plots of mEPSC
0.0 0.0
0 10 20 30 40 50 60
amplitude before or after leptin (p ⬎ .05,
0 1 2 3 4 5
Kolmogorov-Smirnov test). (F) Bar graph
inter-event interval (s) mEPSC peak amplitude (pA)
illustrating a significant increase in the
paired pulse ratio after leptin administra-
* tion (*p ⬍ .05). (G) Example recording of
1.0 paired (50 msec interval) AMPAR EPSCs
before (black) and after leptin (gray) appli-
Paired Pulse Ratio (P2/P1)

0.8 cation. Scale bars: 20 msec, 50 pA. Stimu-


lus artifact has been removed for clarity.
0.6 Bars represent mean ⫾ SEM. VEH, vehicle.

0.4

0.2

0.0
Baseline Leptin

presynaptic inhibition of glutamate release via retrograde med- (100 nmol/L) (5,13) abolished the leptin-induced synaptic depression
iators. Another possibility is that activation of presynaptic LepRbs (Figure 4C,D; baseline: 102% ⫾ 2% vs. leptin: 96% ⫾ 5%, n ¼ 7,
induces a suppression of glutamate release. To investigate if p ⬎ .05). Furthermore, with postsynaptic inhibition of PI3K, leptin
postsynaptic receptor activation was required for leptin-induced significantly depressed AMPAR EPSCs (Figure 4E,F; baseline:
synaptic depression, we applied AG-490 via the patch pipette. 102% ⫾ 2% vs. leptin: 82% ⫾ 6%; n ¼ 7, p ⬍ .05). These data
Leptin-induced a significant synaptic depression of AMPAR- suggest that leptin likely activates presynaptic LepRbs to inhibit
mediated synaptic transmission with intracellular inhibition of glutamate release and that leptin activation of Jak2-PI3K is necessary
Jak2 in dopamine neurons (Figure 3C,D, baseline: 99% ⫾ 4% vs. to induce a reduction in synaptic efficacy of VTA dopamine neuron
leptin: 78% ⫾ 7%; n ¼ 5, p ⬍ .05). inputs.
To test the involvement of the SHP-2/ERK pathway, we pre- Discussion
incubated slices with U0126 (10 mmol/L) (13) a MEK2 inhibitor
upstream of ERK activation. In the presence of U0126, leptin The data presented here establish a novel mechanism for
significantly depressed AMPAR-mediated EPSCs (Figure 4A,B, base- leptin action in the mesolimbic incentive motivation/reward
line: 100% ⫾ 2% vs. leptin 80% ⫾ 5%, n ¼ 5, p ⬍ .05). These data system. Leptin reduced both NMDAR- and AMPAR-mediated
indicate that unlike leptin suppression of firing rate (13), leptin- synaptic transmission onto dopamine neurons by reducing
induced synaptic depression is via a different signaling pathway. We glutamate release from presynaptic terminals. This effect was
next tested the involvement of the Jak2/PI3K pathway using the mediated by presynaptic Jak2 and PI3K signaling, suggesting that
PI3K inhibitor, wortmannin. Extracellular application of wortmannin leptin is not directly activating dopamine neurons to induce

www.sobp.org/journal
864 BIOL PSYCHIATRY 2013;73:860–868 J.L. Thompson and S.L. Borgland

Figure 3. Leptin-induced synaptic inhibi-


tion requires PI3K activation. Evoked
a-amino-3-hydroxy-5-methyl-4-isoxazole-
propionic acid receptor (AMPAR) excitatory
postsynaptic currents (EPSCs) were recorded
before, during, and after bath application of
leptin (100 nmol/L) for 15 min. Slices were
preincubated in inhibitors for a minimum
20 min before and throughout the experi-
ment. Filled bars indicate the presence of
leptin in the bath. (A) Bath application of
AG490 (50 mmol/L) abolished leptin-induced
synaptic depression. (B) Example recording
of AMPAR EPSCs from a representative
dopamine neuron in the presence of
AG490 and leptin. Inset, example traces
before (black) and after (gray) leptin applica-
tion. (C) AG490 (50 mmol/L) applied intracel-
lularly did not inhibit leptin-induced synaptic
depression. (D) Example recording of
AMPAR EPSCs before and after leptin appli-
cation in the presence of intracellular AG-
490. Inset, example traces before (black) and
after (gray) leptin application. Scale bars: 20
msec, 100 pA. Stimulus artifacts have been
removed for clarity. Error bars indicate SEM.

synaptic inhibition. A model of leptin inhibition of excitatory Several lines of evidence suggest that leptin modulates reward-
synaptic transmission onto VTA dopamine neurons is illustrated seeking behavior. Intra-VTA leptin reduced the reward value of
in Figure 5. sucrose via a reduction in dopamine neuronal activity (11), intra-VTA
Previous studies have implicated leptin action in the VTA in or intracerebroventricular leptin suppressed the rewarding effects of
reducing food intake (5,40,41). Interestingly, Morton et al. (40) intracranial self-stimulation (41), intracerebroventricular leptin
demonstrated that leptin’s effect on food intake was not sensitive attenuates acute food deprivation-induced relapse to drug seeking
to PI3K inhibition, suggesting that our results may dissociate (40) and reverses conditioned place preference for sucrose (49) or
from the effects of leptin on food intake. To inhibit PI3K, this high fat foods (41). Furthermore, firing rate of putative dopamine
study used a concentration of LY-294002 (2 mmol/L) intra-VTA, neurons is decreased after intravenous leptin administration or bath
which may have several off-target effects including action at application of leptin to midbrain slices (5,13). Studies using leptin
several ion channels (42–44), phosphodiesterases (45), and the deficient ob/ob mice have provided conflicting results. Fulton et al.
estrogen receptor (46), potentially confounding the feeding (6) reported that the baseline and electrically evoked release of
results. Furthermore, it is important to consider that leptin acting dopamine in the nucleus accumbens was significantly reduced in
on GABAergic inputs to VTA neurons (excluded in our study) may ob/ob mice. In contrast, Rosenberry et al. (50) demonstrated that
play a role in feeding. Indeed, leptin potentiates inhibitory basal or cocaine-induced dopamine levels in the nucleus accum-
synaptic transmission in the hippocampus (47). Future studies bens was increased in ob/ob mice when measured via microdialysis.
will be aimed at directly testing leptin modulation of GABAergic This discrepancy is likely due to technical differences such as the use
inputs to VTA dopamine neurons. Interestingly, knocking down of in vivo microdialysis (50) compared with amperometry in
LepRb expression within the VTA (using short hairpin RNA forebrain slices (6). Amphetamine-induced locomotion and sensiti-
[shRNA]) or knocking out LepRb on VTA dopamine neurons zation was reduced in ob/ob mice compared with wildtype mice (6).
yielded differential effects with regards to feeding. Although However, basal locomotor activity is lower in ob/ob mice compared
neither method alters body weight, shRNA to LepRb in VTA with wildtype mice, potentially confounding these results (50–54).
increased consumption of high fat food and increased the work Taken together, these studies suggest that leptin in the VTA reduces
required to obtain sucrose (5,10). In contrast, there was no activity of dopamine neurons and modulates reward seeking
difference in regular or high fat food intake in mice with a behavior. Our findings that leptin reduces excitatory synaptic
conditional LepRb knockout in dopamine transporter expressing transmission onto dopamine neurons are consistent with these
neurons compared with wildtype mice (48). Possible discrepan- studies and suggest that in the constitutive presence of leptin,
cies between these studies may be because LepRb is removed selective glutamatergic afferents to the VTA are tonically dampened.
only from dopamine neurons of the substantia nigra and VTA in Leptin can modulate excitatory synaptic transmission in other
the conditional knockout mice, whereas the LepRb shRNA would brain regions. For example, mice with mutations in LepRb (db/db
target dopamine as well as GABAergic neurons only in the VTA. mice or fa/fa rats) have decreased LTP or LTD of hippocampal
Furthermore, conditional LepRb mice are missing LepRbs neurons (55). Leptin bath applied to hippocampal slices facilitates
throughout development, possibly leading to compensatory LTP (56), likely because of leptin-induced forward trafficking on
changes. NMDARs to the synapse (26). Additionally, leptin is reported

www.sobp.org/journal
J.L. Thompson and S.L. Borgland BIOL PSYCHIATRY 2013;73:860–868 865

Figure 4. Leptin-induced synaptic inhibi-


tion requires PI3K activation. Evoked
a-amino-3-hydroxy-5-methyl-4-isoxazole-
propionic acid receptor (AMPAR) excita-
tory postsynaptic currents (EPSCs) were
recorded before, during, and after bath
application of leptin (100 nmol/L) for 15
min. Slices were preincubated in inhibitors
for a minimum 20 min before and
throughout the experiment. Filled bars
indicate the presence of leptin in the bath.
(A) U0126 (10 mmol/L) does not alter
leptin-induced depression of AMPAR
EPSCs of ventral tegmental area dopamine
neurons. (B) Example recording of AMPAR
EPSCs from a representative dopamine
neuron in the presence of U0126 and
leptin. (C) Bath application of wortmannin
(100 nmol/L) abolished leptin-induced
synaptic depression. (D) Example record-
ing of AMPAR EPSCs from a representative
dopamine neuron in the presence of
wortmannin and leptin. Inset, example
traces before (black) and after (gray) leptin
application. Scale bars: 20 msec, 100 pA.
(E) Wortmannin (100 nmol/L) applied
intracellularly did not inhibit leptin-
induced synaptic depression. (F) Example
recording of AMPAR EPSCs before and
after leptin application in the presence of
intracellular wortmannin. Inset, example
traces before (black) and after (gray) leptin
application. Scale bars: 20 msec, 100 pA.
Stimulus artifacts have been removed for
clarity. Error bars indicate SEM.

to preferentially enhance NR2B-mediated NMDAR responses in the effect of leptin, suggesting that LepRb activation does not
cerebellar granule cells (28). Thus, unlike the postsynaptically occur directly on dopamine neurons.
mediated alteration of glutamatergic synapses in the hippocam- Several studies have demonstrated that there is high LepRb
pus or cerebellum described earlier, leptin action in the VTA expression in the VTA using immunohistochemistry to LepRb
caused a presynaptic suppression of glutamate release. protein in rats (57); fluorescent in situ hybridization to LepRb
Interestingly, leptin-induced synaptic depression was not due messenger RNA colocalized with TH messenger RNA in rats (5)
to a direct effect of leptin on dopamine neurons. Several lines of and immunohistochemistry to leptin activated pSTAT3 colocaliza-
evidence support our conclusion that depression of AMPAR- tion with TH in mice (6). However, in mice expressing enhanced
mediated EPSCs by leptin was mediated presynaptically. First, green fluorescent protein (EGFP) in LepRb-containing neurons, only
leptin depressed NMDAR-mediated EPSCs with a similar time approximately 6% overlapped with TH expression in the VTA (8).
course and maximum to that of AMPAR EPSCs, suggesting Low promoter activity driving EGFP expression in the LepRbEGFP
reduced availability of synaptic glutamate to activate either mice may account for some of this discrepancy; however, it is clear
glutamate receptor subtype as opposed to a postsynaptic that LepRb expression on dopamine neurons is less than originally
decrease in number or function of both receptors. Second, described. Our results are consistent with the idea that leptin
decreased mEPSC frequency concomitant with unchanged mEPSC control of the mesolimbic dopamine system occurs indirectly.
amplitude suggests that leptin reduced synaptic glutamate with- Leptin acting at LepRb expressed on presynaptic glutamatergic
out affecting the number or function of AMPARs on dopaminergic terminals in the VTA may be one mechanism through which leptin
neurons. Third, leptin induced paired-pulse facilitation suggesting inhibits excitatory synaptic transmission. Terminals expressing LepR-
a decreased release probability. Finally, while postsynaptic inhibi- bEGFP in the VTA include those from the lateral hypothalamus (LH),
tion of Jak2 or PI3K did not alter leptin-induced synaptic periaqueductal gray area, and hypothalamic preoptic area (8).
depression, bath application of AG490 or wortmannin abolished Whereas POA neurons are mainly inhibitory (58,59), periaqueductal

www.sobp.org/journal
866 BIOL PSYCHIATRY 2013;73:860–868 J.L. Thompson and S.L. Borgland

Figure 5. Putative mechanisms of leptin-


induced synaptic depression onto ventral
tegmental area (VTA) dopamine neurons.
STAT3 Leptin may act via presynaptic LepRbs on
glutamatergic terminals. Inhibition of Jak2
STAT3 and PI3K, but not MEK1/2, could block the
leptin-induced reduction in presynaptic
LepRb glutamate release. Leptin may also act
U0126 via PI3K-dependent upregulation of glu-
tamate transporters (i.e., EAAT4) to
MEK1/2 K2 increase clearance of glutamate from the
JA
wortmannin K2 synapse.
JA
PI3-K
? ? AG490
glutamate

EAAT4

VTA dopamine neuron

gray area neurons provide glutamatergic input to the VTA (60). leptin-induced suppression of firing was mediated by direct or
Interestingly, activation of LepRb-expressing neurons in the LH indirect depolarization of dopamine neurons, it does suggest that
modulates dopamine production and content in the VTA, con- leptin-modulation of firing rate is mediated by an independent
tributing to a decrease in incentive value of food (61). However, mechanism to leptin-induced synaptic inhibition.
LepRb expression was mainly on LH GABAergic neurons. One interesting hypothesis for the mechanism underlying a
Although it is possible that LepRb-containing LH GABAergic leptin-induced synaptic inhibition is that leptin may increase
terminals within the VTA synapse onto glutamatergic afferents glutamate clearance from the synapse by upregulating transpor-
to control glutamate release, this effect is unlikely to occur in our ter function. Recent evidence demonstrated that leptin activation
slices because of the presence of picrotoxin in all extracellular of Jak2 led to upregulation of Na⫹/K⫹-dependent excitatory
solutions. Another possibility is that leptin activates LepRb on amino acid transporters (EAATs), particularly EAAT2 and EAAT4
somata outside the VTA in our horizontal midbrain slices. These (63). Consistent with this, PI3K/Akt signaling has been implicated
slices contain a significant portion of the posterior hypothalamic in forward trafficking of both EAAT2 and EAAT4 (64,65). Interest-
area, a region with dense LepRb expression (2). Therefore, it is ingly, in addition to astrocytic EAAT4 expression, EAAT4 is
feasible that leptin could activate receptors on cell bodies in densely expressed on neurons within the VTA (66). Notably,
these regions that could project directly or indirectly to the VTA, leptin caused modest paired pulse facilitation in our experiments,
thus inhibiting glutamate release. It should be noted that suggesting that while synaptic glutamate release was depressed,
differences in leptin signaling and receptor expression levels the probability of release was less affected, consistent with an
may differ between young mice used in this study and adult increase in glutamate reuptake. Thus, leptin-induced synaptic
animals used in other studies. However, location of receptor depression may be due to an upregulation of glutamate
expression is not expected to change with age. Furthermore, clearance in the VTA.
others have reported consistent effects of leptin in the VTA of Increased excitatory synaptic transmission onto dopamine
adult and 3 week old mice (13). Taken together, leptin likely neurons is associated with learning of cues predicting food
activates LepRbs on glutamatergic inputs directly or indirectly reward (67). A reduction in excitatory synaptic transmission
projecting to the VTA. may raise the threshold required for burst firing of dopamine
Interestingly, activation of PI3K, but not ERK, was required for neurons (17). Because burst firing has been implicated in
leptin-induced synaptic depression in the VTA. Leptin-induced promoting dopamine release and driving salience of food related
activation of Jak2 recruits insulin receptor substrate-1 (IRS-1), cues, a reduction of salience to food-related cues may be a
leading to PI3K activation. Inhibition of PI3K by the potent functional consequence of leptin-induced inhibition of glutamate
inhibitor wortmannin blocked leptin-induced synaptic depression release. This idea is consistent with studies demonstrating a
only when applied in the bath solution. In hippocampal cultures, leptin-induced reduction in hedonic food intake (11) and reduced
leptin caused a PI3K-dependent forward trafficking of postsynap- motivation to obtain food (10). Because leptin’s action was
tic GluA1 subunits (62). This discrepancy is likely because in VTA, selective to presynaptic glutamatergic inputs to the VTA, it is
leptin inhibition of excitatory synaptic transmission is mediated possible that circulating leptin may tonically dampen select
presynaptically. Leptin-induced inhibition of firing of VTA neu- inputs to the VTA, thereby reducing their influence to promote
rons was mediated by ERK signaling as bath application of the burst firing of dopamine neurons. Fasting decreases circulating
MEK inhibitor U0126 abolished leptin-induced suppression of leptin (68); therefore, one can speculate that tonic suppression
firing rate (13). Although these experiments did not test whether of glutamatergic inputs to the VTA would be relieved in

www.sobp.org/journal
J.L. Thompson and S.L. Borgland BIOL PSYCHIATRY 2013;73:860–868 867

this situation, resulting in heightened salience to food-related firing pattern in midbrain dopamine neurons. J Neurophysiol 91:
cues. 346–357.
Here, we propose a novel mechanism for leptin signaling in the 19. Borgland SL, Taha SA, Sarti F, Fields HL, Bonci A (2006): Orexin a in the
VTA is critical for the induction of synaptic plasticity and behavioral
VTA that is, to our knowledge, the first report of a presynaptic
sensitization to cocaine. Neuron 49:589–601.
inhibition of excitatory synaptic transmission induced by leptin. 20. Borgland SL, Chang SJ, Bowers MS, Thompson JL, Vittoz N, Floresco SB,
Leptin-induced synaptic inhibition in the VTA is dependent on et al. (2009): Orexin a/hypocretin-1 selectively promotes motivation for
activation of PI3K of nondopaminergic neurons. Taken together, this positive reinforcers. J Neurosci 29:11215–11225.
study provides new insight for how leptin modulates the mesolimbic 21. Abizaid A, Liu ZW, Andrews ZB, Shanabrough M, Borok E, Elsworth JD,
dopamine system to suppress reward-seeking behaviors. et al. (2006): Ghrelin modulates the activity and synaptic input
organization of midbrain dopamine neurons while promoting appe-
tite. J Clin Invest 116:3229–3239.
This work was supported by an Natural Sciences and Engineer- 22. Thompson JL, Borgland SL (2011): A role for hypocretin/orexin in
ing Research Council discovery grant and a Canadian Institutes of motivation. Behav Brain Res 217:446–453.
Health Research new investigator award to SLB. We thank Haiyan 23. Blum ID, Patterson Z, Khazall R, Lamont EW, Sleeman MW, Horvath TL,
Zou for breeding the Pitx3 mice. et al. (2009): Reduced anticipatory locomotor responses to scheduled
The authors have no biomedical financial interests or potential meals in ghrelin receptor deficient mice. Neuroscience 164:351–359.
24. Moult PR, Milojkovic B, Harvey J (2009): Leptin reverses long-term
conflicts of interest. potentiation at hippocampal CA1 synapses. J Neurochem 108:
685–696.
1. Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM (1996): Leptin 25. Durakoglugil M, Irving AJ, Harvey J (2005): Leptin induces a novel
enters the brain by a saturable system independent of insulin. form of NMDA receptor-dependent long-term depression. J Neuro-
Peptides 17:305–311. chem 95:396–405.
2. Elmquist JK, Bjorbaek C, Ahima RS, Flier JS, Saper CB (1998): 26. Shanley LJ, Irving AJ, Harvey J (2001): Leptin enhances NMDA
Distributions of leptin receptor mRNA isoforms in the rat brain. receptor function and modulates hippocampal synaptic plasticity. J
J Comp Neurol 395:535–547. Neurosci 21:RC186.
3. Oswal A, Yeo G (2010): Leptin and the control of body weight: A 27. Glaum SR, Hara M, Bindokas VP, Lee CC, Polonsky KS, Bell GI, et al.
review of its diverse central targets, signaling mechanisms, and role (1996): Leptin, the obese gene product, rapidly modulates synaptic
in the pathogenesis of obesity. Obesity (Silver Spring) 18:221–229. transmission in the hypothalamus. Mol Pharmacol 50:230–235.
4. Munzberg H, Myers MG Jr (2005): Molecular and anatomical determi- 28. Irving AJ, Wallace L, Durakoglugil D, Harvey J (2006): Leptin enhances
nants of central leptin resistance. Nat Neurosci 8:566–570. NR2b-mediated N-methyl-D-aspartate responses via a mitogen-
5. Hommel JD, Trinko R, Sears RM, Georgescu D, Liu ZW, Gao XB, et al. activated protein kinase-dependent process in cerebellar granule
(2006): Leptin receptor signaling in midbrain dopamine neurons cells. Neuroscience 138:1137–1148.
regulates feeding. Neuron 51:801–810. 29. Zhao S, Maxwell S, Jimenez-Beristain A, Vives J, Kuehner E, Zhao J,
6. Fulton S, Pissios P, Manchon RP, Stiles L, Frank L, Pothos EN, et al. et al. (2004): Generation of embryonic stem cells and transgenic mice
(2006): Leptin regulation of the mesoaccumbens dopamine pathway. expressing green fluorescence protein in midbrain dopaminergic
Neuron 51:811–822. neurons. Eur J Neurosci 19:1133–1140.
7. Figlewicz DP, Evans SB, Murphy J, Hoen M, Baskin DG (2003): 30. Paxinos G, Watson C (1998): The Rat Brain in Stereotaxic Coordinates,
Expression of receptors for insulin and leptin in the ventral tegmental 4th Ed. San Diego, CA: Academic Press.
area/substantia nigra (VTA/SN) of the rat. Brain Res 964:107–115. 31. Dodt HU, Eder M, Schierloh A, Zieglgansberger W (2002): Infrared-
8. Leshan RL, Opland DM, Louis GW, Leinninger GM, Patterson CM, guided laser stimulation of neurons in brain slices. Sci STKE 2002:l2.
Rhodes CJ, et al. (2010): Ventral tegmental area leptin receptor 32. Grace AA, Onn SP (1989): Morphology and electrophysiological
neurons specifically project to and regulate cocaine- and amphetamine- properties of immunocytochemically identified rat dopamine neurons
regulated transcript neurons of the extended central amygdala. recorded in vitro. J Neurosci 9:3463–3481.
J Neurosci 30:5713–5723. 33. Lacey MG, Mercuri NB, North RA (1990): Actions of cocaine on rat
9. Berridge KC (2012): From prediction error to incentive salience: dopaminergic neurones in vitro. Br J Pharmacol 99:731–735.
Mesolimbic computation of reward motivation. Eur J Neurosci 35: 34. Johnson SW, North RA (1992): Two types of neurone in the rat ventral
1124–1143. tegmental area and their synaptic inputs. J Physiol 450:455–468.
10. Davis JF, Choi DL, Schurdak JD, Fitzgerald MF, Clegg DJ, Lipton JW, 35. Cameron DL, Wessendorf MW, Williams JT (1997): A subset of ventral
et al. (2011): Leptin regulates energy balance and motivation through tegmental area neurons is inhibited by dopamine, 5-hydroxytryptamine
action at distinct neural circuits. Biol Psychiatry 69:668–674. and opioids. Neuroscience 77:155–166.
11. Domingos AI, Vaynshteyn J, Voss HU, Ren X, Gradinaru V, Zang F, 36. Ford CP, Mark GP, Williams JT (2006): Properties and opioid inhibition
et al. (2011): Leptin regulates the reward value of nutrient. Nat of mesolimbic dopamine neurons vary according to target location.
Neurosci 14:1562–1568. J Neurosci 26:2788–2797.
12. Narayanan NS, Guarnieri DJ, DiLeone RJ (2010): Metabolic hormones, 37. Malenka RC, Nicoll AR (1999): Long-term potentiation—a decade of
dopamine circuits, and feeding. Front Neuroendocrinol 31:104–112. progress? Science 285:1870–1874.
13. Trinko R, Gan G, Gao XB, Sears RM, Guarnieri DJ, DiLeone RJ (2011): 38. Katz B (1971): Quantal mechanism of neural transmitter release.
ERK1/2 mediates leptin receptor signaling in the ventral tegmental Science 173:123–126.
area. PLoS One 6:e27180. 39. Jo YH, Chen YJ, Chua SC Jr, Talmage DA, Role LW (2005): Integration
14. Bonci A, Malenka RC (1999): Properties and plasticity of excitatory of endocannabinoid and leptin signaling in an appetite-related neural
synapses on dopaminergic and GABAergic cells in the ventral circuit. Neuron 48:1055–1066.
tegmental area. J Neurosci 19:3723–3730. 40. Morton GJ, Blevins JE, Kim F, Matsen M, Figlewicz DP (2009): The
15. Overton PG, Richards CD, Berry MS, Clark D (1999): Long-term action of leptin in the ventral tegmental area to decrease food intake
potentiation at excitatory amino acid synapses on midbrain dopa- is dependent on Jak-2 signaling. Am J Physiol Endocrinol Metab 297:
mine neurons. Neuroreport 10:221–226. E202–E210.
16. Chergui K, Suaud-Chagny MF, Gonon F (1994): Nonlinear relationship 41. Bruijnzeel AW, Corrie LW, Rogers JA, Yamada H (2011): Effects of
between impulse flow, dopamine release and dopamine elimination insulin and leptin in the ventral tegmental area and arcuate
in the rat brain in vivo. Neuroscience 62:641–645. hypothalamic nucleus on food intake and brain reward function in
17. Overton PG, Clark D (1997): Burst firing in midbrain dopaminergic female rats. Behav Brain Res 219:254–264.
neurons. Brain Res Brain Res Rev 25:312–334. 42. Welling A, Hofmann F, Wegener JW (2005): Inhibition of l-type CAV1.2
18. Komendantov AO, Komendantova OG, Johnson SW, Canavier CC CA2⫹ channels by 2,(4-morpholinyl)-8-phenyl-4h-1-benzopyran-4-
(2004): A modeling study suggests complementary roles for GABAA one (ly294002) and 2-[1-(3-dimethyl-aminopropyl)-5-methoxyindol-3-
and NMDA receptors and the SK channel in regulating the yl]-3-(1h-indol-3-yl) maleimide (gö6983). Mol Pharmacol 67:541–544.

www.sobp.org/journal
868 BIOL PSYCHIATRY 2013;73:860–868 J.L. Thompson and S.L. Borgland

43. Sun H, Oudit GY, Ramirez RJ, Costantini D, Backx PH (2004): The 56. Wayner MJ, Armstrong DL, Phelix CF, Oomura Y (2004): Orexin-a
phosphoinositide 3-kinase inhibitor ly294002 enhances cardiac myo- (hypocretin-1) and leptin enhance LTP in the dentate gyrus of rats
cyte contractility via a direct inhibition of ik,slow currents. Cardiovasc in vivo. Peptides 25:991–996.
Res 62:509–520. 57. Figlewicz DP, Bennett J, Evans SB, Kaiyala K, Sipols AJ, Benoit SC
44. Wu J, Ding WG, Matsuura H, Tsuji K, Zang WJ, Horie M (2009): (2004): Intraventricular insulin and leptin reverse place preference
Inhibitory actions of the phosphatidylinositol 3-kinase inhibitor conditioned with high-fat diet in rats. Behav Neurosci 118:
ly294002 on the human kv1.5 channel. Br J Pharmacol 156:377–387. 479–487.
45. Abbott BM, Thompson PE (2004): Pde2 inhibition by the PI3 kinase 58. Maeda H, Mogenson GJ (1980): An electrophysiological study of
inhibitor ly294002 and analogues. Bioorg Med Chem Lett 14: inputs to neurons of the ventral tegmental area from the nucleus
2847–2851. accumbens and medial preoptic-anterior hypothalamic areas. Brain Res
46. Pasapera Limón AM, Herrera-Muñoz J, Gutiérrez-Sagal R, Ulloa-Aguirre A 197:365–377.
(2003): The phosphatidylinositol 3-kinase inhibitor ly294002 binds the 59. Luo AH, Aston-Jones G (2009): Circuit projection from suprachias-
estrogen receptor and inhibits 17b-estradiol-induced transcriptional matic nucleus to ventral tegmental area: A novel circadian output
activity of an estrogen sensitive reporter gene. Mol Cell Endocrinol 200: pathway. Eur J Neurosci 29:748–760.
199–202. 60. Omelchenko N, Sesack SR (2010): Periaqueductal gray afferents
47. Solovyova N, Moult PR, Milojkovic B, Lambert JJ, Harvey J (2009): synapse onto dopamine and GABA neurons in the rat ventral
Bi-directional modulation of fast inhibitory synaptic transmission by tegmental area. J Neurosci Res 88:981–991.
leptin. J Neurochem 108:190–201. 61. Leinninger GM, Jo YH, Leshan RL, Louis GW, Yang H, Barrera JG, et al.
48. Liu J, Perez SM, Zhang W, Lodge DJ, Lu XY (2011): Selective deletion (2009): Leptin acts via leptin receptor-expressing lateral hypothalamic
of the leptin receptor in dopamine neurons produces anxiogenic-like neurons to modulate the mesolimbic dopamine system and suppress
behavior and increases dopaminergic activity in amygdala. Mol feeding. Cell Metab 10:89–98.
Psychiatry 16:1024–1038. 62. Moult PR, Cross A, Santos SD, Carvalho AL, Lindsay Y, Connolly CN,
49. Figlewicz DP, Higgins MS, Ng-Evans SB, Havel PJ (2001): Leptin et al. (2010): Leptin regulates AMPA receptor trafficking via PTEN
reverses sucrose-conditioned place preference in food-restricted rats. inhibition. J Neurosci 30:4088–4101.
Physiol Behav 73:229–234. 63. Hosseinzadeh Z, Bhavsar SK, Sopjani M, Alesutan I, Saxena A,
50. Roseberry AG, Painter T, Mark GP, Williams JT (2007): Decreased Dermaku-Sopjani M, et al. (2011): Regulation of the glutamate
vesicular somatodendritic dopamine stores in leptin-deficient mice. transporters by Jak2. Cell Physiol Biochem 28:693–702.
J Neurosci 27:7021–7027. 64. Bohmer C, Philippin M, Rajamanickam J, Mack A, Broer S, Palmada M, et al.
51. Dauncey MJ (1986): Activity-induced thermogenesis in lean and (2004): Stimulation of the EAAT4 glutamate transporter by SGK protein
genetically obese (ob/ob) mice. Cell Mol Life Sci 42:547–549. kinase isoforms and PKB. Biochem Biophys Res Commun 324:1242–1248.
52. Calcagnetti DJ, Flynn JJ, Margules DL (1987): Opioid-induced linear 65. Wu X, Kihara T, Akaike A, Niidome T, Sugimoto H (2010): PI3K/AKT/
running in obese (ob/ob) and lean mice. Pharmacol Biochem Behav 26: mTOR signaling regulates glutamate transporter 1 in astrocytes.
743–747. Biochem Biophys Res Commun 393:514–518.
53. Dauncey MJ, Brown D (1987): Role of activity-induced thermogenesis 66. Massie A, Cnops L, Smolders I, McCullumsmith R, Kooijman R, Kwak S,
in twenty-four hour energy expenditure of lean and genetically obese et al. (2008): High-affinity Na⫹/K⫹-dependent glutamate transporter
(ob/ob) mice. Exp Physiol 72:549–559. EAAT4 is expressed throughout the rat fore- and midbrain. J Comp
54. Collin M, Håkansson-Ovesjö M-L, Misane I, Ögren SO, Meister B (2000): Neurol 511:155–172.
Decreased 5-HT transporter mRNA in neurons of the dorsal raphe 67. Stuber GD, Klanker M, de Ridder B, Bowers MS, Joosten RN, Feenstra MG,
nucleus and behavioral depression in the obese leptin-deficient et al. (2008): Reward-predictive cues enhance excitatory synaptic strength
ob/ob mouse. Mol Brain Res 81:51–61. onto midbrain dopamine neurons. Science 321:1690–1692.
55. Li XL, Aou S, Oomura Y, Hori N, Fukunaga K, Hori T (2002): Impairment 68. Weigle DS, Duell PB, Connor WE, Steiner RA, Soules MR, Kuijper JL
of long-term potentiation and spatial memory in leptin receptor- (1997): Effect of fasting, refeeding, and dietary fat restriction on
deficient rodents. Neuroscience 113:607–615. plasma leptin levels. J Clin Endocrinol Metab 82:561–565.

www.sobp.org/journal

S-ar putea să vă placă și