ABSTRACT
Chronic stress affects brain functions, leading to the development of mental disorders like anxiety and depression, as well as cognitive decline and social dysfunction. Among many biological changes in chronically stressed brains, disruptions in AMPA Receptor (AMPAR)‐mediated synaptic transmission in the hippocampus are associated with stress responses. We have revealed that low‐dose ketamine rapidly induces the expression of GluA1‐containing, GluA2‐lacking Ca2+‐Permeable AMPARs (CP‐AMPARs), which enhances glutamatergic synaptic strength in hippocampal neurons. Additionally, subanesthetic low‐dose ketamine decreases anxiety‐ and depression‐like behaviors in naïve animals. In addition to reducing depression, some research indicates that ketamine may have protective effects against chronic stress in both humans and animals. However, the role of CP‐AMPARs in the actions of ketamine's antistress effects is largely unknown. We use whole‐cell patch‐clamp recordings from CA1 pyramidal neurons in female and male hippocampal slices and multiple behavioral assays, including reciprocal social interaction, contextual fear conditioning, and tail suspension test. We demonstrate that low‐dose ketamine treatment reverses chronic restraint stress (CRS)‐induced social dysfunction, a loss of hippocampus‐dependent fear learning and memory, and depression‐like behavior in both female and male mice. Furthermore, we show that the ketamine‐induced antistress effects on these behaviors are dependent on CP‐AMPAR expression. Our findings suggest that subanesthetic low‐dose ketamine rapidly triggers the expression of CP‐AMPARs in the hippocampus, which induces antidepressant and antistress effects.
Keywords: behaviors, Ca2+‐permeable AMPA receptors, chronic stress, ketamine
Low‐dose ketamine rapidly induces the expression of GluA1‐containing, GluA2‐lacking Ca2+‐Permeable AMPARs (CP‐AMPARs) in the hippocampus via NMDAR antagonism. When ketamine is administered, CP‐AMPAR‐mediated Ca2+ influx may replace NMDAR‐dependent Ca2+ signaling. This increases neural plasticity, which leads to antistress effects in animals. Therefore, inhibition of CP‐AMPARs using antagonists prevents ketamine's effects. Our findings suggest that subanesthetic low‐dose ketamine rapidly triggers the expression of CP‐AMPARs in the hippocampus, which induces antistress effects.

Abbreviations
- BBB
blood–brain barrier
- CFC
contextual fear conditioning
- CP‐AMPARs
Ca2+‐Permeable AMPA receptors
- CRS
chronic restraint stress
- FST
forced swim test
- OFT
open filed test
- TST
tail suspension test
- uEPSCs
uncaging‐induced excitatory postsynaptic currents
1. Introduction
The World Health Organization (WHO) estimates that over 300 million people worldwide have depression (4.4% of the global population), which makes depression the leading cause of disability worldwide. The US Food and Drug Administration (FDA) approved esketamine (the S enantiomer form of ketamine) for the treatment of depression in 2019, sparking a surge in clinical and public interest around the world [1]. Although the antidepressant effects of ketamine become evident within a few hours or 1 day of a single infusion, the benefits generally disappear within 1 week [2]. More recent studies have shown that repeated ketamine infusions somewhat sustain the effects [3, 4, 5]. However, relapse of depression still occurs after cessation of treatment (on average, 18–19 days post‐treatment) [3, 4]. Moreover, because ketamine is associated with psychomimetic side effects and risk of addiction, it is typically given in just a single dose or short series of doses rather than repeated treatments [6, 7, 8]. Some patients receiving ketamine therapy are also advised to stay on other traditional antidepressants while receiving ketamine [9]. However, reports have shown that some patients have been inflicted with adverse effects mimicking psychosis following ketamine administration on top of their regular polydrug regimen [10, 11, 12]. It is unclear which medications (or set of medications) negatively interact with ketamine [9]. Additionally, ketamine is found to be effective in reducing suicidality in treatment‐resistant depression patients (10%–20% of the depression patients) [13, 14]. For these patients, ketamine + other traditional antidepressants is likely ineffective. Given all these problems, the field requires the development of new strategies that will permit a reduction in the frequency of ketamine administration after response to acute treatment, with maintenance of its beneficial effects for its broader application [5]. Unfortunately, achieving these goals is considerably more challenging because the exact mechanisms behind ketamine's antidepressant effects are still unclear. Importantly, ketamine‐induced neuroplasticity has been proposed as a mechanism underlying its antidepressant effects [15]. However, still unclear, is when and how neuroplasticity is induced. Specifically, molecular and cellular processes of how ketamine induces neuroplasticity remain to be elucidated.
One mechanism postulated for ketamine's rapid antidepressant actions involves the enhancement of excitatory synaptic drive in the hippocampus [16, 17, 18]. However, it has been demonstrated that ketamine is a noncompetitive NMDA receptor (NMDAR) antagonist that inhibits excitatory synaptic transmission [19]. This poses a contradictory question of how ketamine enhances excitatory activity and synaptic Ca2+ signaling while blocking NMDARs. This can be explained by an increase in AMPA receptor (AMPAR)‐mediated synaptic activity, but the mechanisms are unknown [20]. In addition to reducing depression, some research indicates that ketamine may have protective effects against chronic stress in both humans and animals [21, 22, 23, 24, 25, 26, 27, 28, 29]. Chronic stress affects brain functions, resulting in the development of mental disorders like anxiety and depression, as well as cognitive decline and social dysfunction [30, 31, 32]. Among many biological changes in chronically stressed brains, disruptions in AMPAR‐mediated synaptic transmission in the hippocampus are associated with stress responses in animal models and individuals with depression [33]. Thus, recovery from AMPAR‐mediated synaptic dysfunction may be the key target of ketamine's antistress and antidepressant effects.
There are two general types of AMPARs formed through combination of their subunits: GluA2‐containing Ca2+‐impermeable AMPARs and GluA2‐lacking, GluA1‐containing Ca2+‐Permeable AMPARs (CP‐AMPARs) [34]. Compared to Ca2+‐impermeable AMPARs, CP‐AMPARs have higher single channel conductance, which contributes to synaptic plasticity [35, 36, 37, 38, 39, 40, 41, 42, 43, 44]. Both preclinical and clinical studies show that chronic stress significantly reduces GluA1 levels in hippocampal synapses, while there are conflicting results describing alterations in hippocampal GluA2 under chronic stress [33]. These results suggest that the stress‐induced decrease in synaptic GluA1 levels diminishes excitatory synaptic transmission in the hippocampus and exacerbates negative behaviors in chronic stress. Therefore, recovery from GluA1‐mediated synaptic dysfunction may be the key target of ketamine's antidepressant and antistress effects. We have revealed that low‐dose ketamine (1 μM, a concentration that blocks approximately 50% of NMDAR‐induced currents) [45] rapidly induces the expression of CP‐AMPARs in cultured hippocampal neurons, which is mediated by increasing GluA1 phosphorylation and its surface expression [46]. Moreover, we have shown that this ketamine‐induced CP‐AMPAR expression enhances glutamatergic synaptic strength in cultured neurons [46]. Additionally, subanesthetic low‐dose ketamine (5–10 mg/kg) decreases anxiety‐ and depression‐like behaviors in naïve animals [46]. In fact, an increase in hippocampal activity is known to improve anxiety‐ and depression‐like behaviors, fear learning and memory, and social behavior in mice [16, 17, 18, 47, 48, 49, 50, 51]. This suggests that ketamine‐induced recovery from disruptions in AMPAR‐mediated synaptic transmission and behaviors in stressed animals is likely associated with an increase in synaptic GluA1 expression in the hippocampus [52, 53, 54].
Although we have shown that subanesthetic low‐dose ketamine selectively increases GluA1 expression in the hippocampus and induces antidepressant effects via CP‐AMPAR expression in naïve mice [46], much research with hippocampal neural circuits is required to fully comprehend ketamine's actions on chronic stress‐induced behavioral alterations. Here, we use an electrophysiological approach in hippocampal slices to confirm that low‐dose ketamine treatment indeed induces CP‐AMPAR expression in hippocampal pyramidal neurons. We further show that ketamine at the low dosages reverses chronic restraint stress (CRS)‐induced behavioral changes in mice, which is dependent on the expression of CP‐AMPARs. Our findings thus suggest that subanesthetic low‐dose ketamine rapidly triggers the expression of CP‐AMPARs in the hippocampus, which in turn enhances synaptic strength and compensates for reduced NMDAR antagonism‐mediated synaptic Ca2+ signaling to induce antidepressant and antistress effects.
2. Methods
2.1. Animals
C57Bl6J (Jax 000664) mice were obtained from Jackson Laboratory and bred in the animal facility at Colorado State University (CSU). Animals were housed under a 12:12 h light/dark cycle. To collect brain tissues from animals, mice were deeply anesthetized and euthanized by CO2 asphyxiation. All efforts were made to minimize animal suffering. CSU's Institutional Animal Care and Use Committee (IACUC) reviewed and approved the animal care and protocol (3408). Results were reported following the ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines [55].
2.2. Chronic Restraint Stress (CRS)
3‐month‐old C57Bl6 female and male mice were individually placed into 50 mL polypropylene conical tubes with multiple holes for ventilation, and they were exposed to restraint stress (2 h/day) for 14 consecutive days. After restraint stress, the mice were returned to the home cage. Following 2‐week CRS, drug injections, blood collection, and multiple behavior assays were carried out as shown in the experimental timeline in Figure 1.
FIGURE 1.

Experimental timeline of chronic restraint stress (CRS) that is applied to mice for 2 weeks, drug treatment, blood collection, and behavioral tests—reciprocal social interaction test, open‐field test (OFT), contextual fear conditioning (CFC) day 1 and day 2, and tail suspension test (TST). Single intraperitoneal injection of ketamine (5 mg/kg for females and 10 mg/kg for males) or saline in CRS mice in the presence or absence of IEM‐1460 (10 mg/kg for both females and males). We optimized the test order from least to most stressful in order to prevent the possibility that multiple behavioral tests with the same mouse can induce stress, especially if the tests are performed too closely together.
2.3. Drug Treatment
Ketamine hydrochloride (VetOne, 510 189) was used in both in vitro and in vivo experiments. 5 mg/kg and 10 mg/kg ketamine were intraperitoneally injected into 3‐month‐old female and male C57Bl6 mice, respectively (the conditions have been shown to exclusively increase hippocampal GluA1 and induce antidepressant effects on naïve animals' behaviors) [46], and saline was administered to controls. We have approval from IACUC to use ketamine and have the United States Drug Enforcement Administration license to use ketamine for research purposes (DEA# RK0573863). 100 μM 1‐Naphthyl acetyl spermine trihydrochloride (NASPM, Tocris Bioscience) was used to block CP‐AMPARs in slice electrophysiology. 10 mg/kg IEM‐1460 (Tocris Bioscience, 1636) was intraperitoneally injected into 3‐month‐old male and female C57Bl6J mice to inhibit in vivo CP‐AMPAR activity because it is blood–brain barrier (BBB)‐permeable [56, 57, 58]. Following CRS, we performed the experiments in the presence or absence of IEM‐1460 and compared outcomes to examine whether ketamine treatment reversed the CRS‐induced effects via the expression of CP‐AMPARs.
2.4. Brain Slice Preparation and Electrophysiology
The mice were deeply anesthetized with isoflurane, and their brains were removed. The brains were submerged in cold, oxygenated (95% O2 and 5% CO2) slicing medium containing (in mM): 85 NaCl, 1.25 NaH2PO4, 4 MgCl2, 0.5 CaCl2, 24 NaHCO3, 2.5 KCl, 75 sucrose, and 25 glucose. Horizontal hippocampal slices (350 μm thick) were cut using a vibratome (Leica VT 1000S; Leica Microsystems). The solution was continuously oxygenated with 95% O2 and 5% CO2. Slices were initially maintained at 33°C for 30 min, then at room temperature (22°C± 1°C) until they were used for electrophysiological recordings. Whole‐cell patch‐clamp recording procedures used in the present study were like those described previously [59, 60, 61]. Electrophysiological recordings were performed on CA1 pyramidal cells at 33°C in oxygenated (95% O2 and 5% CO2) artificial cerebrospinal fluid (ACSF) containing (in mM): 126 NaCl, 2.5 KCl, 26 NaHCO3, 2 CaCl2, 2 MgCl2, 1.25 NaH2PO4, and 10 glucose. Slices were superfused with ACSF at a rate of 2.0 mL/min. Individual neurons were visualized using an upright microscope (Olympus BX51WI) with infrared differential interference contrast (DIC) optics. CA1 pyramidal cells in the stratum pyramidale (the pyramidal cell layer), visualized using differential interference contrast microscopy, will be selected based on their morphology as described previously [61]. Whole‐cell patch‐clamp recordings were obtained from CA1 pyramidal cells with borosilicate patch pipettes (2–4 MΩ) filled with an internal solution containing (in mM): 126 K‐gluconate, 4 KCl, 10 HEPES, 4 ATP‐Mg, 0.3 GTP‐Na, and 10 phosphocreatine. The pH and osmolarity of the internal solution were adjusted to 7.2 and 290 mOsm, respectively. Recordings were obtained using a MultiClamp 700B amplifier (Molecular Devices). After forming the whole‐cell configuration, the recording was allowed to rest for at least 5 min prior to data acquisition. Signals were sampled at 10 kHz, low‐pass filtered at 3 kHz using a Digidata 1440A analog‐to‐digital digitizer (Molecular Devices) and stored on a computer for subsequent analysis using pClamp 10 software (Molecular Devices). Series resistances were continuously monitored with recordings being discarded if the series resistance increased > 20%. We then measured glutamate uncaging‐induced excitatory postsynaptic currents (uEPSCs) in hippocampal CA1 neurons 1 h following a single dose of ketamine (5 mg/kg for females and 10 mg/kg for males). For glutamate uncaging, 100 μM MNI‐caged L‐glutamate was added to ACSF, and epi‐illumination photolysis (390 nm, 0.12 mW/mm2, 50 ms) was applied near the soma of a neuron. 1 μM TTX was added to ACSF for preventing action potential‐dependent network activity. The average of uEPSC amplitudes (Vhold = −70 and + 60 mV) in CA1 pyramidal cells was compared between saline and ketamine treatment. To examine whether CP‐AMPARs played a role in uEPSCs, we recorded uEPSCs with a bath application of 100 μM NASPM, a CP‐AMPAR blocker. Additionally, we added saline to ACSF as a control for NASPM treatment. Each recording was performed in different slices. We then analyzed the amplitudes of uEPSCs in each condition using Clampfit 10.7 (Molecular Devices).
2.5. Reciprocal Social Interaction
We performed the reciprocal social interaction test to examine social interaction between two freely moving mice [62, 63]. A test mouse (3‐month‐old) and a stranger mouse of identical genotypes that were previously housed in different cages were placed into the chamber (40 W × 40 L × 40 H cm) and allowed to explore freely for 20 min. Social behavior was recorded using a camera mounted overhead. Social contacts were determined by the sniffing time that was defined as each instance in which a test mouse's nose came within 2 cm of a stranger mouse. The total number of reciprocal interactions and total duration of reciprocal interactions were measured manually. Animals that escaped the chamber or showed aggressive behaviors (e.g., attacking a stranger mouse) were excluded from the analysis.
2.6. Open Field Test (OFT)
We measured locomotor activity and anxiety‐like behavior using OFT as carried out previously [46, 64, 65]. A test mouse was first placed in the center of the open field chamber (40 W × 40 L × 40 H cm) for 5 min. Animals were then allowed to explore the chamber for 20 min. The behavior was recorded by a video camera. A 20 × 20 cm center square was defined as the inside zone. Data were analyzed using the ANY‐maze tracking program (Stoelting Co.) to acquire total traveled distance (locomotor activity) and time spent outside and inside (anxiety‐like behavior). Animals that escaped the chamber or showed significantly decreased locomotor activity were excluded from the analysis.
2.7. Contextual Fear Conditioning (CFC)
CFC (Habitest Modular System, Coulbourn Instrument) was carried out as described previously [64, 66, 67]. On Day 1, a test mouse was placed in a novel rectangular chamber with a grid floor. After a 3 min baseline period, the test animal was given one shock (a 2 s, 0.5 mA shock) and stayed in the chamber for an additional 1 min after the shock before being returned to the home cage overnight. A contextual memory test was conducted the next day (Day 2) in the same conditioning chamber for 3 min. Fear memory was determined by measuring the percentage of the freezing response (immobility excluding respiration and heartbeat) using an automated tracking program (FreezeFrame). Mice that did not freeze were excluded from the analysis.
2.8. Tail Suspension Test (TST)
It has been suggested that the forced swim test (FST) and TST are based on the same principle: measurement of the duration of immobility when rodents are exposed to an inescapable situation [68]. Importantly, TST is known to be more sensitive to antidepressant agents than FST because the animals remain immobile longer in TST than FST [69]. We thus used TST to examine depression‐like behavior as described previously [46, 66, 70]. A test mouse was suspended by its tail from a rod suspended 20 cm above the tabletop surface with adhesive tape placed 1 cm from the tip of the tail. Animals were immobile when they exhibited no body movement and hung passively for > 3 s. The time during which mice remained immobile was quantified over a period of 6 min. The behavior was recorded by a video camera, and data were analyzed using the ANY‐maze tracking program to acquire immobility (depression‐like behavior). Mice that successfully climbed their tails to escape were excluded from the analysis.
2.9. Corticosterone
Corticosterone levels in mice were measured as described previously [71, 72]. 50 μL tail‐tip whole blood samples were collected and centrifuged at 5000 rpm for 10 min. 10 μL of serum was harvested and stored at −20°C until assay. 5 μL of serum samples in each condition were assayed using the DetectX Corticosterone Multi‐Format Enzyme‐linked immunosorbent assay (ELISA) kit (Arbor Assays Inc. K014‐H). To prevent possible artifacts during the ELISA experiment, we measured the corticosterone levels twice using the same sample.
2.10. Statistical Analysis
All behavior tests were blindly scored by more than two investigators. The Franklin A. Graybill Statistical Laboratory at Colorado State University was consulted for statistical analysis in the current study, including sample size determination, randomization, experiment conception and design, data analysis, and interpretation. We used the GraphPad Prism 10 software to determine statistical significance (set at p < 0.05). Grouped results of single comparisons were tested for normality with the Shapiro–Wilk normality or Kolmogorov–Smirnov test and analyzed using an unpaired two‐tailed Student's t‐test when data are normally distributed. Differences between multiple groups were assessed by N‐way analysis of variance (ANOVA) with the Tukey test. The graphs were presented as mean ± Standard Deviation (SD). We discard data points that are located further than two SD above or below the average as outliers.
3. Results
3.1. Subanesthetic Low‐Dose Ketamine Induces the Expression of CP‐AMPARs in Hippocampal CA1 Pyramidal Neurons
The hippocampus is one of the key brain regions controlling cognition, motivation, and social behavior [47, 73, 74, 75, 76, 77, 78, 79, 80, 81]. An increase in hippocampal activity reverses stress‐induced memory impairment, social dysfunction, and mood disorder‐associated behaviors [16, 17, 18, 47, 48, 49, 50, 51, 82]. More importantly, the hippocampus is preferably targeted by ketamine at low doses [83]. This suggests that the hippocampus is the primary brain region responsible for ketamine's antistress effects. In fact, we have discovered that subanesthetic low‐dose ketamine significantly increases synaptic GluA1 but not GluA2, an indication of CP‐AMPAR expression, in the hippocampus of naïve animals [46]. We thus decided to examine whether low‐dose ketamine was able to induce CP‐AMPAR expression in hippocampal neurons using whole‐cell patch clamping with glutamate uncaging. We intraperitoneally injected 5 mg/kg and 10 mg/kg ketamine to female and male C57Bl6 mice, respectively, and saline was administered to controls. These ketamine concentrations were shown to exclusively increase hippocampal GluA1 and induce antidepressant effects on naïve animals' behaviors [46]. 1 h following injection, hippocampal slices were obtained, and we then measured the amplitudes of glutamate uncaging‐induced excitatory postsynaptic currents (uEPSCs) (Vhold = −70 and + 60 mV) in CA1 pyramidal cells. We found that ketamine treatment significantly elevated the amplitudes of uEPSCs (Vhold = −70 mV) in these neurons when compared to control cells (CTRL) (FEMALE: CTRL, 86.330 ± 23.107 pA and Ketamine, 113.208 ± 30.563 pA, p = 0.0367. MALE: CTRL, 83.915 ± 27.768 pA and Ketamine, 109.329 ± 28.293 pA, p = 0.0471) (Figure 2a and Table 1). However, when ketamine had no effect on uEPSCs (Vhold = +60 mV) (FEMALE: CTRL, 292.223 ± 97.252 pA and Ketamine, 360.238 ± 173.766 pA, p = 0.5017. MALE: CTRL, 286.115 ± 129.521 pA and Ketamine, 372.100 ± 126.540 pA, p = 0.2718) (Figure 2b and Table 2). To examine whether CP‐AMPARs were involved in ketamine's effects on uEPSCs, we added 100 μM NASPM, a CP‐AMPAR antagonist, to ACSF and measured the amplitudes of uEPSCs in CA1 pyramidal cells from saline‐ or ketamine‐injected animals. We found that NASPM treatment significantly reduced the amplitudes of uEPSCs (Vhold = −70 and + 60 mV) following ketamine treatment (Vhold = −70 mV FEMALE: Ketamine + NASPM, 58.671 ± 22.786 pA, p < 0.0001. MALE: Ketamine + NASPM, 55.159 ± 26.805 pA, p < 0.0001. Vhold = +60 mV; FEMALE: Ketamine + NASPM 213.793 ± 111.514 pA, p = 0.0170. MALE: Ketamine + NASPM, 233.635 ± 130.302 pA, p = 0.0133) (Figure 2a,b, Tables 1 and 2). However, NASPM had no effect on the amplitudes of uEPSCs (Vhold = −70 and + 60 mV) measured in slices from saline‐injected mice (Vhold = −70 mV; FEMALE: NASPM, 61.702 ± 21.324 pA, p = 0.0575. MALE: NASPM, 77.500 ± 16.628 pA, p = 0.9248. Vhold = +60 mV; FEMALE: NASPM, 276.986 ± 97.353 pA, p = 0.9885. MALE: NASPM, 284.958 ± 123.642 pA, p > 0.9999) (Figure 2a,b, Tables 1 and 2). These findings demonstrate that subanesthetic low‐dose ketamine induces the expression of CP‐AMPARs in hippocampal CA1 pyramidal neurons.
FIGURE 2.

Subanesthetic low‐dose ketamine induces the expression of CP‐AMPARs in hippocampal CA1 pyramidal neurons. Representative traces and summary of the amplitudes of uEPSCs (a) Vhold = −70 mV and (b) Vhold = +60 mV) in each condition (n = number of cells [n = number of animals]. FEMALE: CTRL = 13 [3], NASPM = 14 [3], Ketamine = 13 [3], and Ketamine + NASPM = 14 [3]. MALE: CTRL = 13 [4], NASPM = 14 [4], Ketamine = 13 [3], and Ketamine + NASPM = 14 [3]). *p < 0.05 and ****p < 0.0001, Two‐Way ANOVA, Tukey test. A bar indicates photostimulation. Ketamine: 5 mg/kg for females and 10 mg/kg for males. NASPM: 100 μM.
TABLE 1.
Statistical analysis of the amplitudes of uEPSCs (Vhold = −70 mV) in hippocampal CA1 pyramidal neurons.
| Female (−70 mV) | |||||
|---|---|---|---|---|---|
| ANOVA table | SS (Type III) | DF | MS | F (DFn, DFd) | p |
| Interaction | 3015 | 1 | 3015 | F (1, 50) = 4.979 | p = 0.0302 |
| Row factor (Saline vs. Ket) | 1917 | 1 | 1917 | F (1, 50) = 3.165 | p = 0.0813 |
| Column factor (Saline vs. NASPM) | 21 122 | 1 | 21 122 | F (1, 50) = 34.88 | p < 0.0001 |
| Residual | 30 277 | 50 | 605.5 | ||
| Tukey's multiple comparisons test | Predicted (LS) mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| CTRL vs. NASPM | 24.63 | −0.5609 to 49.82 | 0.0575 |
| CTRL vs. ketamine | −26.88 | −52.53 to −1.227 | 0.0367 |
| CTRL vs. ketamine + NASPM | 27.66 | 2.470 to 52.85 | 0.0262 |
| NASPM vs. ketamine | −51.51 | −76.69 to −26.32 | < 0.0001 |
| NASPM vs. ketamine + NASPM | 3.031 | −21.69 to 27.75 | 0.9879 |
| Ketamine vs. ketamine + NASPM | 54.54 | 29.35 to 79.72 | < 0.0001 |
| Male (−70 mV) | |||||
|---|---|---|---|---|---|
| ANOVA table | SS (Type III) | DF | MS | F (DFn, DFd) | p |
| Interaction | 7866 | 1 | 7866 | F (1, 55) = 11.82 | p = 0.0011 |
| Row factor (Saline vs. Ket) | 59.69 | 1 | 59.69 | F (1, 55) = 0.08970 | p = 0.7657 |
| Column factor (Saline vs. NASPM) | 12 776 | 1 | 12 776 | F (1, 55) = 19.20 | P < 0.0001 |
| Residual | 36 598 | 55 | 665.4 | ||
| Tukey's multiple comparisons test | Predicted (LS) mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| CTRL vs. NASPM | 6.415 | −20.94 to 33.77 | 0.9248 |
| CTRL vs. ketamine | −25.41 | −50.59 to −0.2344 | 0.0471 |
| CTRL vs. ketamine + NASPM | 27.76 | 2.577 to 52.94 | 0.0253 |
| NASPM vs. ketamine | −31.83 | −57.60 to −6.062 | 0.0097 |
| NASPM vs. ketamine + NASPM | 21.34 | −4.426 to 47.11 | 0.1376 |
| Ketamine vs. ketamine + NASPM | 53.17 | 29.73 to 76.61 | < 0.0001 |
TABLE 2.
Statistical analysis of the amplitudes of uEPSCs (Vhold = +60 mV) in hippocampal CA1 pyramidal neurons.
| Female (+60 mV) | |||||
|---|---|---|---|---|---|
| ANOVA table | SS (Type III) | DF | MS | F (DFn, DFd) | p |
| Interaction | 58 023 | 1 | 58 023 | F (1, 50) = 3.814 | p = 0.0564 |
| Row factor (Saline vs. Ket) | 78.38 | 1 | 78.38 | F (1, 50) = 0.005152 | p = 0.9431 |
| Column factor (Saline vs. NASPM) | 88 106 | 1 | 88 106 | F (1, 50) = 5.791 | p = 0.0198 |
| Residual | 760 699 | 50 | 15 214 | ||
| Tukey's multiple comparisons test | Predicted (LS) mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| CTRL vs. NASPM | 15.24 | −111.0 to 141.5 | 0.9885 |
| CTRL vs. ketamine | −68.02 | −196.6 to 60.56 | 0.5017 |
| CTRL vs. ketamine + NASPM | 78.43 | −47.83 to 204.7 | 0.3602 |
| NASPM vs. ketamine | −83.25 | −209.5 to 43.00 | 0.3082 |
| NASPM vs. ketamine + NASPM | 63.19 | −60.70 to 187.1 | 0.5327 |
| Ketamine vs. ketamine + NASPM | 146.4 | 20.19 to 272.7 | 0.0170 |
| Male (+60 mV) | |||||
|---|---|---|---|---|---|
| ANOVA table | SS (Type III) | DF | MS | F (DFn, DFd) | p |
| Interaction | 67 842 | 1 | 67 842 | F (1, 55) = 4.158 | p = 0.0462 |
| Row factor (Saline vs. Ket) | 4323 | 1 | 4323 | F (1, 55) = 0.2650 | p = 0.6088 |
| Column factor (Saline vs. NASPM) | 70 148 | 1 | 70 148 | F (1, 55) = 4.300 | p = 0.0428 |
| Residual | 897 324 | 55 | 16 315 | ||
| Tukey's multiple comparisons test | Predicted (LS) mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| CTRL vs. NASPM | 1.157 | −134.3 to 136.6 | > 0.9999 |
| CTRL vs. ketamine | −85.98 | −210.7 to 38.70 | 0.2718 |
| CTRL vs. ketamine + NASPM | 52.48 | −72.20 to 177.2 | 0.6818 |
| NASPM vs. ketamine | −87.14 | −214.7 to 40.45 | 0.2799 |
| NASPM vs. ketamine + NASPM | 51.32 | −76.27 to 178.9 | 0.7116 |
| Ketamine vs. ketamine + NASPM | 138.5 | 22.39 to 254.5 | 0.0133 |
3.2. CRS‐Induced Social Dysfunction Is Reversed by Ketamine via the Expression of CP‐AMPARs
Repeated stress can induce the development of social dysfunction [30]. Importantly, it has been shown that an increase in hippocampal activity reverses stress‐induced social dysfunction [47, 48]. We thus conducted a reciprocal social interaction test to examine whether chronic stress induced social dysfunction and if ketamine reversed this via CP‐AMPARs. For chronic stress in mice, we used chronic restraint stress (CRS) because it has been widely used as a model of chronic psychoemotional stress to induce depression‐ and anxiety‐like behaviors, learning and memory deficits, social dysfunction, and hippocampal neuronal damage in mice [84, 85, 86]. After restraint stress, mice were returned to the home cage. Following 2‐week CRS, the reciprocal social interaction test was carried out as shown in the experimental timeline in Figure 1. First, we found that CRS significantly decreased the total number of reciprocal interactions (FEMALE: Naïve, 32.67 ± 8.41 and CRS, 16.83 ± 4.31, p = 0.0425. MALE: Naïve, 33.00 ± 10.94 and CRS, 16.77 ± 7.18, p = 0.0028) and total time of reciprocal interactions (FEMALE: Naïve, 16.28 ± 3.55 s and CRS, 7.37 ± 4.83 s, p = 0.0315. MALE: Naïve, 19.03 ± 6.56 s and CRS, 9.02 ± 2.22 s, p = 0.0026) in both female and male mice when compared to Naïve controls, an indication of social dysfunction (Figure 3, Tables 3 and 4). This suggests that CRS significantly disrupted social behavior in animals. We then injected low‐dose ketamine to CRS animals and examined whether ketamine reversed CRS‐induced social dysfunction 1 h after the treatment. We discovered that ketamine markedly increased the total number of reciprocal interactions (FEMALE: CRS + Ket, 44.29 ± 12.41, p < 0.0001. MALE: CRS + Ket, 33.25 ± 11.83, p = 0.0001) and total time of reciprocal interactions (FEMALE: CRS + Ket, 23.09 ± 6.29 s, p < 0.0001. MALE: CRS + Ket, 17.71 ± 8.13 s, p = 0.0009) in both female and male mice (Figure 3, Tables 3 and 4). This suggests that subanesthetic low‐dose ketamine can restore normal social behavior in chronically stressed animals. Finally, we tested whether these ketamines effects on social behavior were mediated by CP‐AMPAR expression; we treated IEM‐1460 (IEM), a BBB‐permeable CP‐AMPAR antagonist, with low‐dose ketamine in CRS mice and carried out the social behavior test. We revealed that IEM treatment significantly reduced the total number of reciprocal interactions (FEMALE: CRS + Ket + IEM, 19.23 ± 9.08, p < 0.0001. MALE: CRS + Ket + IEM, 16.61 ± 7.33, p < 0.0001) and total time of reciprocal interactions (FEMALE: CRS + Ket + IEM, 11.60 ± 5.03 s, p < 0.0001. MALE: CRS + Ket + IEM, 12.04 ± 4.38 s, p = 0.0286) in both female and male mice when compared to ketamine‐treated CRS mice (Figure 3, Tables 3 and 4). These findings demonstrate that CRS impairs animals' social behavior, which is reversed by low‐dose ketamine treatment, and the ketamines effects are mediated by CP‐AMPARs.
FIGURE 3.

CRS‐induced social dysfunction is reversed by ketamine via the expression of CP‐AMPARs. Summary of total numbers and time of reciprocal interactions in each condition (n = number of animals. FEMALE: Naïve = 5, CRS = 6, CRS + Ket = 14, and CRS + Ket + IEM = 13. MALE: Naïve = 7, CRS = 12, CRS + Ket = 14, and CRS + Ket + IEM = 17). *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001, One‐Way ANOVA, Tukey test. Ketamine: 5 mg/kg for females and 10 mg/kg for males. IEM‐1460: 10 mg/kg for both females and males.
TABLE 3.
Statistical analysis of the total numbers of reciprocal interactions in the reciprocal social interaction test.
| Female | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 5447 | 3 | 1816 | F (3, 35) = 18.49 | p < 0.0001 |
| Residual (within columns) | 3437 | 35 | 98.21 | ||
| Total | 8885 | 38 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | 15.83 | 0.4028 to 31.26 | 0.0425 |
| Naive vs. CRS + Ket | −11.62 | −24.66 to 1.422 | 0.0952 |
| Naive vs. CRS + Ket + IEM | 13.44 | 0.2451 to 26.63 | 0.0446 |
| CRS vs. CRS + Ket | −27.45 | −40.49 to −14.41 | < 0.0001 |
| CRS vs. CRS + Ket + IEM | −2.397 | −15.59 to 10.79 | 0.9608 |
| CRS + Ket vs. CRS + Ket + IEM | 25.05 | 14.76 to 35.35 | < 0.0001 |
| Male | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 3594 | 3 | 1198 | F (3, 50) = 13.76 | p < 0.0001 |
| Residual (within columns) | 4352 | 50 | 87.03 | ||
| Total | 7945 | 53 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | 16.23 | 4.608 to 27.85 | 0.0028 |
| Naive vs. CRS + Ket | −0.2500 | −11.49 to 10.99 | > 0.9999 |
| Naive vs. CRS + Ket + IEM | 16.39 | 5.345 to 27.43 | 0.0014 |
| CRS vs. CRS + Ket | −16.48 | −25.74 to −7.223 | 0.0001 |
| CRS vs. CRS + Ket + IEM | 0.1581 | −8.866 to 9.182 | > 0.9999 |
| CRS + Ket vs. CRS + Ket + IEM | 16.64 | 8.120 to 25.16 | < 0.0001 |
TABLE 4.
Statistical analysis of the total time of reciprocal interactions in the reciprocal social interaction test.
| Female | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 1401 | 3 | 466.9 | F (3, 35) = 16.40 | p < 0.0001 |
| Residual (within columns) | 996.4 | 35 | 28.47 | ||
| Total | 2397 | 38 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | 8.917 | 0.6088 to 17.22 | 0.0315 |
| Naive vs. CRS + Ket | −6.802 | −13.82 to 0.2191 | 0.0605 |
| Naive vs. CRS + Ket + IEM | 4.683 | −2.419 to 11.79 | 0.3005 |
| CRS vs. CRS + Ket | −15.72 | −22.74 to −8.698 | < 0.0001 |
| CRS vs. CRS + Ket + IEM | −4.233 | −11.34 to 2.869 | 0.3876 |
| CRS + Ket vs. CRS + Ket + IEM | 11.49 | 5.943 to 17.03 | < 0.0001 |
| Male | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 788.4 | 3 | 262.8 | F (3, 50) = 8.035 | p = 0.0002 |
| Residual (within columns) | 1635 | 50 | 32.71 | ||
| Total | 2424 | 53 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | 10.01 | 2.888 to 17.14 | 0.0026 |
| Naive vs. CRS + Ket | 1.322 | −5.565 to 8.210 | 0.9563 |
| Naive vs. CRS + Ket + IEM | 6.990 | 0.2196 to 13.76 | 0.0406 |
| CRS vs. CRS + Ket | −8.691 | −14.37 to −3.016 | 0.0009 |
| CRS vs. CRS + Ket + IEM | −3.024 | −8.555 to 2.508 | 0.4734 |
| CRS + Ket vs. CRS + Ket + IEM | 5.667 | 0.4452 to 10.89 | 0.0286 |
3.3. A CRS‐Induced Loss of Fear Learning and Memory Is Reversed by Ketamine via the Expression of CP‐AMPARs
Repeated stress can impair learning and memory [87]. In fact, it has been shown that CRS disrupts hippocampus‐dependent fear learning and memory in mice [84]. We thus examined whether low‐dose ketamine reversed a CRS‐induced loss of hippocampus‐dependent fear learning and memory via the expression of CP‐AMPARs. To measure hippocampus‐dependent fear learning and memory, CFC was used [50]. As shown previously [84], we found that freezing was significantly lower in CRS female and male animals compared to naïve controls, an indication of fear memory loss (FEMALE: Naïve, 33.55% ± 11.50% and CRS, 15.24% ± 5.64%, p = 0.0026. MALE: Naïve, 26.45% ± 11.14% and CRS, 16.35% ± 7.64%, p = 0.0133) (Figure 4 and Table 5). Next, low‐dose ketamine was intraperitoneally injected to CRS female and male mice, and we carried out CFC to address the ketamine's effects on fear memory. We discovered that ketamine treatment significantly increased freezing in CRS female and male mice (FEMALE: CRS + Ket, 28.16% ± 13.92%, p = 0.0454. MALE: CRS + Ket, 27.79% ± 11.50%, p = 0.0021) (Figure 4 and Table 5). This suggests that subanesthetic low‐dose ketamine can restore normal hippocampus‐dependent fear memory in chronically stressed animals. To address that this ketamine's effect was mediated by CP‐AMPAR expression, we treated ketamine and IEM together in CRS mice. We revealed that blocking CP‐AMPARs markedly reduced freezing in ketamine‐treated CRS female and male animals compared to ketamine‐treated CRS mice (FEMALE: CRS + Ket + IEM, 11.65% ± 4.45%, p = 0.0092. MALE: CRS + Ket + IEM, 19.06% ± 5.34%, p = 0.0322) (Figure 4 and Table 5). These data suggest that CRS disrupts hippocampus‐dependent fear memory in animals, which is reversed by low‐dose ketamine treatment, and the ketamine's effect is dependent on CP‐AMPARs. Because we also injected ketamine and IEM‐1460 on Day 1, a learning period, it is possible that ketamine reverses CRS‐induced impairment of hippocampus‐dependent fear learning via CP‐AMPARs.
FIGURE 4.

A loss of CRS‐induced fear learning and memory is reversed by ketamine via the expression of CP‐AMPARs. Summary of fear memory in each condition (n = number of animals. FEMALE: Naïve = 8, CRS = 9, CRS + Ket = 8, and CRS + Ket + IEM = 8. MALE: Naïve = 9, CRS = 15, CRS + Ket = 11, and CRS + Ket + IEM = 14). *p < 0.05 and **p < 0.01, One‐Way ANOVA, Tukey test. Ketamine: 5 mg/kg for females and 10 mg/kg for males. IEM‐1460: 10 mg/kg for both females and males.
TABLE 5.
Statistical analysis of contextual fear conditioning.
| Female | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 2638 | 3 | 879.4 | F (3, 29) = 9.531 | p = 0.0002 |
| Residual (within columns) | 2676 | 29 | 92.27 | ||
| Total | 5314 | 32 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | 18.31 | 5.595 to 31.03 | 0.0026 |
| Naive vs. CRS + Ket | 5.393 | −7.692 to 18.48 | 0.6786 |
| Naive vs. CRS + Ket + IEM | 21.90 | 8.811 to 34.98 | 0.0005 |
| CRS vs. CRS + Ket | −12.92 | −25.63 to −0.2016 | 0.0454 |
| CRS vs. CRS + Ket + IEM | 3.584 | −9.132 to 16.30 | 0.8682 |
| CRS + Ket vs. CRS + Ket + IEM | 16.50 | 3.417 to 29.59 | 0.0092 |
| Male | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 1164 | 3 | 387.9 | F (3, 46) = 6.701 | p = 0.0008 |
| Residual (within columns) | 2663 | 46 | 57.89 | ||
| Total | 3826 | 49 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | 10.11 | 1.656 to 18.56 | 0.0133 |
| Naive vs. CRS + Ket | −1.334 | −10.45 to 7.781 | 0.9796 |
| Naive vs. CRS + Ket + IEM | 7.394 | −1.270 to 16.06 | 0.1190 |
| CRS vs. CRS + Ket | −11.44 | −19.38 to −3.496 | 0.0021 |
| CRS vs. CRS + Ket + IEM | −2.712 | −10.13 to 4.710 | 0.7649 |
| CRS + Ket vs. CRS + Ket + IEM | 8.728 | 0.5568 to 16.90 | 0.0322 |
3.4. CRS‐Induced Depression‐Like Behavior Is Reversed by Ketamine via the Expression of CP‐AMPARs
Repeated stress is a known trigger for depression in humans and depression‐like behavior in animals [88, 89]. Indeed, CRS is known to induce depression‐like behavior in rodents [90]. Moreover, disruptions in AMPAR‐mediated synaptic transmission in the hippocampus are associated with stress responses in animal models and individuals with depression [33]. We thus employed TST to examine whether low‐dose ketamine reversed CRS‐induced depression‐like behavior in mice via the expression of CP‐AMPARs. As shown previously [90], CRS significantly elevated immobility in female and male mice when compared to Naïve controls, an indication of depression‐like behavior (FEMALE: Naïve, 97.07 ± 42.05 s and CRS, 168.80 ± 40.18 s, p = 0.0130. MALE: Naïve, 78.80 ± 29.96 s and CRS, 139.10 ± 36.04 s, p = 0.0016) (Figure 5 and Table 6). Importantly, low‐dose ketamine treatment markedly decreased immobility in CRS female and male animals, an indication of antidepressant effects (FEMALE: CRS + Ket, 79.43 ± 35.91 s, p = 0.0006. MALE: CRS + Ket, 90.25 ± 37.03 s, p = 0.0066) (Figure 5 and Table 6). We further discovered that immobility in IEM and ketamine‐treated CRS mice was significantly higher than that of ketamine‐treated CRS mice (FEMALE: CRS + Ket + IEM, 131.50 ± 61.21 s, p = 0.0472. MALE: CRS + Ket + IEM, 128.30 ± 31.56 s, p = 0.0400) (Figure 5 and Table 6). The data show that CRS induces depression‐like behavior in animals, which is reversed by low‐dose ketamine treatment via the expression of CP‐AMPARs.
FIGURE 5.

CRS‐induced depression‐like behavior is reversed by ketamine via the expression of CP‐AMPARs. Summary of immobility in each condition (n = number of animals. FEMALE: Naïve = 9, CRS = 8, CRS + Ket = 13, and CRS + Ket + IEM = 10. MALE: Naïve = 10, CRS = 10, CRS + Ket = 14, and CRS + Ket + IEM = 11). *p < 0.05, **p < 0.01, and ***p < 0.001, One‐Way ANOVA, Tukey test. Ketamine: 5 mg/kg for females and 10 mg/kg for males. IEM‐1460: 10 mg/kg for both females and males.
TABLE 6.
Statistical analysis of the tail suspension test.
| Female | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 45 229 | 3 | 15 076 | F (3, 36) = 7.272 | p = 0.0006 |
| Residual (within columns) | 74 637 | 36 | 2073 | ||
| Total | 119 866 | 39 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | −71.77 | −131.4 to −12.18 | 0.0130 |
| Naive vs. CRS + Ket | 17.64 | −35.54 to 70.81 | 0.8084 |
| Naive vs. CRS + Ket + IEM | −34.42 | −90.77 to 21.92 | 0.3669 |
| CRS vs. CRS + Ket | 89.41 | 34.30 to 144.5 | 0.0006 |
| CRS vs. CRS + Ket + IEM | 37.35 | −20.82 to 95.52 | 0.3239 |
| CRS + Ket vs. CRS + Ket + IEM | −52.06 | −103.6 to −0.4780 | 0.0472 |
| Male | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 27 140 | 3 | 9047 | F (3, 41) = 7.801 | p = 0.0003 |
| Residual (within columns) | 47 549 | 41 | 1160 | ||
| Total | 74 689 | 44 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | −60.31 | −101.1 to −19.53 | 0.0016 |
| Naive vs. CRS + Ket | −11.45 | −49.20 to 26.30 | 0.8484 |
| Naive vs. CRS + Ket + IEM | −49.48 | −89.32 to −9.640 | 0.0097 |
| CRS vs. CRS + Ket | 48.86 | 11.11 to 86.61 | 0.0066 |
| CRS vs. CRS + Ket + IEM | 10.83 | −29.01 to 50.67 | 0.8854 |
| CRS + Ket vs. CRS + Ket + IEM | −38.03 | −74.77 to −1.292 | 0.0400 |
3.5. No Abnormality in the Open Field Test
As animals' locomotor capacities can influence behavior, we carried out OFT to examine whether there were any abnormalities in animals' locomotion in each condition. We found that there was no difference in total distance traveled, total time spent outside, and total time spent inside between the conditions (Figure 6 and Table 7). Moreover, as time spent outside and inside can be interpreted as anxiety‐like behavior [46], normal behaviors in OFT indicate that the animals used in our study had normal locomotor activity and no anxiety‐like behavior. These findings suggest that the behavioral alterations we described above are not due to changes in animals' movement and anxiety levels.
FIGURE 6.

No abnormality in the open field test. Summary of total distance traveled, total time spent outside, and total time spent inside in each condition (n = number of animals. FEMALE: Naïve = 9, CRS = 9, CRS + Ket = 16, and CRS + Ket + IEM = 14. MALE: Naïve = 8, CRS = 15, CRS + Ket = 16, and CRS + Ket + IEM = 18). Ketamine: 5 mg/kg for females and 10 mg/kg for males. IEM‐1460: 10 mg/kg for both females and males.
TABLE 7.
Summary of the open field test.
| FEMALE | MALE | |||||||
|---|---|---|---|---|---|---|---|---|
| CTRL | CRS | CRS + Ket | CRS + Ket + IEM | CTRL | CRS | CRS + Ket | CRS + Ket + IEM | |
| Total distance traveled (m) | 37.52 ± 15.31 | 45.02 ± 12.62 | 47.37 ± 16.47 | 45.43 ± 12.24 | 36.55 ± 9.70 | 42.37 ± 12.86 | 39.34 ± 10.56 | 40.07 ± 14.65 |
| Total time spent outside (sec) | 962.8 ± 85.57 | 989.6 ± 122.0 | 1029 ± 115.9 | 991.8 ± 106.0 | 947.6 ± 167.2 | 1044 ± 60.74 | 1042 ± 101.2 | 975.9 ± 128.1 |
| Total time spent inside (sec) | 237.2 ± 85.57 | 210.4 ± 122.0 | 170.8 ± 115.9 | 208.2 ± 106.0 | 252.4 ± 167.2 | 155.6 ± 60.74 | 158.4 ± 101.2 | 224.1 ± 128.1 |
3.6. Ketamine Reverses the CRS‐Induced Elevation of Corticosterone Levels, Which Is Not Dependent on the Expression of CP‐AMPARs
Chronic stress activates the hypothalamic–pituitary–adrenal axis, resulting in the release of glucocorticoid class stress hormones [91, 92]. Cortisol is the primary endogenous adrenal steroid in most mammals, including humans, whereas corticosterone is the primary adrenal corticosteroid in rodents [93, 94, 95, 96]. Chronic stress can keep these hormone levels high, which in turn alters brain functions [91, 97]. In fact, CRS is known to increase corticosterone levels in animals, which is associated with stress‐induced behavioral changes [98]. We thus examined whether low‐dose ketamine reversed the CRS‐induced elevation of corticosterone levels via the expression of CP‐AMPARs. As shown previously [99], we found that CRS significantly increased serum corticosterone levels in both female and male animals when compared to naïve mice (FEMALE: Naïve, 831.1 ± 376.8 pg/mL and CRS, 2374 ± 1101 pg/mL, p = 0.0001. MALE: Naïve, 756.1 ± 210.6 pg/mL and CRS, 2140 ± 721.1 pg/mL, p = 0.0005) (Figure 7 and Table 8). Ketamine is known to significantly decrease the CRS‐induced increase in serum corticosterone levels in mice [99]. Consistent with this finding, we discovered that low‐dose ketamine significantly reduced serum corticosterone levels in CRS animals (FEMALE: CRS + Ket, 834.3 ± 372.0 pg/mL, p = 0.0001. MALE: CRS + Ket, 1089 ± 851.3 pg/mL, p = 0.0146) (Figure 7 and Table 8). Finally, we tested whether the ketamine's effects on corticosterone levels were mediated by CP‐AMPARs. In contrast to behavioral assays, serum corticosterone levels of ketamine‐treated CRS animals did not differ in the presence or absence of IEM (FEMALE: CRS + Ket + IEM, 1095 ± 481.9 pg/mL, p = 0.9247. MALE: CRS + Ket + IEM, 1082 ± 383.5 pg/mL, p > 0.9999) (Figure 7 and Table 8). The data show that low‐dose ketamine treatment reverses the CRS‐induced increase in serum corticosterone levels, which is independent of the expression of CP‐AMPARs.
FIGURE 7.

Ketamine reverses the CRS‐induced elevation of corticosterone levels, which is not dependent on the expression of CP‐AMPARs. Summary of serum corticosterone levels in each condition (n = number of animals with duplication. FEMALE: Naïve = 5, CRS = 5, CRS + Ket = 5, and CRS + Ket + IEM = 2. MALE: Naïve = 4, CRS = 4, CRS + Ket = 3, and CRS + Ket + IEM = 3). *p < 0.05 and ***p < 0.001, One‐Way ANOVA, Tukey test. n.s.—not significant. Ketamine: 5 mg/kg for females and 10 mg/kg for males. IEM‐1460: 10 mg/kg for both females and males.
TABLE 8.
Statistical analysis of corticosterone levels.
| Female | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 15 981 461 | 3 | 5 327 154 | F (3, 30) = 11.31 | p < 0.0001 |
| Residual (within columns) | 14 132 346 | 30 | 471 078 | ||
| Total | 30 113 807 | 33 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | −1543 | −2378 to −708.6 | 0.0001 |
| Naive vs. CRS + Ket | −12.17 | −846.8 to 822.4 | > 0.9999 |
| Naive vs. CRS + Ket + IEM | −264.2 | −1368 to 839.9 | 0.9145 |
| CRS vs. CRS + Ket | 1531 | 696.5 to 2366 | 0.0001 |
| CRS vs. CRS + Ket + IEM | 1279 | 175.0 to 2383 | 0.0183 |
| CRS + Ket vs. CRS + Ket + IEM | −252.0 | −1356 to 852.1 | 0.9247 |
| Male | |||||
|---|---|---|---|---|---|
| ANOVA table | SS | DF | MS | F (DFn, DFd) | p |
| Treatment (between columns) | 8 562 805 | 3 | 2 854 268 | F (3, 24) = 8.244 | P = 0.0006 |
| Residual (within columns) | 8 309 607 | 24 | 346 234 | ||
| Total | 16 872 412 | 27 | |||
| Tukey's multiple comparisons test | Mean diff. | 95.00% CI of diff. | Adjusted p |
|---|---|---|---|
| Naive vs. CRS | −1384 | −2196 to −572.3 | 0.0005 |
| Naive vs. CRS + Ket | −332.8 | −1209 to 543.9 | 0.7239 |
| Naive vs. CRS + Ket + IEM | −325.8 | −1202 to 550.9 | 0.7366 |
| CRS vs. CRS + Ket | 1051 | 174.5 to 1928 | 0.0146 |
| CRS vs. CRS + Ket + IEM | 1058 | 181.5 to 1935 | 0.0139 |
| CRS + Ket vs. CRS + Ket + IEM | 7.000 | −930.2 to 944.2 | > 0.9999 |
4. Discussion and Conclusions
4.1. Ketamine's Unknown Mechanisms
It is challenging to understand how ketamine works due to its complex dose‐dependent effects, with multiple pharmacologically active metabolites and various mechanisms associated with the facilitation of synaptic plasticity [1]. Importantly, the subanesthetic low‐dose ketamine‐induced increase of glutamatergic synaptic strength is presumed to underlie antidepressant effects in humans and animals [16, 17, 18]. These actions of ketamine are likely mediated by enhancing AMPAR activity due to its nature of NMDAR antagonism [20]. However, the entire role of AMPARs in the actions of ketamine remains undetermined. Additionally, treatment with ketamine is associated with psychomimetic side effects and risk of addiction, which prevent its broader application [6]. Therefore, it is critical to identify the mechanisms of ketamine's effects, particularly on glutamatergic synapses, to guide the development of safer fast‐acting therapy.
4.2. The Role of CP‐AMPARs in Ketamine‐Induced Antistress Effects
Disruptions of AMPAR‐mediated synaptic transmission in diverse cell types in the hippocampus are one of the major biological changes in chronically stressed brains of animal models and individuals with depression [33]. Importantly, recovery from GluA1‐mediated synaptic dysfunction is thought to be the key target of ketamine's antidepressant and antistress effects. Moreover, ketamine‐induced recovery from disruptions in AMPAR‐mediated synaptic transmission and behaviors in stressed animals is suggested to be associated with an increase in synaptic GluA1 expression in the hippocampus [52, 53]. In fact, we have discovered that low‐dose ketamine rapidly induces the expression of CP‐AMPARs in cultured hippocampal neurons, which is mediated by increasing GluA1 phosphorylation and its surface expression through a reduction of neuronal Ca2+ and Ca2+‐dependent phosphatase calcineurin (CaN) activity [46] (Figure 8). In particular, GluA1‐containing CP‐AMPARs have higher single channel conductance, which contributes to synaptic plasticity [35, 36, 37, 38, 39, 40, 41, 42, 43, 44]. We have indeed shown that ketamine‐induced CP‐AMPAR expression enhances glutamatergic synaptic strength and chemically induced long‐term potentiation (cLTP) in cultured neurons [46]. cLTP is shown to rely on a rise in postsynaptic Ca2+ concentration, specifically through the activation of NMDARs [100]. Therefore, when ketamine is treated, CP‐AMPAR‐mediated Ca2+ influx may replace NMDAR‐dependent Ca2+ signaling. We further show that subanesthetic low‐dose ketamine decreases anxiety and depression‐like behaviors in naïve animals [46]. Importantly, an increase in hippocampal activity is known to improve stress‐induced negative behaviors in mice [16, 17, 18, 47, 48, 49, 50, 51], which further supports our findings. However, the mechanisms of ketamine's protective effects against stress‐induced disorders are not completely understood. Here, our electrophysiological data demonstrate that low‐dose ketamine induces CP‐AMPAR expression in pyramidal neurons of the hippocampus CA1 area. Additionally, the current study reveals that subanesthetic low‐dose ketamine can rapidly reverse social dysfunction, fear memory loss, and depression‐like behavior caused by CRS in mice via CP‐AMPAR expression (Figure 8). This suggests that low‐dose ketamine induces the expression of CP‐AMPARs in the hippocampus, which in turn enhances synaptic strength to reverse hippocampus‐dependent behavioral dysfunctions in chronically stressed animals.
FIGURE 8.

A schematic model of ketamine's antistress effects. Because ketamine is a noncompetitive NMDAR antagonist, a therapeutic dose is enough to block NMDAR‐mediated Ca2+ influx in excitatory synapses. This can lower the activity of calcineurin (CaN), a Ca2+‐dependent phosphatase that dephosphorylates the AMPA receptor (AMPAR) subunit GluA1, and promote GluA1 phosphorylation, resulting in increased synaptic expression of GluA2‐lacking, GluA1‐containing Ca2+‐permeable AMPARs (CP‐AMPARs). When ketamine is administered, CP‐AMPAR‐mediated Ca2+ influx may replace NMDAR‐dependent Ca2+ signaling. This increases neural plasticity, which leads to antistress effects in animals. Therefore, inhibition of CP‐AMPARs using antagonists such as NASPM and IEM‐1460 prevents the ketamine's effects.
4.3. The Ketamine's Rapid Onset and Long‐Lasting Effects
Given its brief half‐life (3 h in humans and 13 min in mice), the ketamine's rapid onset and long‐lasting effects (which persist for days in humans and 24 h in mice) are not fully understood by synaptic mechanisms [7]. One potential mechanism underlying the ketamine's rapid onset and long‐lasting effects is that ketamine induces neural plasticity [15]. Interestingly, homeostatic synaptic plasticity has been implicated in ketamine's rapid action due to ketamine blocking NMDARs that are involved in a specific form of neurotransmission to impact intracellular signaling, whereas long‐lasting effects likely involve structural plasticity [16, 101, 102, 103, 104]. However, still unclear is when and how the plasticity is induced. Specifically, molecular and cellular processes of how ketamine induces neural plasticity remain to be elucidated. In addition to strengthening excitatory synaptic drive, low‐dose ketamine can enhance neuronal Ca2+ signaling in the hippocampus [105], possibly contributing to the ketamine's rapid onset and long‐lasting effects. Although the relationship between ketamine‐triggered alteration in Ca2+ signaling and functional consequences for downstream processes regulating key aspects of neuronal function is known to be important for the antidepressant and antistress effects [106], how ketamine enhances synaptic Ca2+ signaling is not fully understood. Given that CP‐AMPARs are known to contribute to various forms of synaptic plasticity [35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 107, 108, 109], CP‐AMPAR‐mediated neural plasticity can facilitate the ketamine's effects. Therefore, our findings in which low‐dose ketamine rapidly induces hippocampal CP‐AMPARs can be significant because ketamine can enhance not only synaptic transmission but also synaptic Ca2+ signaling in response to partial antagonism of NMDARs (Figure 8). Therefore, the ketamine‐induced expression of CP‐AMPARs can increase glutamatergic activity and compensate for reduced NMDAR inhibition‐mediated synaptic Ca2+ signaling, enabling the ketamine's rapid onset and long‐lasting effects.
4.4. Sex Differences in Ketamine's Effects
Our work shows an important finding: female mice have higher ketamine antidepressant responses than male mice, as seen by synaptic insertion of CP‐AMPARs following a lower ketamine dosage, which induces the antistress effects. Several studies including both male and female animals consistently demonstrate that females are more sensitive to ketamine [110, 111, 112, 113]. These studies collectively suggest that hormonal differences and metabolic factors contribute to the heightened sensitivity of females to ketamine's effects at lower doses. In addition, the distinct pharmacokinetics of ketamine in the animals' brains and plasma could be one reason for the increased ketamine's antistress reactions in female mice [114]. Over the course of 3 h after treatment, female rats' medial prefrontal cortex and hippocampus contain higher levels of ketamine and norketamine, a metabolite of ketamine, than do male rats [114]. The study also shows that female rats' slower clearance rates and longer half‐lives result in higher post‐treatment effects of ketamine and its metabolites [114]. Unfortunately, the potential sex differences in response to ketamine have been particularly understudied at this time, which is an important area for future investigation in order to develop tailored treatment and dosing in clinics.
4.5. Multiple Mechanisms Underlying Ketamine's Effects
The mechanisms underlying ketamine's antidepressant effects are not fully understood, although multiple ideas have been proposed. These hypotheses include (1) direct NMDAR inhibition‐mediated mechanism in excitatory neurons [16, 17, 24, 46, 115, 116]; (2) NMDAR inhibition‐independent, ketamine metabolite‐dependent mechanism [110, 111, 112]; and (3) NMDAR inhibition‐induced disinhibition‐ and BDNF‐induced protein synthesis‐dependent mechanism [117, 118]. Low‐dose MK‐801, an NMDAR antagonist, has been shown to improve depression‐like behavior and working memory in mice [119, 120]. Conversely, others suggest that the metabolism of ketamine to (2R,6R)‐hydroxynorketamine (HNK), devoid of NMDAR inhibition properties, is important for the antidepressant effect [110, 111, 112]. Although multiple studies, including our own, support the NMDAR inhibition‐mediated mechanisms, it is important to investigate how our current findings integrate or act independently from other described mechanisms.
4.6. Brain Region‐Specific Actions of Ketamine
Research identifying the mechanisms of the antidepressant effects of ketamine has largely focused on the hippocampus, since it is known to be dysfunctional in depressive disorders [110, 121, 122, 123]. More recently, other areas of the brain have been investigated, including the lateral habenula [24, 115, 116]. A recent study shows that in CRS animals, ketamine selectively inhibited NMDARs in lateral habenula neurons, but not in hippocampal pyramidal neurons. This suggests that the lateral habenula–NMDAR inhibition–dependent mechanism likely occurs more upstream in the cascade of ketamine signaling in vivo [115]. In contrast, another recent study using a systematic and unbiased mapping approach that provides a comprehensive coverage of all brain regions discovers that ketamine preferably targets the hippocampus [124]. The hippocampus is a crucial brain area that governs learning, memory, social interaction, and depression [51, 125, 126, 127, 128, 129]. Research also demonstrates that an increase in hippocampal activity can be protective against stress [16, 17, 18, 47, 48, 49, 50, 51]. Importantly, one of the most vulnerable targets of stress is the hippocampus, because it abundantly expresses both glucocorticoid and mineralocorticoid receptors [130, 131]. Moreover, stress changes hippocampal neural activity and synaptic plasticity, activating hippocampal glucocorticoid receptors, and decreasing neuronal cell survival and neurogenesis [132, 133, 134]. This suggests that the hippocampus is the important brain region responsible for ketamine's effects. Since ketamine is delivered systemically in most studies, many regions in the brain can potentially be responsive to ketamine. In fact, different brain regions are involved in controlling various aspects of animals' behaviors, including movement, emotion, cognition, learning, and innate responses crucial for survival. Given that ketamine is systemically administered in the current study, it is possible that the ketamine‐induced antistress behavior outcomes involve brain regions outside or in addition to the hippocampus. Therefore, it is crucial to identify the crosstalk from the hippocampus to other brain regions, including the lateral habenula, for better understanding of ketamine's therapeutic effects.
4.7. Potential Role of Ketamine's Anti‐Inflammatory Properties
Chronic stress is known to increase the release of corticosterone in animals [91, 92]. In fact, we discover that ketamine significantly reduces serum corticosterone levels in CRS mice, which is consistent with the previous finding [99]. In contrast to ketamine‐induced protective effects against CRS‐induced behavioral changes, this effect is not dependent on CP‐AMPAR expression. Interestingly, chronic stress can induce disorders through increased pre‐inflammatory cytokines, reactive microglia numbers, and up‐regulated regulatory molecules [135]. Moreover, chronic inflammatory responses directly affect brain structure and function, resulting in pathological behaviors in animals [136]. Importantly, ketamine is known to be associated with anti‐inflammatory effects in humans [137, 138] and animals [139, 140, 141], suggesting that its ability to reduce inflammation may play a role in its protective effects against stress. Therefore, it is possible that the mechanism underlying the ketamine‐induced decrease in stress hormones following chronic stress can be mediated by the hypothalamic–pituitary–adrenal axis rather than the hippocampus. It is thus possible that hypothalamic neurons may not express CP‐AMPARs in response to ketamine treatment. In fact, there are multiple mechanisms proposed to explain ketamine's anti‐inflammatory effects, including the involvement of the gut microbiome [142, 143] and priming of Type 1 T helper (Th1)‐type immune response [144]. However, the clinical relevance of ketamine‐induced inflammatory modulation is still unclear. Therefore, further studies are needed to identify the precise mechanisms of its anti‐inflammatory properties.
5. Conclusions
The current study addresses how chronic stress and ketamine affect hippocampal synaptic activity and hippocampus‐dependent behaviors, providing a new evidence‐based idea that NMDAR inhibition by ketamine leads to the antistress effects through synaptic insertion of CP‐AMPARs. Therefore, our work advances current understandings of the mechanisms of ketamine's antidepressant and antistress effects to guide the developments of new therapies.
Author Contributions
Sang‐Hun Lee and Seonil Kim designed research; Seonil Kim analyzed data; Joshua C. Flowers, Paige E. Vetter, McKennon J. Wiles, Seung Hyun Roh, Ellison R. Black, Evelina Bouckova, Madison H. Wustrau, and Rahmi Lee, performed research; Joshua C. Flowers, Paige E. Vetter, McKennon J. Wiles, Seung Hyun Roh, and Seonil Kim wrote the paper.
Conflicts of Interest
The authors declare no conflicts of interest.
Acknowledgments
We thank members of the Kim laboratory for their generous support. This work is supported by Student Experiential Learning Grants and the College Research Council Shared Research Program from the College of Veterinary Medicine and Biomedical Sciences, Colorado State University, and a research grant from Korea Ginseng Corporation.
Flowers J. C., Vetter P. E., Wiles M. J., et al., “Ketamine Reverses Chronic Stress‐Induced Behavioral Changes via the Expression of Ca2+‐Permeable AMPA Receptors in Mice,” The FASEB Journal 39, no. 16 (2025): e70891, 10.1096/fj.202501763R.
Funding: This work was supported by CSU College of Veterinary Medicine and Biomedical Sciences, Colorado State University (CVMBS, CSU), Korea Ginseng Corporation (KGC).
Joshua C. Flowers, Paige E. Vetter, McKennon J. Wiles, Seung Hyun Roh, Ellison R. Black, Evelina Bouckova, Madison H. Wustrau contributed equally to this study.
Data Availability Statement
Privacy/ethical restrictions.
References
- 1. Kohtala S., “Ketamine‐50 Years in Use: From Anesthesia to Rapid Antidepressant Effects and Neurobiological Mechanisms,” Pharmacological Reports 73 (2021): 323–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Newport D. J., Carpenter L. L., McDonald W. M., et al., “Ketamine and Other NMDA Antagonists: Early Clinical Trials and Possible Mechanisms in Depression,” American Journal of Psychiatry 172 (2015): 950–966. [DOI] [PubMed] [Google Scholar]
- 3. aan het Rot M., Collins K. A., Murrough J. W., et al., “Safety and Efficacy of Repeated‐Dose Intravenous Ketamine for Treatment‐Resistant Depression,” Biological Psychiatry 67 (2010): 139–145. [DOI] [PubMed] [Google Scholar]
- 4. Murrough J. W., Perez A. M., Pillemer S., et al., “Rapid and Longer‐Term Antidepressant Effects of Repeated Ketamine Infusions in Treatment‐Resistant Major Depression,” Biological Psychiatry 74 (2013): 250–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Phillips J. L., Norris S., Talbot J., et al., “Single, Repeated, and Maintenance Ketamine Infusions for Treatment‐Resistant Depression: A Randomized Controlled Trial,” American Journal of Psychiatry 176 (2019): 401–409. [DOI] [PubMed] [Google Scholar]
- 6. Ng S. H., Tse M. L., Ng H. W., and Lau F. L., “Emergency Department Presentation of Ketamine Abusers in Hong Kong: A Review of 233 Cases,” Hong Kong Medical Journal 16 (2010): 6–11. [PubMed] [Google Scholar]
- 7. Silva J. C. H. and Proulx C. D., “Locking Away Depression,” Science 385 (2024): 608–609. [DOI] [PubMed] [Google Scholar]
- 8. Sepulveda Ramos C., Thornburg M., Long K., et al., “The Therapeutic Effects of Ketamine in Mental Health Disorders: A Narrative Review,” Cureus 14 (2022): e23647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Veraart J. K. E., Smith‐Apeldoorn S. Y., Bakker I. M., et al., “Pharmacodynamic Interactions Between Ketamine and Psychiatric Medications Used in the Treatment of Depression: A Systematic Review,” International Journal of Neuropsychopharmacology 24 (2021): 808–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Correia‐Melo F. S., Silva S. S., Araujo‐de‐Freitas L., and Quarantini L. C., “S‐(+)‐Ketamine‐Induced Dissociative Symptoms as a Traumatic Experience in Patients With Treatment‐Resistant Depression,” Braz J Psychiatry 39 (2017): 188–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Paul R., Schaaff N., Padberg F., Moller H. J., and Frodl T., “Comparison of Racemic Ketamine and S‐Ketamine in Treatment‐Resistant Major Depression: Report of Two Cases,” World Journal of Biological Psychiatry: Official Journal of the World Federation of Societies of Biological Psychiatry 10 (2009): 241–244. [DOI] [PubMed] [Google Scholar]
- 12. Szymkowicz S. M., Finnegan N., and Dale R. M., “A 12‐Month Naturalistic Observation of Three Patients Receiving Repeat Intravenous Ketamine Infusions for Their Treatment‐Resistant Depression,” Journal of Affective Disorders 147 (2013): 416–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Serafini G., Howland R. H., Rovedi F., Girardi P., and Amore M., “The Role of Ketamine in Treatment‐Resistant Depression: A Systematic Review,” Current Neuropharmacology 12 (2014): 444–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Papakostas G. I., “Maintaining Rapid Antidepressant Effects Following Ketamine Infusion: A Major Unmet Need,” Journal of Clinical Psychiatry 81 (2020), 10.4088/JCP.19r12859. [DOI] [PubMed] [Google Scholar]
- 15. Wu H., Savalia N. K., and Kwan A. C., “Ketamine for a Boost of Neural Plasticity: How, but Also When?,” Biological Psychiatry 89 (2021): 1030–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kavalali E. T. and Monteggia L. M., “Targeting Homeostatic Synaptic Plasticity for Treatment of Mood Disorders,” Neuron 106 (2020): 715–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Miller O. H., Moran J. T., and Hall B. J., “Two Cellular Hypotheses Explaining the Initiation of Ketamine's Antidepressant Actions: Direct Inhibition and Disinhibition,” Neuropharmacology 100 (2016): 17–26. [DOI] [PubMed] [Google Scholar]
- 18. Aleksandrova L. R., Wang Y. T., and Phillips A. G., “Ketamine and Its Metabolite, (2R,6R)‐HNK, Restore Hippocampal LTP and Long‐Term Spatial Memory in the Wistar‐Kyoto Rat Model of Depression,” Molecular Brain 13 (2020): 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Anis N. A., Berry S. C., Burton N. R., and Lodge D., “The Dissociative Anaesthetics, Ketamine and Phencyclidine, Selectively Reduce Excitation of Central Mammalian Neurones by N‐Methyl‐Aspartate,” British Journal of Pharmacology 79 (1983): 565–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Aleksandrova L. R., Phillips A. G., and Wang Y. T., “Antidepressant Effects of Ketamine and the Roles of AMPA Glutamate Receptors and Other Mechanisms Beyond NMDA Receptor Antagonism,” Journal of Psychiatry & Neuroscience 42 (2017): 222–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ma L., Wang L., Chang L., et al., “A Key Role of miR‐132‐5p in the Prefrontal Cortex for Persistent Prophylactic Actions of (R)‐Ketamine in Mice,” Translational Psychiatry 12 (2022): 417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Ma J. H., Wang S. Y., Yu H. Y., et al., “Prophylactic Use of Ketamine Reduces Postpartum Depression in Chinese Women Undergoing Cesarean Section(✰),” Psychiatry Research 279 (2019): 252–258. [DOI] [PubMed] [Google Scholar]
- 23. Brachman R. A., McGowan J. C., Perusini J. N., et al., “Ketamine as a Prophylactic Against Stress‐Induced Depressive‐Like Behavior,” Biological Psychiatry 79 (2016): 776–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Yang Y., Cui Y., Sang K., et al., “Ketamine Blocks Bursting in the Lateral Habenula to Rapidly Relieve Depression,” Nature 554 (2018): 317–322. [DOI] [PubMed] [Google Scholar]
- 25. Feder A., Parides M. K., Murrough J. W., et al., “Efficacy of Intravenous Ketamine for Treatment of Chronic Posttraumatic Stress Disorder: A Randomized Clinical Trial,” JAMA Psychiatry 71 (2014): 681–688. [DOI] [PubMed] [Google Scholar]
- 26. Lee E. H., Park J. Y., Kwon H. J., and Han P. L., “Repeated Exposure With Short‐Term Behavioral Stress Resolves Pre‐Existing Stress‐Induced Depressive‐Like Behavior in Mice,” Nature Communications 12 (2021): 6682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Taylor J. H., Landeros‐Weisenberger A., Coughlin C., et al., “Ketamine for Social Anxiety Disorder: A Randomized, Placebo‐Controlled Crossover Trial,” Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 43 (2018): 325–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Gill H., Gill B., Rodrigues N. B., et al., “The Effects of Ketamine on Cognition in Treatment‐Resistant Depression: A Systematic Review and Priority Avenues for Future Research,” Neuroscience and Biobehavioral Reviews 120 (2021): 78–85. [DOI] [PubMed] [Google Scholar]
- 29. Zhou Y., Wang C., Lan X., et al., “The Potential Pro‐Cognitive Effects With Intravenous Subanesthetic Ketamine in Adults With Treatment‐Resistant Major Depressive or Bipolar Disorders and Suicidality,” Journal of Psychiatric Research 144 (2021): 312–319. [DOI] [PubMed] [Google Scholar]
- 30. daSilva A. W., Huckins J. F., Wang W., Wang R., Campbell A. T., and Meyer M. L., “Daily Perceived Stress Predicts Less Next Day Social Interaction: Evidence From a Naturalistic Mobile Sensing Study,” Emotion 21 (2021): 1760–1770. [DOI] [PubMed] [Google Scholar]
- 31. Kessler R. C., “The Effects of Stressful Life Events on Depression,” Annual Review of Psychology 48 (1997): 191–214. [DOI] [PubMed] [Google Scholar]
- 32. Lupien S. J., McEwen B. S., Gunnar M. R., and Heim C., “Effects of Stress Throughout the Lifespan on the Brain, Behaviour and Cognition,” Nature Reviews. Neuroscience 10 (2009): 434–445. [DOI] [PubMed] [Google Scholar]
- 33. He J. G., Zhou H. Y., Wang F., and Chen J. G., “Dysfunction of Glutamatergic Synaptic Transmission in Depression: Focus on AMPA Receptor Trafficking,” Biol Psychiatry Glob Open Sci 3 (2023): 187–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Diering G. H. and Huganir R. L., “The AMPA Receptor Code of Synaptic Plasticity,” Neuron 100 (2018): 314–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Purkey A. M. and Dell'Acqua M. L., “Phosphorylation‐Dependent Regulation of ca(2+)‐Permeable AMPA Receptors During Hippocampal Synaptic Plasticity,” Frontiers in Synaptic Neuroscience 12 (2020): 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Sanderson J. L., Gorski J. A., and Dell'Acqua M. L., “NMDA Receptor‐Dependent LTD Requires Transient Synaptic Incorporation of ca(2)(+)‐Permeable AMPARs Mediated by AKAP150‐Anchored PKA and Calcineurin,” Neuron 89 (2016): 1000–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Sanderson J. L., Scott J. D., and Dell'Acqua M. L., “Control of Homeostatic Synaptic Plasticity by AKAP‐Anchored Kinase and Phosphatase Regulation of ca(2+)‐Permeable AMPA Receptors,” Journal of Neuroscience 38 (2018): 2863–2876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Thiagarajan T. C., Lindskog M., and Tsien R. W., “Adaptation to Synaptic Inactivity in Hippocampal Neurons,” Neuron 47 (2005): 725–737. [DOI] [PubMed] [Google Scholar]
- 39. Goel A. and Lee H. K., “Persistence of Experience‐Induced Homeostatic Synaptic Plasticity Through Adulthood in Superficial Layers of Mouse Visual Cortex,” Journal of Neuroscience 27 (2007): 6692–6700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Goel A., Xu L. W., Snyder K. P., et al., “Phosphorylation of AMPA Receptors Is Required for Sensory Deprivation‐Induced Homeostatic Synaptic Plasticity,” PLoS One 6 (2011): e18264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Lee H. K., “Ca‐Permeable AMPA Receptors in Homeostatic Synaptic Plasticity,” Frontiers in Molecular Neuroscience 5 (2012): 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kim S., Violette C. J., and Ziff E. B., “Reduction of Increased Calcineurin Activity Rescues Impaired Homeostatic Synaptic Plasticity in Presenilin 1 M146V Mutant,” Neurobiology of Aging 36 (2015): 3239–3246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Kim S. and Ziff E. B., “Calcineurin Mediates Synaptic Scaling via Synaptic Trafficking of Ca2+−Permeable AMPA Receptors,” PLoS Biology 12 (2014): e1001900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Aoto J., Nam C. I., Poon M. M., Ting P., and Chen L., “Synaptic Signaling by All‐Trans Retinoic Acid in Homeostatic Synaptic Plasticity,” Neuron 60 (2008): 308–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Hare B. D., Shinohara R., Liu R. J., Pothula S., DiLeone R. J., and Duman R. S., “Optogenetic Stimulation of Medial Prefrontal Cortex Drd1 Neurons Produces Rapid and Long‐Lasting Antidepressant Effects,” Nature Communications 10 (2019): 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Zaytseva A., Bouckova E., Wiles M. J., et al., “Ketamine's Rapid Antidepressant Effects Are Mediated by ca(2+)‐Permeable AMPA Receptors,” eLife 12 (2023), 10.7554/eLife.86022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Sun Q., Li X., Li A., et al., “Ventral Hippocampal‐Prefrontal Interaction Affects Social Behavior via Parvalbumin Positive Neurons in the Medial Prefrontal Cortex,” iScience 23 (2020): 100894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Okuyama T., Kitamura T., Roy D. S., Itohara S., and Tonegawa S., “Ventral CA1 Neurons Store Social Memory,” Science 353 (2016): 1536–1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Grieco S. F., Castren E., Knudsen G. M., et al., “Psychedelics and Neural Plasticity: Therapeutic Implications,” Journal of Neuroscience 42 (2022): 8439–8449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Tovote P., Fadok J. P., and Luthi A., “Neuronal Circuits for Fear and Anxiety,” Nature Reviews. Neuroscience 16 (2015): 317–331. [DOI] [PubMed] [Google Scholar]
- 51. Bird C. M. and Burgess N., “The Hippocampus and Memory: Insights From Spatial Processing,” Nature Reviews. Neuroscience 9 (2008): 182–194. [DOI] [PubMed] [Google Scholar]
- 52. Fischell J., Van Dyke A. M., Kvarta M. D., LeGates T. A., and Thompson S. M., “Rapid Antidepressant Action and Restoration of Excitatory Synaptic Strength After Chronic Stress by Negative Modulators of Alpha5‐Containing GABAA Receptors,” Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 40 (2015): 2499–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Li N., Liu R. J., Dwyer J. M., et al., “Glutamate N‐Methyl‐D‐Aspartate Receptor Antagonists Rapidly Reverse Behavioral and Synaptic Deficits Caused by Chronic Stress Exposure,” Biological Psychiatry 69 (2011): 754–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kallarackal A. J., Kvarta M. D., Cammarata E., et al., “Chronic Stress Induces a Selective Decrease in AMPA Receptor‐Mediated Synaptic Excitation at Hippocampal Temporoammonic‐CA1 Synapses,” Journal of Neuroscience 33 (2013): 15669–15674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Kilkenny C., Browne W., Cuthill I. C., Emerson M., Altman D. G., and NC3Rs Reporting Guidelines Working Group , “Animal Research: Reporting In Vivo Experiments: The ARRIVE Guidelines,” British Journal of Pharmacology 160 (2010): 1577–1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Adotevi N., Lewczuk E., Sun H., et al., “Alpha‐Amino‐3‐Hydroxy‐5‐Methyl‐4‐Isoxazolepropionic Acid Receptor Plasticity Sustains Severe, Fatal Status Epilepticus,” Annals of Neurology 87 (2020): 84–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Szczurowska E. and Mares P., “An Antagonist of Calcium Permeable AMPA Receptors, IEM1460: Anticonvulsant Action in Immature Rats?,” Epilepsy Research 109 (2015): 106–113. [DOI] [PubMed] [Google Scholar]
- 58. Wiltgen B. J., Royle G. A., Gray E. E., et al., “A Role for Calcium‐Permeable AMPA Receptors in Synaptic Plasticity and Learning,” PLoS One 5 (2010): e12818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Kang Y. J., Lewis H. E. S., Young M. W., et al., “Cell Type‐Specific Intrinsic Perithreshold Oscillations in Hippocampal GABAergic Interneurons,” Neuroscience 376 (2018): 80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Govindaiah G., Kang Y. J., Lewis H. E. S., et al., “Group I Metabotropic Glutamate Receptors Generate Two Types of Intrinsic Membrane Oscillations in Hippocampal Oriens/Alveus Interneurons,” Neuropharmacology 139 (2018): 150–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kang Y. J., Clement E. M., Sumsky S. L., et al., “The Critical Role of Persistent Sodium Current in Hippocampal Gamma Oscillations,” Neuropharmacology 162 (2020): 107787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Matsuo N., Tanda K., Nakanishi K., et al., “Comprehensive Behavioral Phenotyping of Ryanodine Receptor Type 3 (RyR3) Knockout Mice: Decreased Social Contact Duration in Two Social Interaction Tests,” Frontiers in Behavioral Neuroscience 3, no. 3 (2009), 10.3389/neuro.08.003.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bolivar V. J., Walters S. R., and Phoenix J. L., “Assessing Autism‐Like Behavior in Mice: Variations in Social Interactions Among Inbred Strains,” Behavioural Brain Research 176 (2007): 21–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Shou J., Tran A., Snyder N., Bleem E., and Kim S., “Distinct Roles of GluA2‐Lacking AMPA Receptor Expression in Dopamine D1 or D2 Receptor Neurons in Animal Behavior,” Neuroscience 398 (2018): 102–112. [DOI] [PubMed] [Google Scholar]
- 65. Kang Y. J., Clement E. M., Park I. H., L. J. Greenfield, Jr. , Smith B. N., and Lee S. H., “Vulnerability of Cholecystokinin‐Expressing GABAergic Interneurons in the Unilateral Intrahippocampal Kainate Mouse Model of Temporal Lobe Epilepsy,” Experimental Neurology 342 (2021): 113724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kim S., Pick J. E., Abera S., et al., “Brain Region‐Specific Effects of cGMP‐Dependent Kinase II Knockout on AMPA Receptor Trafficking and Animal Behavior,” Learning and Memory 23 (2016): 435–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Lee R., Kim G., and Kim S., “Co‐Activation of Selective Nicotinic Acetylcholine Receptor Subtypes Is Required to Reverse Hippocampal Network Dysfunction and Prevent Fear Memory Loss in Alzheimer's Disease,” bioRxiv (2024): 586204, 10.1101/2024.07.08.602576. [DOI] [Google Scholar]
- 68. Castagne V., Moser P., Roux S., and Porsolt R. D., “Rodent models of depression: forced swim and tail suspension behavioral despair tests in rats and mice,” Current Protocols in Neuroscience/Editorial Board Jacqueline N. Crawley 1 (2011): 5–8. [DOI] [PubMed] [Google Scholar]
- 69. Cryan J. F., Mombereau C., and Vassout A., “The Tail Suspension Test as a Model for Assessing Antidepressant Activity: Review of Pharmacological and Genetic Studies in Mice,” Neuroscience and Biobehavioral Reviews 29 (2005): 571–625. [DOI] [PubMed] [Google Scholar]
- 70. Kim S., Shou J., Abera S., and Ziff E. B., “Sucrose Withdrawal Induces Depression and Anxiety‐Like Behavior by Kir2.1 Upregulation in the Nucleus Accumbens,” Neuropharmacology 130 (2018): 10–17. [DOI] [PubMed] [Google Scholar]
- 71. Luo E., Stephens S. B., Chaing S., Munaganuru N., Kauffman A. S., and Breen K. M., “Corticosterone Blocks Ovarian Cyclicity and the LH Surge via Decreased Kisspeptin Neuron Activation in Female Mice,” Endocrinology 157 (2016): 1187–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. McCosh R. B., Kreisman M. J., Tian K., Thomas S. A., and Breen Church K. M., “Norepinephrine Neurons in the Nucleus of the Solitary Tract Suppress Luteinizing Hormone Secretion in Female Mice,” Journal of Neuroscience 44 (2024): e0084242024, 10.1523/JNEUROSCI.0084-24.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Hong I. and Kaang B. K., “The Complexity of Ventral CA1 and Its Multiple Functionalities,” Genes, Brain, and Behavior 21 (2022): e12826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Watarai A., Tao K., Wang M. Y., and Okuyama T., “Distinct Functions of Ventral CA1 and Dorsal CA2 in Social Memory,” Current Opinion in Neurobiology 68 (2021): 29–35. [DOI] [PubMed] [Google Scholar]
- 75. Kim W. B. and Cho J. H., “Encoding of Contextual Fear Memory in Hippocampal‐Amygdala Circuit,” Nature Communications 11 (2020): 1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Yoshida K., Drew M. R., Kono A., Mimura M., Takata N., and Tanaka K. F., “Chronic Social Defeat Stress Impairs Goal‐Directed Behavior Through Dysregulation of Ventral Hippocampal Activity in Male Mice,” Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 46 (2021): 1606–1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Felix‐Ortiz A. C., Burgos‐Robles A., Bhagat N. D., Leppla C. A., and Tye K. M., “Bidirectional Modulation of Anxiety‐Related and Social Behaviors by Amygdala Projections to the Medial Prefrontal Cortex,” Neuroscience 321 (2016): 197–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Wang X. and Zhan Y., “Regulation of Social Recognition Memory in the Hippocampal Circuits,” Frontiers in Neural Circuits 16 (2022): 839931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Tao K., Chung M., Watarai A., Huang Z., Wang M. Y., and Okuyama T., “Disrupted Social Memory Ensembles in the Ventral Hippocampus Underlie Social Amnesia in Autism‐Associated Shank3 Mutant Mice,” Molecular Psychiatry 27 (2022): 2095–2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Phillips M. L., Robinson H. A., and Pozzo‐Miller L., “Ventral Hippocampal Projections to the Medial Prefrontal Cortex Regulate Social Memory,” eLife 8 (2019): e44182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Schumacher A., Vlassov E., and Ito R., “The Ventral Hippocampus, but Not the Dorsal Hippocampus is Critical for Learned Approach‐Avoidance Decision Making,” Hippocampus 26 (2016): 530–542. [DOI] [PubMed] [Google Scholar]
- 82. Su T., Lu Y., Fu C., Geng Y., and Chen Y., “GluN2A Mediates Ketamine‐Induced Rapid Antidepressant‐Like Responses,” Nature Neuroscience 26 (2023): 1751–1761. [DOI] [PubMed] [Google Scholar]
- 83. Davoudian P. A., Shao L. X., and Kwan A. C., “Shared and Distinct Brain Regions Targeted for Immediate Early Gene Expression by Ketamine and Psilocybin,” ACS Chemical Neuroscience 14 (2023): 468–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Yun J., Koike H., Ibi D., et al., “Chronic Restraint Stress Impairs Neurogenesis and Hippocampus‐Dependent Fear Memory in Mice: Possible Involvement of a Brain‐Specific Transcription Factor Npas4,” Journal of Neurochemistry 114 (2010): 1840–1851. [DOI] [PubMed] [Google Scholar]
- 85. Huang P., Li C., Fu T., et al., “Flupirtine Attenuates Chronic Restraint Stress‐Induced Cognitive Deficits and Hippocampal Apoptosis in Male Mice,” Behavioural Brain Research 288 (2015): 1–10. [DOI] [PubMed] [Google Scholar]
- 86. Park M. J., Seo B. A., Lee B., Shin H. S., and Kang M. G., “Stress‐Induced Changes in Social Dominance Are Scaled by AMPA‐Type Glutamate Receptor Phosphorylation in the Medial Prefrontal Cortex,” Scientific Reports 8 (2018): 15008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Kim E. J. and Kim J. J., “Neurocognitive Effects of Stress: A Metaparadigm Perspective,” Molecular Psychiatry 28 (2023): 2750–2763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Tafet G. E. and Nemeroff C. B., “The Links Between Stress and Depression: Psychoneuroendocrinological, Genetic, and Environmental Interactions,” Journal of Neuropsychiatry and Clinical Neurosciences 28 (2016): 77–88. [DOI] [PubMed] [Google Scholar]
- 89. Richter‐Levin G. and Xu L., “How Could Stress Lead to Major Depressive Disorder?,” IBRO Reports 4 (2018): 38–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Mao Y., Xu Y., and Yuan X., “Validity of Chronic Restraint Stress for Modeling Anhedonic‐Like Behavior in Rodents: A Systematic Review and Meta‐Analysis,” Journal of International Medical Research 50 (2022): 3000605221075816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. McEwen B. S., “Physiology and Neurobiology of Stress and Adaptation: Central Role of the Brain,” Physiological Reviews 87 (2007): 873–904. [DOI] [PubMed] [Google Scholar]
- 92. Phillips L. J., McGorry P. D., Garner B., et al., “Stress, the Hippocampus and the Hypothalamic‐Pituitary‐Adrenal axis: Implications for the Development of Psychotic Disorders,” Australian and New Zealand Journal of Psychiatry 40 (2006): 725–741. [DOI] [PubMed] [Google Scholar]
- 93. Raff H., Sharma S. T., and Nieman L. K., “Physiological Basis for the Etiology, Diagnosis, and Treatment of Adrenal Disorders: Cushing's Syndrome, Adrenal Insufficiency, and Congenital Adrenal Hyperplasia,” Comprehensive Physiology 4 (2014): 739–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Raff H., Skelton M. M., and A. W. Cowley, Jr. , “Feedback Control of Vasopressin and Corticotrophin Secretion in Conscious Dogs: Effect of Hypertonic Saline,” Journal of Endocrinology 122 (1989): 41–48. [DOI] [PubMed] [Google Scholar]
- 95. Usa K., Singh R. J., Netzel B. C., Liu Y., Raff H., and Liang M., “Renal Interstitial Corticosterone and 11‐Dehydrocorticosterone in Conscious Rats,” American Journal of Physiology. Renal Physiology 293 (2007): F186–F192. [DOI] [PubMed] [Google Scholar]
- 96. Yu T., Xu H., Wang W., Li S., Chen Z., and Deng H., “Determination of Endogenous Corticosterone in Rodent's Blood, Brain and Hair With LC‐APCI‐MS/MS,” Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences 1002 (2015): 267–276. [DOI] [PubMed] [Google Scholar]
- 97. Burke M. R., Sotiropoulos I., and Waites C. L., “The Multiple Roles of Chronic Stress and Glucocorticoids in Alzheimer's Disease Pathogenesis,” Trends in Neurosciences 47 (2024): 933–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Marin M. T., Cruz F. C., and Planeta C. S., “Chronic Restraint or Variable Stresses Differently Affect the Behavior, Corticosterone Secretion and Body Weight in Rats,” Physiology & Behavior 90 (2007): 29–35. [DOI] [PubMed] [Google Scholar]
- 99. Tan S., Wang Y., Chen K., Long Z., and Zou J., “Ketamine Alleviates Depressive‐Like Behaviors via Down‐Regulating Inflammatory Cytokines Induced by Chronic Restraint Stress in Mice,” Biological & Pharmaceutical Bulletin 40 (2017): 1260–1267. [DOI] [PubMed] [Google Scholar]
- 100. Molnar E., “Long‐Term Potentiation in Cultured Hippocampal Neurons,” Seminars in Cell & Developmental Biology 22 (2011): 506–513. [DOI] [PubMed] [Google Scholar]
- 101. Kim J. W., Suzuki K., Kavalali E. T., and Monteggia L. M., “Bridging Rapid and Sustained Antidepressant Effects of Ketamine,” Trends in Molecular Medicine 29 (2023): 364–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Kavalali E. T. and Monteggia L. M., “Rapid Homeostatic Plasticity and Neuropsychiatric Therapeutics,” Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 48 (2023): 54–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Krystal J. H., Kavalali E. T., and Monteggia L. M., “Ketamine and Rapid Antidepressant Action: New Treatments and Novel Synaptic Signaling Mechanisms,” Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 49 (2024): 41–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Aleksandrova L. R. and Phillips A. G., “Neuroplasticity as a Convergent Mechanism of Ketamine and Classical Psychedelics,” Trends in Pharmacological Sciences 42 (2021): 929–942. [DOI] [PubMed] [Google Scholar]
- 105. Zucker R. S., “Calcium‐ and Activity‐Dependent Synaptic Plasticity,” Current Opinion in Neurobiology 9 (1999): 305–313. [DOI] [PubMed] [Google Scholar]
- 106. Lisek M., Zylinska L., and Boczek T., “Ketamine and Calcium Signaling‐A Crosstalk for Neuronal Physiology and Pathology,” International Journal of Molecular Sciences 21 (2020): 8410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Man H. Y., “GluA2‐Lacking, Calcium‐Permeable AMPA Receptors Inducers of Plasticity?,” Current Opinion in Neurobiology 21 (2011): 291–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Cull‐Candy S. G. and Farrant M., “Ca(2+) ‐Permeable AMPA Receptors and Their Auxiliary Subunits in Synaptic Plasticity and Disease,” Journal of Physiology 599 (2021): 2655–2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Guire E. S., Oh M. C., Soderling T. R., and Derkach V. A., “Recruitment of Calcium‐Permeable AMPA Receptors During Synaptic Potentiation Is Regulated by CaM‐Kinase I,” Journal of Neuroscience 28 (2008): 6000–6009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Zanos P., Moaddel R., Morris P. J., et al., “NMDAR Inhibition‐Independent Antidepressant Actions of Ketamine Metabolites,” Nature 533 (2016): 481–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Franceschelli A., Sens J., Herchick S., Thelen C., and Pitychoutis P. M., “Sex Differences in the Rapid and the Sustained Antidepressant‐Like Effects of Ketamine in Stress‐Naive and “Depressed” Mice Exposed to Chronic Mild Stress,” Neuroscience 290 (2015): 49–60. [DOI] [PubMed] [Google Scholar]
- 112. Carrier N. and Kabbaj M., “Sex Differences in the Antidepressant‐Like Effects of Ketamine,” Neuropharmacology 70 (2013): 27–34. [DOI] [PubMed] [Google Scholar]
- 113. Dossat A. M., Wright K. N., Strong C. E., and Kabbaj M., “Behavioral and Biochemical Sensitivity to Low Doses of Ketamine: Influence of Estrous Cycle in C57BL/6 Mice,” Neuropharmacology 130 (2018): 30–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Saland S. K. and Kabbaj M., “Sex Differences in the Pharmacokinetics of Low‐Dose Ketamine in Plasma and Brain of Male and Female Rats,” Journal of Pharmacology and Experimental Therapeutics 367 (2018): 393–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Chen M., Ma S., Liu H., et al., “Brain Region‐Specific Action of Ketamine as a Rapid Antidepressant,” Science 385 (2024): eado7010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Ma S., Chen M., Jiang Y., et al., “Sustained Antidepressant Effect of Ketamine Through NMDAR Trapping in the LHb,” Nature 622 (2023): 802–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Deyama S. and Duman R. S., “Neurotrophic Mechanisms Underlying the Rapid and Sustained Antidepressant Actions of Ketamine,” Pharmacology, Biochemistry, and Behavior 188 (2020): 172837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Gerhard D. M., Pothula S., Liu R. J., et al., “GABA Interneurons Are the Cellular Trigger for Ketamine's Rapid Antidepressant Actions,” Journal of Clinical Investigation 130 (2020): 1336–1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Yang B., Ren Q., Ma M., Chen Q. X., and Hashimoto K., “Antidepressant Effects of (+)‐MK‐801 and (−)‐MK‐801 in the Social Defeat Stress Model,” International Journal of Neuropsychopharmacology 19 (2016): pyw080, 10.1093/ijnp/pyw080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Wesierska M. J., Duda W., and Dockery C. A., “Low‐Dose Memantine‐Induced Working Memory Improvement in the Allothetic Place Avoidance Alternation Task (APAAT) in Young Adult Male Rats,” Frontiers in Behavioral Neuroscience 7 (2013): 203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Duman R. S., Aghajanian G. K., Sanacora G., and Krystal J. H., “Synaptic Plasticity and Depression: New Insights From Stress and Rapid‐Acting Antidepressants,” Nature Medicine 22 (2016): 238–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Carreno F. R., Donegan J. J., Boley A. M., et al., “Activation of a Ventral Hippocampus‐Medial Prefrontal Cortex Pathway Is Both Necessary and Sufficient for an Antidepressant Response to Ketamine,” Molecular Psychiatry 21 (2016): 1298–1308. [DOI] [PubMed] [Google Scholar]
- 123. Moda‐Sava R. N., Murdock M. H., Parekh P. K., et al., “Sustained Rescue of Prefrontal Circuit Dysfunction by Antidepressant‐Induced Spine Formation,” Science 364 (2019): eaat8078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Davoudian P. A., Shao L.‐X., and Kwan A. C., “Shared and Distinct Brain Regions Targeted for Immediate Early Gene Expression by Ketamine and Psilocybin,” ACS Chemical Neuroscience 14, no. 3 (2022): 468–480, 10.1021/acschemneuro.2c00637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Xu S., Jiang M., Liu X., et al., “Neural Circuits for Social Interactions: From Microcircuits to Input‐Output Circuits,” Frontiers in Neural Circuits 15 (2021): 768294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Campbell S. and Macqueen G., “The Role of the Hippocampus in the Pathophysiology of Major Depression,” Journal of Psychiatry & Neuroscience 29 (2004): 417–426. [PMC free article] [PubMed] [Google Scholar]
- 127. Ghasemi M., Navidhamidi M., Rezaei F., Azizikia A., and Mehranfard N., “Anxiety and Hippocampal Neuronal Activity: Relationship and Potential Mechanisms,” Cognitive, Affective, & Behavioral Neuroscience 22 (2022): 431–449. [DOI] [PubMed] [Google Scholar]
- 128. Jarrard L. E., “On the Role of the Hippocampus in Learning and Memory in the Rat,” Behavioral and Neural Biology 60 (1993): 9–26. [DOI] [PubMed] [Google Scholar]
- 129. Fortin N. J., Agster K. L., and Eichenbaum H. B., “Critical Role of the Hippocampus in Memory for Sequences of Events,” Nature Neuroscience 5 (2002): 458–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Smith M. A., “Hippocampal Vulnerability to Stress and Aging: Possible Role of Neurotrophic Factors,” Behavioural Brain Research 78 (1996): 25–36. [DOI] [PubMed] [Google Scholar]
- 131. Uno H., Tarara R., Else J. G., Suleman M. A., and Sapolsky R. M., “Hippocampal Damage Associated With Prolonged and Fatal Stress in Primates,” Journal of Neuroscience 9 (1989): 1705–1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Egeland M., Zunszain P. A., and Pariante C. M., “Molecular Mechanisms in the Regulation of Adult Neurogenesis During Stress,” Nature Reviews. Neuroscience 16 (2015): 189–200. [DOI] [PubMed] [Google Scholar]
- 133. Kim J. J. and Diamond D. M., “The Stressed Hippocampus, Synaptic Plasticity and Lost Memories,” Nature Reviews. Neuroscience 3 (2002): 453–462. [DOI] [PubMed] [Google Scholar]
- 134. Ryu J. R., Hong C. J., Kim J. Y., Kim E. K., Sun W., and Yu S. W., “Control of Adult Neurogenesis by Programmed Cell Death in the Mammalian Brain,” Molecular Brain 9 (2016): 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Liu Y. Z., Wang Y. X., and Jiang C. L., “Inflammation: The Common Pathway of Stress‐Related Diseases,” Frontiers in Human Neuroscience 11 (2017): 316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. McEwen B. S., Nasca C., and Gray J. D., “Stress Effects on Neuronal Structure: Hippocampus, Amygdala, and Prefrontal Cortex,” Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 41 (2016): 3–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Dale O., Somogyi A. A., Li Y., Sullivan T., and Shavit Y., “Does Intraoperative Ketamine Attenuate Inflammatory Reactivity Following Surgery? A Systematic Review and Meta‐Analysis,” Anesthesia and Analgesia 115 (2012): 934–943. [DOI] [PubMed] [Google Scholar]
- 138. Nikkheslat N., “Targeting Inflammation in Depression: Ketamine as an Anti‐Inflammatory Antidepressant in Psychiatric Emergency,” Brain Behav Immun Health 18 (2021): 100383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Spencer H. F., Berman R. Y., Boese M., et al., “Effects of an Intravenous Ketamine Infusion on Inflammatory Cytokine Levels in Male and Female Sprague‐Dawley Rats,” Journal of Neuroinflammation 19 (2022): 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Wang N., Yu H. Y., Shen X. F., et al., “The Rapid Antidepressant Effect of Ketamine in Rats Is Associated With Down‐Regulation of Pro‐Inflammatory Cytokines in the Hippocampus,” Upsala Journal of Medical Sciences 120 (2015): 241–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Ommati M. M., Mobasheri A., Niknahad H., et al., “Low‐Dose Ketamine Improves Animals' Locomotor Activity and Decreases Brain Oxidative Stress and Inflammation in Ammonia‐Induced Neurotoxicity,” Journal of Biochemical and Molecular Toxicology 37 (2023): e23468. [DOI] [PubMed] [Google Scholar]
- 142. Getachew B., Aubee J. I., Schottenfeld R. S., Csoka A. B., Thompson K. M., and Tizabi Y., “Ketamine Interactions With Gut‐Microbiota in Rats: Relevance to Its Antidepressant and Anti‐Inflammatory Properties,” BMC Microbiology 18 (2018): 222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Zhang J. C., Yao W., Dong C., et al., “Blockade of Interleukin‐6 Receptor in the Periphery Promotes Rapid and Sustained Antidepressant Actions: A Possible Role of Gut‐Microbiota‐Brain Axis,” Translational Psychiatry 7 (2017): e1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Ohta N., Ohashi Y., and Fujino Y., “Ketamine Inhibits Maturation of Bone Marrow‐Derived Dendritic Cells and Priming of the Th1‐Type Immune Response,” Anesthesia and Analgesia 109 (2009): 793–800. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Privacy/ethical restrictions.
