All experiments involving animals were performed in accordance with protocols approved by the Institutional Animal Care and Use Committee of the Academy of Military Medical Sciences.
CUMS model
We used a CUMS regimen that followed the procedure originally described by Willner et al. [30] and that was subsequently adapted to mice [29]. This stress model involves the repeated application of mild physical and psychological stressors. The CUMS paradigm provides a model of a chronic-depressive-like state that develops gradually over time in response to stress. Hence, CUMS is considered more naturalistic in its induction [31]. Here, we subjected the mice to different types of stressors several times a day for 4 weeks in a chronic, inevitable, and unpredictable way. For ethical reasons, the stress procedure did not involve food and water deprivation or immobilization. The stressors used here were as follows: damp sawdust; changing the sawdust; placement in an empty cage or an empty cage with water covering the floor; placement in a soiled cage with an aversive odor; cage tilting (45° for 12 h); noise stress (15 min); inversion of the 12:12 h light:dark cycle; lights on or off for a short time during the dark phase or light phase, respectively; foot shock (150 mA, 30 min); and ice-water swimming (15°C, 5 min). We administered these stressors in a pseudorandom manner so that they could occur at any time of the day or night, and we changed the stressor sequence weekly to ensure that the stress procedure was unpredictable. During the behavioral tests, the stress procedure was slightly modified: we reduced the number of stressors applied during the light period to avoid interference with the tests. In addition, we did not subject test mice to any stressors for 12 h before the behavioral tests. Nonstressed mice were left undisturbed in their home cages. In all experiments, the first 2 weeks of CUMS were DBS-free, whereas the second 2 weeks of CUMS also included the administration of DBS. To determine the behavioral effects of the CUMS regimen and DBS treatment and to confirm the induction of depression-like behaviors, we examined sucrose consumption, tail suspension, and forced swimming in mice. We also measured locomotor activity using an open field test. We isolated nonstressed animals for 1 day before the open field test to match the conditions used for the CUMS mice.
Deep brain stimulation
Before surgery, 6-week-old C57BL/6J mice were handled for 7 days to habituate them to the experimenter and the stimulation procedure. Subsequently, we anesthetized the mice intraperitoneally (pentobarbital sodium, 0.7%, 10 μL/μg) and, using a stereotaxic frame, implanted electrodes (bipolar, two parallel tungsten wires, twisted, 0.22 mm in diameter) reaching the bilateral NAc (coordinates: 1.18 mm anterior, ±1.35 mm lateral, and −4.50 mm ventral to bregma). After 5–7 days of recovery and 2 weeks of a 4-week CUMS regime, we subjected the mice to high-frequency stimulations (100 Hz, 100 μA, 100 μs pulse width) daily for 1 h per day (30 min/unilateral) (i.e., NAc-DBS). Thus, stimulations took place from the second week of CUMS, and they continued for another 2 weeks (i.e., the end of CUMS). We controlled current intensity using a constant-current isolated stimulator, and we synchronized our stimulation protocols using Master-8 (A.M.P.I., Israel). We also placed control animals (CUMS-sham) into stimulation chambers daily for 1 h per day and connected them to the stimulator without applying any current.
Drug treatments
Bicuculline (Bic, 2 mg/kg/day; Apexbt, TX, USA) and Sulfobutylether (SBE)-β-Cyclodextrin (10% in normal saline; MCE, China) were administered via intraperitoneal (i.p.) injection for 2 weeks, which was accompanied by the onset of DBS treatment. Mice were kept on drug treatment until the completion of DBS. Control mice received SBE.
Behavioral studies
All behavioral tests were performed on mice 1 h after the completion of the NAc-DBS or CUMS-sham protocols. The sucrose preference test (SPT), forced swim test (FST), and tail suspension test (TST) were used to assess depression-like behavior [32, 33], whereas general locomotor activity was measured in the open field test (OFT). All behavioral tests were performed according to protocols established in our laboratory.
SPT. The SPT is widely accepted to measure the extent of anhedonic-like behavior [34, 35]. During the course of the CUMS protocol and DBS treatment, preference for sucrose-sweetened water was measured in mice once a week in their home cages. In mice deprived of food and water for 12 h, exposure to 2% sucrose is used to detect sucrose consumption over 2 h. Bottles were weighed before and after use by experimenters blinded to the mouse group allocation. The sucrose bottle was placed in the position of the original water bottle in the cage. During the ensuing 4 weeks, the intake of 2% sucrose by mice was measured once a week on the day without exposure to stress. The consumption of 2% sucrose solution per unit weight of mice can be calculated according to the following equation: sucrose consumption (g)/body weight (g). Sucrose solution bottles were prepared 24 h before their use. A random sampling of bottles placed in empty cages during SP testing demonstrated that leakage was negligible.
FST. The FST was performed in a glass cylinder (2-L beaker) filled with fresh tap water that was maintained at 15°C with an illumination of 100 Lux. Animals were placed individually in the cylinder for 5 min, and their behavior was recorded using a video camera positioned above the basin. The total amount of immobility was scored manually by a trained observer blinded to the treatment groups. An animal was considered immobile when it was floating passively in water, performing only movements that were necessary to keep the nose above the water surface [36].
TST. A metal hook was securely fixed with medical adhesive tape to the tip of the tails of the mice, and the animals were suspended using the hook. During the 6-min testing period, the animals were videotaped and total immobility (reflecting behavioral despair; in which the mice hung without any limb movement) for 5 min (the first and last 30 s were not considered) was scored automatically using analytical software (BIOSEB, In Vivo Research Instruments, France).
OFT. The OFT was used to assess anxiety-like behavior [37, 38]. Mice were individually placed into the periphery of the open field (blue plastic box, 50 × 50 × 50 cm; center defined as the inner 25 × 25 cm area; floor illumination, 150 Lux). The overall distance traveled by each mouse was measured using an automated activity monitoring system (DigBehv Video Analysis System, Shanghai Jiliang Software Technology Co., Ltd.) for 5 min.
Brain microdialysis
The C57BL/6J mice used in this experiment were aged 6–8 weeks. Concentric dialysis probes with a 1.0 mm membrane length were used (CMA 7, Sweden) to detect the concentration of GABA in the VTA. Mice were anesthetized intraperitoneally with pentobarbital sodium (pentobarbital sodium, 0.7%, 10 μL/μg) and placed in a stereotaxic apparatus. The skull was exposed, and a small hole was drilled on one side to expose the dura. The microdialysis catheter was aimed at the unilateral VTA (coordinates: −3.2 mm anterior, 0.4 mm lateral, and 3.0–3.5 mm ventral to bregma). After 5–7 days of recovery, experiments were performed on mice under anesthesia after NAc-DBS. Sterile and oxygenated (95% O2, 5 min) artificial cerebrospinal fluid (NaCl, 145 mM; KCl, 3.8 mM; MgCl2, 1.2 mM; CaCl2, 1.2 mM; pH 7.4) was pumped through the dialysis probe at a constant rate of 1 μL/min (BASi, West Lafayette, USA). A 2-h stabilization period was allowed before the collection of dialysates for analysis, and samples were taken every 30 min from the VTA. Dialysates were automatically collected with a refrigerated autosampler. The three fractions were collected after stimulation. Dialysate samples (15 μL) were injected without purification into the high-performance liquid chromatography-electrochemical detector (HPLC-ECD) apparatus equipped with a chromatographic column (Quattro 3 C18, 3 μm particle size, 150 × 2.1 mm; Chrom4, USA) using a refrigerated autoinjector to quantitate GABA (0.85 V, 35°C). The mobile phase was composed of methanol and buffer salt solution (0.1 M NaH2PO4; 1:9 (v/v)). This method can only be used after removing bacteria using a 0.22 μm filter and removing bubbles by ultrasonic exposure. Samples or standards were derivatized with phthaldialdehyde (OPA)-Na2SO3 (37 mM OPA, 50 mM Na2SO3, 90 mM boric acid, 5% methanol, pH 10.4; 10:1 (v/v)). The mobile phase was pumped at a flow rate of 0.3 mL/min. The retention time for GABA was 30 min. GABA was identified through its migration time and spike profiles. Peak values were normalized to an internal standard curve for a current signal (y, nA × min) and quantified by comparison with an external standard curve (Antec-ALEXYS On-Line Analysis System, Antec®, Netherlands). The values were corrected for in vitro probe recovery, which was approximately 10%.
Recombinant lentivirus-mediated RNA interference
The lentivirus-mediated interfering RNA (RNAi) was constructed and synthesized by Shanghai GeneChem Co., Ltd. (Shanghai, China). The target sequence used against the mouse GABA A receptor subunit alpha 1 (GABAA-α1) gene (Gene ID: 14394) was as follows: 5′-TGC CTA ATA AGC TCC TGC GTA-3′. The recombinant lentivirus LV-Gabra1-RNAi_87-GFP (i_87, virus titer: 3 × 109 infectious units mL−1) was produced by cotransfecting 293T cells with the lentivirus expression plasmid and packaging plasmids using Lipofectamine 2000. C57BL/6J mice (6–8 weeks of age) were used for stereotactic viral injections. Moreover, lentivirus-mediated RNAi (i_87) knockdown of the GABAA-α1 gene was performed via injection into the bilateral VTA (coordinates: −3.2 mm anterior, ±0.4 mm lateral, and 4.1–4.6 mm ventral to bregma). During pentobarbital sodium anesthesia (as described above), the skull was exposed via a small incision, and a small hole was drilled for injection. A modified microliter syringe (Hamilton) with a 22-gage needle was used: the tip of the needle was placed at the target region, and injection was performed at a speed of 50 nL/min using a micromanipulator. The needle was left in place for 10 min after the injection.
Neuron-specific adeno-associated viruses stereotaxic injections and optic fiber implantation
The C57BL/6J mice used in this experiment were aged 6–8 weeks. The animals were anesthetized with 0.7% pentobarbital sodium and placed in a stereotaxic frame (RWD, Shenzhen, China). To reveal the type of medium spiny neurons (MSN) projecting to the cell-specific neurons in the VTA, the anterograde monosynaptic transneuronal rAAV-hSyn-Cre-WRPE-PA plasmid (1.0 × 1013 infectious units mL−1) was injected into the NAc, and a Cre-inducible virus (a double inverted open reading frame (Dio) adeno-associated virus (AAV)) containing the enhanced yellow fluorescent protein (EYFP) (rAAV-Ef1α-Dio-EYFP-WPRE-PA; 1.82 × 1013 infectious units mL−1) in which the expression of EYFP is driven by the Cre–Dio reaction was injected into the VTA. For calcium imaging in the VTA, a neuron-specific AAV (rAAV2/9-mTH-Gcamp6m-WPRE-pA (5.81 × 1012 infectious units mL−1; BrainVTA, Wuhan, China)) and rAAV2/9-mDlx-GCaMP6m-WPRE-pA (1.91 × 1013 infectious units mL−1; BrainVTA, Wuhan, China) were injected into the VTA at two sites (0.4 μL per hemisphere; 0.2 μL at stereotaxic coordinates from bregma: anterior/posterior, −3.2 mm; medial/lateral, ±0.4 mm; dorsal/ventral, −4.1 mm; and 0.2 μL at dorsal/ventral, −4.6 mm, respectively), and an optic fiber was implanted unilaterally at 0.5 mm above the VTA (coordinates: −3.2 mm anterior, ±0.4 mm lateral, and −4.1 mm ventral to bregma). For optogenetic manipulations of the NAc input, we used a combination of a retrograding AAV serotype 2/R carrying Cre-inducible transgenes fused with enhanced green fluorescent protein (eGFP) under the control of the mDLx promoter in inhibitory neurons (rAAV2/R-mDLx-Cre-EGFP-WPRE-pA, 4.72 × 1012 infectious units mL−1) (BrainVTA, Wuhan, China), which was bilaterally injected at two sites of the VTA, to retrogradely trace input neurons to the NAc (0.3 μL per hemisphere; 0.15 μL at stereotaxic coordinates from bregma: anterior/posterior, −3.3 mm; medial/lateral, ±0.5 mm; dorsal/ventral, −4.1 mm; and 0.15 μL at dorsal/ventral, −4.6 mm). Furthermore, a Cre-inducible virus (neuron-specific AAV serotype 2/9 carrying the excitatory optogenetic protein oChIEF and a FLEx element fused to tdTomato under the control of the CAG promoter (rAAV2/9-CAG-FLEx-oChIEF-tdTomato-WPRE-pA, 1.54 × 1013 infectious units mL−1, Taitool Bioscience, Shanghai, China) were bilaterally injected into the NAc (0.2 μL per hemisphere; stereotaxic coordinates from bregma: anterior/posterior, 1.18 mm; medial/lateral, ±1.35 mm; dorsal/ventral, −4.5 mm). For optogenetic manipulations of the VTA input, a neuron-specific AAV serotype 2/9 carrying Cre fused with eGFP under the control of the NLS promoter in tyrosine hydroxylase (TH)-positive neurons (rAAV2/9-mTH-NLS-Cre-EGFP-WPRE, 2.34 × 1012 infectious units mL−1, Taitool Bioscience, Shanghai, China) and a Cre-inducible virus (AAV serotype 2/9 carrying oChIEF and a FLEx element fused to tdTomato under the control of the CAG promoter (rAAV2/9-CAG-FLEx-oChIEF-tdTomato-WPRE-pA, 1.54 × 1013 infectious units mL−1; Taitool Bioscience, Shanghai, China)) were mixed and bilaterally injected at two sites of the VTA (0.3 μL per hemisphere; 0.15 μL at stereotaxic coordinates from bregma: anterior/posterior, −3.3 mm; medial/lateral, ±0.5 mm; dorsal/ventral, −4.1 mm; and 0.15 μL at dorsal/ventral, −4.6 mm). For optogenetic manipulation experiments, mice were also implanted with a bilateral fiber-optic cannula that was secured to the skull using dental cement. Fiber-optic cannulas were 200 μm (fiber core diameter) for optogenetic and fiber photometry experiments. We used the following stereotactic coordinates in the VTA: anterior/posterior, −3.3 mm; medial/lateral, ±1.8 mm; dorsal/ventral, −4.6 mm; 20° angle.
Optogenetic manipulation
To record calcium imaging in cell-type-specific neurons in the VTA, rAAV2/9-mTH-GCaMP6m and rAAV2/9-mDlx-GCaMP6s were used for injection into the VTA. Fiber photometry allows the real-time excitation and recording of fluorescence from genetic-encoded calcium indicators in freely moving mice. Mice were habituated to the fiber patch cord for at least 15 min per day for 3 days before tests were conducted inside home cages. The fiber photometry system consisted of an excitation light-emitting diode (LED) light (470 nm, Inper Technology Co., Ltd., Hangzhou, China), which was reflected off a dichroic mirror with a 435–488 nm reflection band and a 503–538 nm transmission band (Inper Technology Co., Ltd., Hangzhou, China), and coupled to a 200-μm 0.37 N.A. optical fiber (Inper Technology Co., Ltd., Hangzhou, China) by an objective lens. Laser power was adjusted at the tip of the optical fiber to a low level of 20–30 µW to minimize bleaching. The GCaMP6 fluorescence was bandpass-filtered (MF525-39, Thorlabs) and focused by a 20× objective lens. The fluorescence signal was collected using a CMOS camera. The end of the fiber was imaged at a frame rate of 1–320 Hz. The ROI area size and mean value were set through Inper Studio. Moreover, 470 and 410 nm light sources were used alternately, with 410 nm being used as the control. Analysis of the resulting signal was performed using custom-written MATLAB software. To selectively activate the VTA-GABA neurons projecting MSNs in the NAc, projection-specific expression of oChIEF in the NAc was realized via the combined use of retrograding rAAV2/R-mDLx-Cre-EGFP expression in VTA GABAergic neurons, which was injected into the VTA, and Cre-inducible rAAV-FLEx-oChIEF-tdTomato, which was injected into the NAc. For in vivo experiments, the mice were moved to their home cage and allowed a 30 min acclimatization period, the optical cannula was then connected to the optical patch cable, and the mice were administered high-frequency, phasic (frequency: 30 Hz; pulse width: 5 ms; burst duration: 0.267 s; burst interval: 5 s; eight pulses/5 s) blue-light illumination from a blue laser. To selectively activate the DA neurons in the VTA, cell-type-specific expression of oChIEF in dopaminergic neurons was realized via the combined use of rAAV-mTh-Cre-EGFP and Cre-inducible rAAV-FLEx-oChIEF-tdTomato. For in vivo experiments, the mice were moved to their home cage and allowed a 30 min acclimatization period, the optical cannula was then connected to the optical patch cable, and the mice were administered high-frequency, phasic (frequency: 20 Hz; pulse width: 40 ms; burst duration: 0.25 s; burst interval: 10 s; five pulses/10 s) blue-light stimulations from a blue laser. In mice expressing oChIEF, a 473 nm blue laser diode and a stimulator were used to generate blue-light pulses. Fibers measuring at least 10 mW were utilized. Phasic light pulses were delivered for 30 min per day for 14 consecutive days. Behavioral testing was resumed at 4 or 24 h poststimulation.
Histological examination and immunohistochemistry
After completion of all experiments, the mice were anesthetized with pentobarbital sodium intraperitoneally (0.7%, 10 μL/μg) and perfused transcardially with normal saline, followed by 4% paraformaldehyde (PFA). Brains were removed, and coronal sections (40 mm) through the target area were prepared. We confirmed the location of the stimulating electrodes, probes, and optic fibers on bright-field photomicrographs. Only mice with electrodes, probes, and optic fiber placed in the areas of interest were included in subsequent data analyses. Virus-injected brains were fixed overnight in a 4% PFA solution and then equilibrated in 30% sucrose in PBS. Next, 40-μm coronal slices were cut using a freezing microtome. Slices were stored in a cryoprotection solution at 4°C until further processed. The sections were incubated with primary antibodies (mouse anti-GABAA receptor alpha 1 antibody #ab3299, Abcam, 1:1000; rabbit anti-dopamine receptor D1 antibody #ab40653, Abcam, 1:1000; rabbit anti-GABA antibody #ab62669, Abcam, 1:1000; rabbit anti-TH antibody #AB152, Millipore, 1:2000; or mouse anti-Cre recombinase antibody #MAB3120, MERCK, 1:1000) overnight at 4°C. Alexa Fluor®488-, 594-, or 647-conjugated goat antirabbit or antimouse IgG antibodies, or antirabbit IgG antibodies (1:500; Invitrogen, CA, USA) were used as secondary antibodies. Nuclei were counterstained using DAPI. The fluorescent signals were imaged with A1R laser-scanning confocal microscopy using a 10× or 20× objective (Nikon, Japan).
Western blotting
Brains were immediately flash-frozen after dissection, and defined tissue punches of VTA were then collected using a 1-mm round tissue punch while sectioning brains on a cryostat. Total proteins extracted from the VTA using sodium dodecyl sulfate (SDS) lysis buffer (2% SDS, 10% glycerol, 0.1 mM dithiothreitol, and 0.2 M Tris-HCl, pH 6.8) were subjected to western blotting using the indicated primary (mouse anti-GABAA receptor alpha 1 #ab3299, Abcam, 1:200; rabbit antibeta actin #ab8227, Abcam, 1:1000) and secondary antibodies. Bound antibodies were visualized using enhanced chemiluminescence detection. The band optical densities were quantified by the Image Quant software.
Data analysis
Data are presented as the mean ± standard error of the mean (SEM). Exact n numbers are given in the table and figure legends. Statistical analysis was performed using GraphPad Prism 7.0 (GraphPad Software, San Diego, CA, USA). Data were statistically analyzed using one-way ANOVA (with post hoc Bonferroni test) or unpaired/paired Student’s t-tests. Statistical significance was set at P < 0.05. In vivo calcium signal analysis/photometry data were exported as MATLAB mat files for further analysis after recording. To visualize the recording traces of the activity of different VTA neuronal populations during DBS_ON, the first 60 s was chosen as the baseline, followed by the calculation of the fluorescence change (ΔF/F) normalization by the ΔF/F = (F − F0)/F0 method, where F is the normalized fiber photometry signal data and F0 is the average of the fluorescence values during the baseline period (from 60 s preceding the time of DBS_ON). The ΔF/F values are presented with heatmaps or per-event plots, with shaded areas indicating the SEM. All calculations were performed using MATLAB 2014a.