Background

Children with multiple exposures to anesthesia and surgery may have an increased risk of developing cognitive impairment. Sevoflurane, a commonly used anesthetic in children, has been reported to decrease levels of postsynaptic density 95 protein. However, the upstream mechanisms and downstream consequences of the sevoflurane-induced reduction in postsynaptic density 95 protein levels remains largely unknown. We therefore set out to assess whether sevoflurane acts on ubiquitination–proteasome pathway to facilitate postsynaptic density 95 protein degradation.

Methods

Six-day-old wild-type mice received anesthesia with 3% sevoflurane 2 h daily for 3 days starting on postnatal day 6. We determined the effects of the sevoflurane anesthesia on mRNA, protein and ubiquitinated levels of postsynaptic density 95 protein in neurons, and synaptosomes and hippocampus of young mice. Cognitive function in the mice was determined at postnatal day 31 by using a Morris water maze. Proteasome inhibitor MG132 and E3 ligase mouse double mutant 2 homolog inhibitor Nutlin-3 were used for the interaction studies.

Results

The sevoflurane anesthesia decreased protein, but not mRNA, levels of postsynaptic density 95, and reduced ubiquitinated postsynaptic density 95 protein levels in neurons, synaptosomes, and hippocampus of young mice. Both MG132 and Nutlin-3 blocked these sevoflurane-induced effects. Sevoflurane promoted the interaction of mouse double mutant 2 homolog and postsynaptic density 95 protein in neurons. Finally, MG132 and Nutlin-3 ameliorated the sevoflurane-induced cognitive impairment in the mice.

Conclusions

These data suggest that sevoflurane acts on the ubiquitination–proteasome pathway to facilitate postsynaptic density 95 protein degradation, which then decreases postsynaptic density 95 protein levels, leading to cognitive impairment in young mice. These studies would further promote the mechanistic investigation of anesthesia neurotoxicity in the developing brain.

What We Already Know about This Topic
  • In experimental models of neonatal anesthetic neurotoxicity in which cognitive dysfunction occurs, the levels of the scaffolding protein postsynaptic density 95 protein decrease significantly

  • Postsynaptic density 95 protein is a key component of the postsynaptic density and associates with a number of receptor systems, including N-methyl-d-aspartate and α-amino-3-hydroxy-5methyl-4-isoxazolepropionic acid receptor; as such, it plays an essential role in neuroplasticity

What This Article Tells Us That Is New
  • Multiple sevoflurane exposures reduced postsynaptic density 95 protein levels by increasing ubiquitination and proteosomal degradation; inhibition of the ubiquitin–proteosome pathway prevented postsynaptic density 95 protein loss and mitigated cognitive dysfunction

  • The results are consistent with the notion that anesthetic neurotoxicity may result, in part, by reducing postsynaptic density 95 protein levels decreasing neuroplasticity

SEVERAL clinical studies reported that anesthesia and surgery were associated with cognitive impairment in children.1–5  Recent prospective epidemiologic studies show that children do not develop cognitive impairment after single and short duration of anesthesia and surgery.6,7  These findings are consistent with the outcomes reported by Wilder et al.1  that children less than 4 years old who had had multiple (e.g., three times), but not single, anesthesia and surgery might develop a learning disability before age 15. Many preclinical investigations illustrated that anesthetics induce cognitive impairment and neurotoxicity in young rodents8–12  and monkeys.4,5,13–17 

In the mechanistic investigations, Tao et al.12  and Zhang et al.18  reported that anesthesia with 3% sevoflurane 2 h daily for 3 days, starting on postnatal (P) day 6, induced cognitive impairment and reduction in hippocampus levels of postsynaptic density 95 protein (PSD-95), an excitatory postsynaptic marker,19–24  in young mice. However, the upstream mechanism by which sevoflurane decreases PSD-95 levels and the downstream consequences of such reduction in PSD-95 levels remain largely unknown.

PSD-95 regulates adhesion and function of receptors, e.g., affecting N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5methyl-4-isoxazolepropionic acid (AMPA) receptor clustering.25–32  Loss of PSD-95 has been reported to release AMPA receptors from the postsynaptic membrane, allowing for the subsequent removal of PSD-95 from synaptic sites by endocytosis.29,33,34  Given the role of PSD-95 during synaptic development and cognitive function, the studies to assess the effects of anesthesia on PSD-95 levels and the underlying mechanisms in young mice would illustrate new concepts regarding anesthesia neurotoxicity and neurobehavioral deficits in developing brain. Moreover, we could use anesthesia as a tool to further reveal the underlying mechanism of cognitive impairment in young mice.

PSD-95 can be regulated by the ubiquitination–proteasome pathway.35–37  Specifically, PSD-95 is ubiquitinated by the E3 ligase mouse double mutant 2 homolog (MDM2), and the ubiquitinated PSD-95 can then be degraded by the ubiquitin–proteasome system.36,37  Ubiquitination involves the sequential action of three enzymes: ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2), and ubiquitin ligase (E3).38  The key enzyme in the process is the E3, because E3 recognizes and interacts with specific protein substrate, in this case PSD-95. E3 catalyzes and then promotes the transfer of activated ubiquitin to the substrate. Finally, MG132 is an inhibitor of proteasome,39  and Nutlin-3 is an inhibitor of MDM2.40 

Taken together, the objective of the current study is to determine the interaction of sevoflurane and the ubiquitination–proteasome pathway on PSD-95 metabolism. We tested a hypothesis that sevoflurane activates the ubiquitination–proteasome pathway to reduce PSD-95 levels, leading to cognitive impairment in young mice. The effects of sevoflurane on protein and mRNA levels of PSD-95, levels of ubiquitinated PSD-95, and interaction of PSD-95 and MDM2 in vitro (both neurons and synaptosomes) and in vivo were assessed. MG132 and Nutlin-3 were used to further determine the extent to which sevoflurane reduced PSD-95 levels and induced cognitive impairment by activating the ubiquitination–proteasome pathway.

Mice Anesthesia and Treatment

The animal studies were conducted according to the guidelines and regulations of the National Institutes of Health (Bethesda, Maryland). Efforts were made to minimize the number of animals in the studies. The Standing Committee on the Use of Animals in Research and Teaching at Massachusetts General Hospital (Boston, Massachusetts) approved the studies (protocol number 2006N000219). Both female and male mice (C57BL/6J; The Jackson Laboratory, USA) were used in the studies. The mice were randomly assigned into the anesthesia group, control group, or intervention group, including control, sevoflurane plus dimethyl sulfoxide (DMSO), sevoflurane plus MG132, and sevoflurane plus Nutlin-3. The mice received sevoflurane from P6 to P8 and then were decapitated for hippocampus harvest at P8. The studies used a different group of mice in the behavioral studies. These mice received the anesthesia or control conditions from P6 to P8 or on P6, P8, and P10 and then had the Morris water maze (MWM) test from P31 to P37. The mice in the anesthesia group received sevoflurane (3%) plus 60% oxygen (balanced with nitrogen) 2 h daily for 3 consecutive days as performed in our previous studies11,12  or for 3 nonconsecutive days (P6, P8, and P10). The 3% sevoflurane is a clinically relevant concentration, and the anesthesia with 3% sevoflurane 2 h daily for 3 days from P6 to P8 or on P6, P8, and P10 conceptually mimics the multiple exposures of anesthesia in patients. The control conditions were oxygen (60% oxygen and balanced with nitrogen) with an equal rate of flow in a chamber that was similar to the anesthesia chamber.11,12  P6 or P8 mice are equal to postconception day 228 or 262 in humans.41  However, the brain growth spurt reaches the peak in humans around birth and at P7 in rodents. 42,43  We therefore administered the sevoflurane anesthesia at P6, P7, and P8 or P6, P8, and P10 in the current studies. Note that the mice in the control conditions were also separated from the dams and given the control gas (60% oxygen) exposures. The induction flow rate of fresh gas was 2 l/min from the start up to 3 min (for the purpose of induction) and then 1 l/min with the rest of the anesthesia (for maintenance). The concentrations of sevoflurane and oxygen were continuously monitored by using a gas analyzer (Dash 4,000; GE Healthcare, USA) during the anesthesia. The anesthesia chamber temperature was monitored and controlled by a feedback-based system with the DC temperature control system (World Precision Instruments, USA), which controls and automatically adjusts the temperature to keep the rectal temperature of each mouse at 37°C (± 0.5°C) by placing a warming pad under this chamber. The previous studies from another lab10  and from our own lab44  demonstrated that the anesthesia with 3% sevoflurane did not significantly change the levels of pH values, oxygen partial pressure, or carbon dioxide partial pressure as compared to the control group in the young mice. We therefore did not perform the blood gas analysis of the mice in the current studies. In the intervention studies, the mice were treated with MG132 (0.5 mg/kg, dissolved in DMSO at the concentration of 0.3 μg/μl; Sigma-Aldrich, USA)45,46  or Nutlin-3 (10 mg/kg, dissolved in DMSO at the concentration of 0.3 μg/μl; Sigma-Aldrich)47  through intraperitoneal administration 30 min before each sevoflurane anesthesia on P6, P7, and P8. The mice in the control group received 0.1 ml of DMSO solution (15 μl of DMSO dissolved in 1 ml of saline), which is the vehicle of MG132 and Nutlin-3.

Harvest of Brain Tissues

The mice were allowed to totally recover from the anesthesia. Each of the mice was euthanized by decapitation at the end of the sevoflurane anesthesia on P8, and the hippocampus tissues were harvested. The collected hippocampus tissues were utilized in the analysis of Western blot. The harvested hippocampus tissues were homogenized on frost by using immunoprecipitation buffer (10 mM Tris-HCl, pH 7.4, 150 mM NaCl, 2 mM EDTA, 0.5% Nonidet P-40) plus protease inhibitor cocktail from Sigma (catalog No. 11836170001). Finally, the lysates were collected, which were then centrifuged 10 min at the speed of 12,000 rpm.

Harvest of Primary Neurons

Hippocampal neurons of mice were harvested as described in our previous studies with modification.44,48  Specifically, the pregnant dams at gestation day 15 were euthanized with carbon dioxide. The embryos were delivered by Caesarian section, and the brains were dissected in a 100-mm dish of phosphate-buffered saline. Then we specifically placed the hippocampi into another 100-mm dish of phosphate-buffered saline. The neurons were dissociated by using trypsinization and trituration. The dissociated neurons were then resuspended in serum-free B27/neurobasal medium and placed into six-well plates.

Isolation of Synaptosomes

The mice were euthanized by decapitation on P6. Hippocampal tissues from the mice were weighed and homogenized in a grinder using Syn-PER synaptic protein extraction reagent, purchased from Thermo Scientific (catalog No. 87793; Rockford, USA). Immediately before use, protease inhibitor cocktail from Sigma (catalog No. 11836170001) was added to the Syn-PER reagent. Mice hippocampus tissues were homogenized using Syn-PER synaptic protein extraction reagent at a volume ratio of 10 using a 7-ml Dounce tissue grinder with 15 up and down strokes. The homogenate was centrifuged at 2,000g for 10 min to remove cell debris. The resulting supernatant was centrifuged at 15,000g for 20 min. The supernatant formed the cytosolic fraction, and the synaptosome pellet was gently resuspended in Syn-PER synaptic protein extraction reagent. The dissociated synaptosomes were then resuspended in medium 199 HEPES modification (M7528; Sigma) medium and placed into six-well plates.

Treatments for the Neurons or Synaptosomes

The neurons or synaptosomes were treated with 21% O2, 5% CO2, and 4.1% sevoflurane (2 minimum alveolar concentration) for 4 h as described by Dong et al.49  We used an anesthesia machine to deliver 21% O2, 5% CO2, and 4.1% sevoflurane to a sealed plastic box in a 37°C incubator containing six-well plates seeded with the neurons. A gas analyzer (Dash 4000; GE Healthcare) was used to continuously monitor the concentrations of delivered carbon dioxide, oxygen, and sevoflurane as performed in our previous studies.49  In the interaction studies, 50 μM MG13250,51  or 10 μM Nutlin-352,53  was administrated to the neurons 1 h before the sevoflurane treatment. Although different in vitro experiments were performed at different times, the anesthesia and control experiments always occurred concurrently, aiming to reduce bias.

Quantification of Protein

The amount of total proteins was quantified via the kit of bicinchoninic acid protein assay (Pierce, USA) as performed in other studies.49 

Reverse Transcriptase Polymerase Chain Reaction

RNA was isolated from mouse hippocampus. Real-time one-Step reverse transcriptase polymerase chain reaction (PCR) was carried out using the QuantiTect SYBR Green real-time polymerase chain reaction kit (Qiagen, USA). PSD-95 messenger RNA levels were determined and standardized with glyceraldehyde-3-phosphate dehydrogenase as an internal control. Primers of mouse PSD-95 (ID No. PPM04258A) and mouse glyceraldehyde-3-phosphate dehydrogenase (ID No. QT01658692) were purchased from Qiagen. PCR was performed at 50°C for 30 min, 95°C for 15 min, followed by 50 cycles of 94°C for 15 s, 55°C for 30 s and 72°C for 30 s, and finally 95°C for 15 s and 55°C for 15 s.

Western Blot Analysis

The studies utilized PSD-95 antibody (1:1,000, molecular weight of 95 kDa; Cell Signaling, USA) to detect PSD-95 level. An β-actin antibody (1:5,000, molecular weight of 42 kDa; Sigma) was used to detect nontargeted protein β-actin. The quantification of Western-blot was accomplished as described in other studies.49  In brief, we analyzed the signal intensity via the Quantity One image analysis program (Bio-Rad, USA). Two steps were used to quantify the Western blots. At the first step, β-actin was used to standardize protein amounts (e.g., calculating the ratio of PSD-95 in relation to β-actin amount) and limit the differences in the protein amount loaded. At the second step, we expressed the protein levels obtained from the treatment as a percentage in relation to control condition.

Coimmunoprecipitation

The coimmunoprecipitation of PSD-95 and ubiquitin or PSD-95 and MDM2 complexes was performed in neurons. The neurons were solubilized by lysis buffer: 0.5% Nonidet P-40, 1 M Tris, pH 8.0, 4 M NaCl, 0.5 M EDTA and stayed at 4°C overnight. After centrifugation at 12,000 × g for 30 min at 4°C, 800 μg of supernatant was incubated with 1 μg of antibody and 30 μl of prewashed protein A or G-Sepharose (50% slurry) overnight at 4°C. The unbound proteins were washed away, and bound proteins were eluted with SDS sample buffer, separated by SDS-PAGE on a 4 to 15% gradient gel, and transferred to nitrocellulose membrane. The other parts were the same as described under “Western Blot Analysis.”

Morris Water Maze

MWM experiments were performed using the methods described in our previous studies.11,12,18  Briefly, water was filled to a round and steel pool (diameter, 150 cm; height, 60 cm) until the water level reached 1.0 cm over the level of a platform (diametric distance, 10 cm). The pool was located in an isolated room and was surrounded by a black curtain. There were four graphic signals located in the black drape covering the MWM pool. The temperature of the water in the pool was kept at 20°C. The water was made opaque by using titanium dioxide. We tested the P31 mice in the MWM for 7 days (P31 to P37) with four trials daily. A video recording device was used to track the motions of the mice during swimming in the pool. Both escape latency (the time for mouse to reach the platform) in the MWM training from P31 to P37 and platform-crossing times (the counts the mouse moved across the original area of the removed platform) in MWM probe test on P37 were recorded to assess the mouse spatial learning and memory function. Mouse body temperature was maintained by using a heating device described in previous studies.11,12,18  Specifically, we placed each mouse under a heat lamp for 5 min in a holding cage to dry the mouse before returning the mouse to its home cage.

Statistics

The data obtained from biochemistry studies and escape latency of MWM are presented as means ± SD. The numbers of the platform-crossing time of MWM were not normally distributed. Hence, the data are presented as medians with interquartile range. The number of samples was 10 to 11 at every group in behavioral studies, 6 in each group in Western blot and PCR, and 3 in each group in coimmunoprecipitation studies. We estimated a difference in mean escape time from the maze of 30 s, with a SD of 20 s. Using two-sided Student’s t test for two sample mean comparison with 5% type 1 error, a sample size of 10 mice in each group will provide us with more than 80% statistical power to detect such a difference. Similarly, a sample size of six mice in each group will provide the same statistical power for Western blot and PCR. We did not quantify the coimmunoprecipitation studies, but the studies were repeated three times. Two-way analysis of variance (ANOVA) with repeated measurement was used to evaluate the difference of escape latency of the mice in the anesthesia group in relation to the mice in the control group in the MWM test. Post hoc analysis was used to compare the change in escape latency of the anesthesia mice as compared to the control mice for every day during the MWM test, cut-off α was adjusted using the Bonferroni method. We used Mann–Whitney U test to compare the difference in the platform-crossing times of the mice in the anesthesia group and the mice in the control group. Two-way ANOVA without repeated measurement was used to evaluate the interaction of group (control group vs. anesthesia group) and treatment (MG132 or Nutlin-3 vs. DMSO) on the levels of PSD-95. The data in the two-way ANOVA without repeated measurement were found to have normal distribution. A Kruskal–Wallis test was used to detect the platform-crossing times among three groups (MG132 or Nutlin-3 vs. DMSO). The studied employed two-tailed hypothesis, and statistically significant P values were less than 0.05. We used the software of Prism 6 (GraphPad, USA) to evaluate all of the study data.

Sevoflurane Did Not Decrease mRNA Level of PSD-95 in Hippocampi of the Mice

Increasing evidence suggests that PSD-95 plays an important role during synaptic development and cognitive function.54–56  We therefore first assessed the effects of sevoflurane on PSD-95 metabolism by determining both mRNA and protein levels in hippocampi of mice after anesthesia with 3% sevoflurane 2 h daily for 3 days on P6, P7, and P8. Immunoblotting of PSD-95 showed that the sevoflurane anesthesia reduced the visibility of the band representing PSD-95 as compared to the control conditions (fig. 1A). There were no significant differences in β-actin levels between the sevoflurane anesthesia and control condition. Quantification of the Western blot, based on the ratio of PSD-95 to β-actin level, revealed that the sevoflurane anesthesia decreased the protein levels of PSD-95: 48% versus 100%, P = 0.002 (fig. 1B). However, the sevoflurane anesthesia did not significantly change the mRNA levels of PSD-95 (fig. 1C, P = 0.950) in the hippocampi of the mice. These results suggest that sevoflurane may decrease the PSD-95 levels by enhancing its posttranslational degradation.

Fig. 1.

Sevoflurane decreased protein, but not mRNA, levels of postsynaptic density 95 protein (PSD-95) in hippocampi of mice. (A) Anesthesia with 3% sevoflurane 2 h daily for 3 days (postnatal day [P] 6, P7, and P8; right lane) decreased protein levels of PSD-95 as compared to control conditions (left lane) in the hippocampi of mice harvested at P8. (B) Quantification of the Western blot showed that the sevoflurane anesthesia (black bar) decreased the protein levels of PSD-95 as compared to control conditions (white bar; P = 0.002). (C) Anesthesia with 3% sevoflurane 2 h daily for 3 days (P6, P7, and P8; black bar) did not significantly change mRNA levels of PSD-95 as compared to control conditions (white bar) in hippocampi of mice harvested at P8. N = 6 in each group.

Fig. 1.

Sevoflurane decreased protein, but not mRNA, levels of postsynaptic density 95 protein (PSD-95) in hippocampi of mice. (A) Anesthesia with 3% sevoflurane 2 h daily for 3 days (postnatal day [P] 6, P7, and P8; right lane) decreased protein levels of PSD-95 as compared to control conditions (left lane) in the hippocampi of mice harvested at P8. (B) Quantification of the Western blot showed that the sevoflurane anesthesia (black bar) decreased the protein levels of PSD-95 as compared to control conditions (white bar; P = 0.002). (C) Anesthesia with 3% sevoflurane 2 h daily for 3 days (P6, P7, and P8; black bar) did not significantly change mRNA levels of PSD-95 as compared to control conditions (white bar) in hippocampi of mice harvested at P8. N = 6 in each group.

Close modal

MG132 Blocked the Sevoflurane-induced Degradation of PSD-95 Level in Neurons, Synaptosomes, and Hippocampi of Mice

Given the findings that sevoflurane specifically affected the degradation of PSD-95, next we asked whether MG132, a proteasome inhibitor of the ubiquitination–proteasome pathway, would block the sevoflurane-induced reduction of PSD-95 levels in neurons, synaptosomes, and hippocampi of mice. In the neurons pretreated with DMSO, sevoflurane (fig. 2A, lane 2) decreased PSD-95 levels as compared to control conditions (fig. 2A, lane 1). However, in the neurons pretreated with MG132, sevoflurane (fig. 2A, lane 4) did not decrease PSD-95 levels as compared to control conditions (fig. 2A, lane 3). There were no significant differences in β-actin levels among these treatments. Quantification of the Western blot showed that sevoflurane (black bar in fig. 2B) reduced PSD-95 level as compared to the control conditions (white bar in fig. 2B), and MG132 blocked the sevoflurane-induced changes in PSD-95 levels (striped bar vs. gray bar in fig. 2B; F = 11.030, P = 0.003, two-way ANOVA).

Fig. 2.

MG132 attenuated the sevoflurane-induced reduction in postsynaptic density 95 protein (PSD-95) level in hippocampus neurons, synaptosomes of mice, and mice hippocampus tissues. (A) Anesthesia with 4.1% sevoflurane plus dimethyl sulfoxide (DMSO) for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus neurons of mice. However, the treatment with sevoflurane plus MG132 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus MG132 (lane 3). (B) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control conditions plus DMSO (white bar). Sevoflurane plus MG132 (striped bar) did not decrease PSD-95 levels as compared to control conditions plus MG132 (gray bar; F = 11.030, P = 0.003, two-way analysis of variance [ANOVA]). (C) Anesthesia with 4.1% sevoflurane plus DMSO for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus synaptosomes of mice. However, the treatment with sevoflurane plus MG132 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus MG132 (lane 3). (D) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control plus DMSO (white bar). Sevoflurane plus MG132 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus MG132 (gray bar; F = 4.728, P = 0.042, two-way ANOVA). (E) Anesthesia with 3% sevoflurane 2 h daily for 3 days plus DMSO (postnatal day [P] 6, P7, and P8; lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in the hippocampus tissues of mice harvested at P8. However, the sevoflurane anesthesia plus MG132 (lane 4) did not significantly decrease PSD-95 levels as compared to the control conditions plus MG132 (lane 3). (F) Quantification of the Western blot showed that the sevoflurane anesthesia plus DMSO (black bar) decreased PSD-95 levels as compared to the control conditions plus DMSO (white bar). The sevoflurane anesthesia plus MG132 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus MG132 (gray bar; F = 11.030, P = 0.003, two-way ANOVA). N = 6 in each group.

Fig. 2.

MG132 attenuated the sevoflurane-induced reduction in postsynaptic density 95 protein (PSD-95) level in hippocampus neurons, synaptosomes of mice, and mice hippocampus tissues. (A) Anesthesia with 4.1% sevoflurane plus dimethyl sulfoxide (DMSO) for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus neurons of mice. However, the treatment with sevoflurane plus MG132 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus MG132 (lane 3). (B) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control conditions plus DMSO (white bar). Sevoflurane plus MG132 (striped bar) did not decrease PSD-95 levels as compared to control conditions plus MG132 (gray bar; F = 11.030, P = 0.003, two-way analysis of variance [ANOVA]). (C) Anesthesia with 4.1% sevoflurane plus DMSO for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus synaptosomes of mice. However, the treatment with sevoflurane plus MG132 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus MG132 (lane 3). (D) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control plus DMSO (white bar). Sevoflurane plus MG132 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus MG132 (gray bar; F = 4.728, P = 0.042, two-way ANOVA). (E) Anesthesia with 3% sevoflurane 2 h daily for 3 days plus DMSO (postnatal day [P] 6, P7, and P8; lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in the hippocampus tissues of mice harvested at P8. However, the sevoflurane anesthesia plus MG132 (lane 4) did not significantly decrease PSD-95 levels as compared to the control conditions plus MG132 (lane 3). (F) Quantification of the Western blot showed that the sevoflurane anesthesia plus DMSO (black bar) decreased PSD-95 levels as compared to the control conditions plus DMSO (white bar). The sevoflurane anesthesia plus MG132 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus MG132 (gray bar; F = 11.030, P = 0.003, two-way ANOVA). N = 6 in each group.

Close modal

Next, synaptosomes were employed in the studies. The successful extraction of synaptosome was demonstrated by higher levels of PSD-95 and synaptophysin in the synatosomes than those in homogenates and cytosol (Supplemental Digital Content, https://links.lww.com/ALN/B541). The quantitative Western blot showed that MG132 blocked the sevoflurane-induced changes in PSD-95 levels in neurons (fig. 2, A and B; F = 11.030, P = 0.003, two-way ANOVA), synaptosomes (fig. 2, C and D; F = 4.728, P = 0.042, two-way ANOVA), and hippocampi of mice (fig. 2, E and F; F = 11.030, P = 0.003, two-way-ANOVA). MG132 is a general inhibitor of proteasome; thus, these data suggest that sevoflurane may decrease PSD-95 levels through ubiquitination–proteasome pathway.

Nutlin-3 Blocked the Sevoflurane-induced Reduction in PSD-95 Levels in Neurons, Synaptosomes, and Hippocampi of Mice

Given the findings that sevoflurane may decrease PSD-95 levels through ubiquitination–proteasome pathway and Nutlin-3 is an inhibitor of MDM2, the E3 ligase of PSD-95 ubiquitination, next we determined the effects of Nutlin-3 on the sevoflurane-induced reduction in PSD-95 levels in neurons, synaptosomes, and hippocampi of mice. The quantitative Western blot showed that Nutlin-3 blocked the sevoflurane-induced changes in neurons (fig. 3, A and B; F = 5.202, P = 0.034, two-way ANOVA), synaptosomes (fig. 3, C and D; F = 12.680, P = 0.002, two-way ANOVA), and hippocampi of the mice (fig. 3, E and F; F = 9.045, P = 0.007, two-way-ANOVA). Taken together, these data showed that both MG132 and Nutlin-3 blocked the sevoflurane-induced changes in PSD-95 levels and suggest that sevoflurane may decrease PSD-95 levels through promoting the ubiquitination of PSD-95.

Fig. 3.

Nutlin-3 attenuated the sevoflurane-induced reduction in postsynaptic density 95 protein (PSD-95) level in the hippocampus neurons, synaptosomes of mice, and hippocampus tissues of mice. (A) Anesthesia with 4.1% sevoflurane plus dimethyl sulfoxide (DMSO) for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus neurons of mice. The treatment with sevoflurane plus Nutlin-3 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus Nutlin-3 (lane 3). (B) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control conditions plus DMSO (white bar). Sevoflurane plus Nutlin-3 (striped bar) did not decrease PSD-95 levels as compared to control conditions plus Nutlin-3 (gray bar; F = 5.202, P = 0.034, two-way analysis of variance [ANOVA]). (C) Anesthesia with 4.1% sevoflurane plus DMSO for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus synaptosomes of mice. The treatment with sevoflurane plus Nutlin-3 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus Nutlin-3 (lane 3). (D) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control plus DMSO (white bar). Sevoflurane plus Nutlin-3 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus Nutlin-3 (gray bar; F = 12.680, P = 0.002, two-way ANOVA). (E) Anesthesia with 3% sevoflurane 2 h daily for 3 days plus DMSO (postnatal day [P] 6, P7, and P8; lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in the hippocampus tissues of mice harvested at P8. The sevoflurane anesthesia plus Nutlin-3 (lane 4) did not significantly decrease PSD-95 levels as compared to the control conditions plus Nutlin-3 (lane 3). (F) Quantification of the Western blot showed that the sevoflurane anesthesia plus DMSO (black bar) decreased PSD-95 levels as compared to the control conditions plus DMSO (white bar). The sevoflurane anesthesia plus Nutlin-3 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus Nutlin-3 (gray bar; F = 9.045, P = 0.007, two-way ANOVA). N = 6 in each group.

Fig. 3.

Nutlin-3 attenuated the sevoflurane-induced reduction in postsynaptic density 95 protein (PSD-95) level in the hippocampus neurons, synaptosomes of mice, and hippocampus tissues of mice. (A) Anesthesia with 4.1% sevoflurane plus dimethyl sulfoxide (DMSO) for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus neurons of mice. The treatment with sevoflurane plus Nutlin-3 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus Nutlin-3 (lane 3). (B) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control conditions plus DMSO (white bar). Sevoflurane plus Nutlin-3 (striped bar) did not decrease PSD-95 levels as compared to control conditions plus Nutlin-3 (gray bar; F = 5.202, P = 0.034, two-way analysis of variance [ANOVA]). (C) Anesthesia with 4.1% sevoflurane plus DMSO for 4 h (lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in hippocampus synaptosomes of mice. The treatment with sevoflurane plus Nutlin-3 (lane 4) did not reduce PSD-95 levels as compared to the treatment with control conditions plus Nutlin-3 (lane 3). (D) Quantification of the Western blot showed that sevoflurane plus DMSO (black bar) decreased PSD-95 levels as compared to control plus DMSO (white bar). Sevoflurane plus Nutlin-3 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus Nutlin-3 (gray bar; F = 12.680, P = 0.002, two-way ANOVA). (E) Anesthesia with 3% sevoflurane 2 h daily for 3 days plus DMSO (postnatal day [P] 6, P7, and P8; lane 2) decreased PSD-95 levels as compared to control conditions plus DMSO (lane 1) in the hippocampus tissues of mice harvested at P8. The sevoflurane anesthesia plus Nutlin-3 (lane 4) did not significantly decrease PSD-95 levels as compared to the control conditions plus Nutlin-3 (lane 3). (F) Quantification of the Western blot showed that the sevoflurane anesthesia plus DMSO (black bar) decreased PSD-95 levels as compared to the control conditions plus DMSO (white bar). The sevoflurane anesthesia plus Nutlin-3 (striped bar) did not reduce PSD-95 levels as compared to control conditions plus Nutlin-3 (gray bar; F = 9.045, P = 0.007, two-way ANOVA). N = 6 in each group.

Close modal

MG132 Blocked the Sevoflurane-induced Reduction in Ubiquitinated PSD-95 Levels in Cultured Hippocampal Neurons

Ubiquitin, a 76-amino acid protein, is covalently attached to substrate proteins. Once bound with ubiquitin, substrate proteins are often targeted for rapid degradation by the 26S proteasome.38,57  We therefore assessed whether sevoflurane might affect the ubiquitination of PSD-95. Coimmunoprecipitation of ubiquitin and PSD-95 showed that sevoflurane (lane 2) reduced ubiquitinated PSD-95 levels as compared to control conditions (lane 1) in the neurons pretreated with DMSO (top panel in fig. 4). However, in the neurons pretreated with MG132, sevoflurane (lane 4) did not reduce the ubiquitinated PSD-95 levels as compared to control conditions (lane 3 in top panel in fig. 4). The studies were repeated three times. Consistent with this finding, MG132 blocked the sevoflurane-induced reduction in PSD-95 levels in the total cell extracts (bottom panel in fig. 4). These data further suggest that sevoflurane may activate the ubiquitination–proteasome pathway, leading to degradation of PSD-95.

Fig. 4.

Sevoflurane reduced ubiquitinated postsynaptic density 95 protein (PSD-95) level in neurons, and MG132 mitigated the effects. (A) Hippocampal neurons were treated with 4.1% sevoflurane plus dimethyl sulfoxide (DMSO) or MG132 for 4 h. PSD-95 was immunoprecipitated first, and then the immunoprecipitated (IP) samples were immunoblotted (IB) with ubiquitin antibody (upper panel) and PSD95 antibody (lower panel). The treatment with 4.1% sevoflurane plus DMSO (lane 2) decreased the levels of ubiquitinated PSD-95 (upper panel) and total PSD-95 (lower panel) as compared to control conditions plus DMSO (lane 1) in the neurons. However, in the neurons pretreated with MG132, the sevoflurane treatment (lane 4) did not reduce the levels of ubiquitinated PSD-95 (upper panel) and total PSD-95 (lower panel) as compared to control conditions (lane 3). (B) In the total neuron extract, the sevoflurane treatment decreased total PSD-95 levels (lane 2 in upper panel) as compared to the control condition. However, in the neurons pretreated with MG132, the sevoflurane treatment (lane 4 in upper panel) did not reduce total PSD-95 levels as compared to control conditions (lane 3 in upper panel). There was no significant difference in the levels of β-actin among the various treatments (lower panel).

Fig. 4.

Sevoflurane reduced ubiquitinated postsynaptic density 95 protein (PSD-95) level in neurons, and MG132 mitigated the effects. (A) Hippocampal neurons were treated with 4.1% sevoflurane plus dimethyl sulfoxide (DMSO) or MG132 for 4 h. PSD-95 was immunoprecipitated first, and then the immunoprecipitated (IP) samples were immunoblotted (IB) with ubiquitin antibody (upper panel) and PSD95 antibody (lower panel). The treatment with 4.1% sevoflurane plus DMSO (lane 2) decreased the levels of ubiquitinated PSD-95 (upper panel) and total PSD-95 (lower panel) as compared to control conditions plus DMSO (lane 1) in the neurons. However, in the neurons pretreated with MG132, the sevoflurane treatment (lane 4) did not reduce the levels of ubiquitinated PSD-95 (upper panel) and total PSD-95 (lower panel) as compared to control conditions (lane 3). (B) In the total neuron extract, the sevoflurane treatment decreased total PSD-95 levels (lane 2 in upper panel) as compared to the control condition. However, in the neurons pretreated with MG132, the sevoflurane treatment (lane 4 in upper panel) did not reduce total PSD-95 levels as compared to control conditions (lane 3 in upper panel). There was no significant difference in the levels of β-actin among the various treatments (lower panel).

Close modal

Sevoflurane Promoted the Interaction of PSD-95 and MDM2

E3 ligase MDM2 interacts with the ubiquitination of protein, e.g., PSD-95. Thus, we determined the interaction of PSD-95 and MDM2 in neurons. Coimmunoprecipitation of MDM2 and PSD-95 showed that sevoflurane increased the interaction of PSD-95 and MDM2 (top panel in fig. 5) in the neurons. The same sevoflurane treatment also decreased PSD-95 levels in total cell extract (bottom panel in fig. 5). Taken together, these data suggest that sevoflurane may reduce PSD-95 levels not only by activating proteasome but also by promoting the interaction of MDM2 and PSD-95 in the ubiquitination–proteasome pathway.

Fig. 5.

Sevoflurane increased the interaction of postsynaptic density 95 protein (PSD-95) and ubiquitin E3 ligase mouse double mutant 2 homolog (MDM2). (A) Hippocampal neurons were treated with 4.1% sevoflurane for 4 h. The extracts of neuron were first subjected to immunoprecipitation (IP) using antibody against MDM2 and then were immunoblotted (IB) with PSD-95 antibody. The sevoflurane treatment induced a robust increase in the interaction of PSD-95 and MDM2 (lane 2 in upper panel) as compared to control conditions (lane 1 in upper panel). (B) In the total neuron extract, the sevoflurane treatment decreased total PSD-95 levels (lane 2 in upper panel) as compared to the control conditions (lane 1 in upper panel). There were equivalent amounts of β-actin in each condition (lower panel).

Fig. 5.

Sevoflurane increased the interaction of postsynaptic density 95 protein (PSD-95) and ubiquitin E3 ligase mouse double mutant 2 homolog (MDM2). (A) Hippocampal neurons were treated with 4.1% sevoflurane for 4 h. The extracts of neuron were first subjected to immunoprecipitation (IP) using antibody against MDM2 and then were immunoblotted (IB) with PSD-95 antibody. The sevoflurane treatment induced a robust increase in the interaction of PSD-95 and MDM2 (lane 2 in upper panel) as compared to control conditions (lane 1 in upper panel). (B) In the total neuron extract, the sevoflurane treatment decreased total PSD-95 levels (lane 2 in upper panel) as compared to the control conditions (lane 1 in upper panel). There were equivalent amounts of β-actin in each condition (lower panel).

Close modal

MG132 and Nutlin-3 Mitigated the Sevoflurane-induced Cognitive Impairment in Young Mice

Finally, the interaction of sevoflurane with MG132 or Nutlin-3 on cognitive function in young mice was determined. In the mice pretreated with DMSO (the vehicle of MG132 and Nutlin-3), the anesthesia with 3% sevoflurane 2 h daily for 3 days induced cognitive impairment tested in MWM from P31 to P37, as evidenced by the significant interaction between treatment (control conditions and sevoflurane anesthesia) and time (P31 to P37) on escape latency of MWM training test (F = 2.202, P = 0.048, two-way ANOVA with repeated measurement; fig. 6A). A post hoc test (Bonferroni) showed that the mice exposed to sevoflurane had longer escape latency to locate the position of platform in MWM pool as compared to the control mice at P35 (P < 0.05), P36 (P < 0.01), and P37 (P < 0.05; fig. 6A). The mice that received the sevoflurane also demonstrated lower platform-crossing times as compared to the mice that received control conditions (fig. 6B; P = 0.036, Mann–Whitney test).

Fig. 6.

MG132 and Nutlin-3 mitigated the sevoflurane-induced cognitive impairment in young mice. (A) In the mice pretreated with dimethyl sulfoxide (DMSO), two-way analysis of variance (ANOVA) with repeated-measurement analysis showed a significant interaction of treatment (control vs. sevoflurane) and time (days 31 to 37) on escape latency of MWM (F = 2.202, P = 0.048). Mice with the sevoflurane anesthesia plus DMSO had longer escape latency compared with that in the mice with control plus DMSO at days 35, 36, and 37. (B) The sevoflurane anesthesia decreased the platform-crossing times compared with the control condition. (C) Two-way ANOVA with repeated-measurement analysis showed no significant interaction of treatment (control vs. sevoflurane) and time (days 31 to 37) on escape latency of MWM in the mice pretreated with MG132 (F = 0.795, P = 0.576). (D) The sevoflurane anesthesia did not decrease the platform-crossing times as compared to the control conditions in the mice pretreated with MG132. (E) In the mice treated with Nutlin-3, two-way ANOVA with repeated-measurement analysis showed no significant interaction of treatment (control vs. sevoflurane) and time (days 31 to 37) on escape latency of the Morris water maze in the mice pretreated with Nutlin-3 (F = 1.238, P = 0.292). (F) The sevoflurane anesthesia did not decrease the platform-crossing times as compared to the control conditions in the mice pretreated with Nutlin-3. N = 10 to 11 in each group.

Fig. 6.

MG132 and Nutlin-3 mitigated the sevoflurane-induced cognitive impairment in young mice. (A) In the mice pretreated with dimethyl sulfoxide (DMSO), two-way analysis of variance (ANOVA) with repeated-measurement analysis showed a significant interaction of treatment (control vs. sevoflurane) and time (days 31 to 37) on escape latency of MWM (F = 2.202, P = 0.048). Mice with the sevoflurane anesthesia plus DMSO had longer escape latency compared with that in the mice with control plus DMSO at days 35, 36, and 37. (B) The sevoflurane anesthesia decreased the platform-crossing times compared with the control condition. (C) Two-way ANOVA with repeated-measurement analysis showed no significant interaction of treatment (control vs. sevoflurane) and time (days 31 to 37) on escape latency of MWM in the mice pretreated with MG132 (F = 0.795, P = 0.576). (D) The sevoflurane anesthesia did not decrease the platform-crossing times as compared to the control conditions in the mice pretreated with MG132. (E) In the mice treated with Nutlin-3, two-way ANOVA with repeated-measurement analysis showed no significant interaction of treatment (control vs. sevoflurane) and time (days 31 to 37) on escape latency of the Morris water maze in the mice pretreated with Nutlin-3 (F = 1.238, P = 0.292). (F) The sevoflurane anesthesia did not decrease the platform-crossing times as compared to the control conditions in the mice pretreated with Nutlin-3. N = 10 to 11 in each group.

Close modal

However, in the mice with the pretreatment of MG132, the sevoflurane anesthesia did not induce cognitive impairment tested in the MWM as evidenced in the fact that there was no significant interaction between treatment (control conditions and sevoflurane anesthesia) versus time (P31 to P37; F = 0.795, P = 0.576, two-way ANOVA with repeated measurement; fig. 6C). Consistent with this finding, the sevoflurane anesthesia did not significantly decrease the platform-crossing times as compared to control conditions in the mice pretreated with MG132 (fig. 6D; P = 0.912, Mann–Whitney test).

Similarly, in the mice with the pretreatment of Nutlin-3, the sevoflurane anesthesia did not induce cognitive impairment tested in MWM as evidenced by no significant interaction (F = 1.238, P = 0.292, two-way ANOVA with repeated measurement; fig. 6E) between treatment (control conditions and sevoflurane anesthesia) and time (P31 to P37) in the training of MWM test and by no difference in platform-crossing times of the probe test (P = 0.350, Mann–Whitney test; fig. 6F).

In addition, there was no significant interaction of treatment (DMSO or MG132 or Nutlin-3) and time (P31 to P37) on escape latency of MWM training test (F = 0.570, P = 0.864, two-way ANOVA; fig. 7A). There were no significant differences of platform-crossing times among these three groups. (fig. 7B; P = 0.247, Kruskal–Wallis test). Taken together, these data indicated that there were no significant differences of the effects of DMSO, MG132, and Nutlin-3 alone on cognitive function in young mice (fig. 7). Collectively, these findings suggest that both MG132 and Nutlin-3 can ameliorate the sevoflurane-induced cognitive impairment in young mice.

Fig. 7.

There were no differences of the effects of dimethyl sulfoxide (DMSO), MG132, and Nutlin-3 on cognitive function in young mice. We compared the effects of DMSO, MG132, or Nutlin-3 on cognitive function in young mice. Two-way analysis of variance with repeated measurement showed no significant interaction of treatment (DMSO, MG132, and Nutlin-3) and time (postnatal days 31 to 37) on escape latency (F = 0.570, P = 0.864). (A) There were no significant differences of platform-crossing times among these three groups. (B) Kruskal–Wallis test (P = 0.247).

Fig. 7.

There were no differences of the effects of dimethyl sulfoxide (DMSO), MG132, and Nutlin-3 on cognitive function in young mice. We compared the effects of DMSO, MG132, or Nutlin-3 on cognitive function in young mice. Two-way analysis of variance with repeated measurement showed no significant interaction of treatment (DMSO, MG132, and Nutlin-3) and time (postnatal days 31 to 37) on escape latency (F = 0.570, P = 0.864). (A) There were no significant differences of platform-crossing times among these three groups. (B) Kruskal–Wallis test (P = 0.247).

Close modal

Nonconsecutive Anesthesia Exposures with Sevoflurane-induced Cognitive Impairment in Young Mice

Finally, we asked whether the sevoflurane-induced cognitive impairment was dependent on the 3-day consecutive anesthesia exposures. The effects of sevoflurane anesthesia on cognitive function in young mice were determined using different anesthesia methods. Specifically, the young mice received 3% sevoflurane 2 h daily for 3 nonconsecutive days (P6, P8, and P10) instead of 3 consecutive days (P6, P7, and P8). We found that the nonconsecutive 3-day sevoflurane anesthesia also induced cognitive impairment in the young mice, as evidenced by a significant interaction of treatment (control and sevoflurane) and time (P31 to P37) on escape latency of MWM (fig. 8A) and significant difference of platform-crossing times between control and sevoflurane (fig. 8B). These data suggest the sevoflurane-induced cognitive impairment in young mice is not dependent on the 3 consecutive day exposures of anesthesia.

Fig. 8.

Nonconsecutive anesthesia exposures with sevoflurane induced cognitive impairment in young mice. The mice received anesthesia with 3% sevoflurane 2 h daily for nonconsecutive days (postnatal day [P] 6, P8, and P10) and were tested from P31 to P37 in the Morris water maze (MWM). (A) Two-way analysis of variance (ANOVA) with repeated-measurement analysis showed a significant interaction of treatment (control vs. sevoflurane) and time (P31 to P37) on escape latency of MWM. The mice that received the sevoflurane anesthesia had longer escape latency as compared with that in the mice in the control group at P34, P35, P36, and P37. (B) The sevoflurane anesthesia decreased the platform-crossing times as compared with the control conditions in the mice at P37. N = 10 in each group. DMSO = dimethyl sulfoxide.

Fig. 8.

Nonconsecutive anesthesia exposures with sevoflurane induced cognitive impairment in young mice. The mice received anesthesia with 3% sevoflurane 2 h daily for nonconsecutive days (postnatal day [P] 6, P8, and P10) and were tested from P31 to P37 in the Morris water maze (MWM). (A) Two-way analysis of variance (ANOVA) with repeated-measurement analysis showed a significant interaction of treatment (control vs. sevoflurane) and time (P31 to P37) on escape latency of MWM. The mice that received the sevoflurane anesthesia had longer escape latency as compared with that in the mice in the control group at P34, P35, P36, and P37. (B) The sevoflurane anesthesia decreased the platform-crossing times as compared with the control conditions in the mice at P37. N = 10 in each group. DMSO = dimethyl sulfoxide.

Close modal

In this mechanistic investigation of anesthesia-induced neurotoxicity and neurobehavioral deficits in developing brain, we demonstrated that sevoflurane activated the ubiquitination–proteasome pathway by stimulating ubiquitination–proteasome pathway and promoting the interaction of PSD-95 and MDM2. This interaction facilitates the ubiquitination of PSD-95, leading to a reduction in protein levels of PSD-95. MG132 (a proteasome inhibitor of the ubiquitination–proteasome pathway) and Nutlin-3 (an inhibitor of MDM2) blocked the sevoflurane-induced PSD-95 metabolism and ameliorated the sevoflurane-induced cognitive impairment in young mice.

The sevoflurane anesthesia decreased the protein level, but not the mRNA level, of PSD-95 (fig. 1). These results suggest that sevoflurane may not regulate the generation of PSD-95 but facilitate PSD-95 degradation. Then the sevoflurane-induced changes in PSD-95 levels were inhibited by MG132 (fig. 2) and Nutlin-3 (fig. 3). These data suggest that sevoflurane decreases PSD-95 levels by activating the ubiquitination–proteasome pathway and specifically through acting on MDM2 and proteasome. The future studies should assess the underlying mechanism by which sevoflurane acts on MDM2 by investigating whether sevoflurane regulates the trafficking of MDM2. Note that the interactions of sevoflurane and MG132 or Nutlin-3 were examined in both neurons and synaptosomes, which would help us to determine age-dependent changes of sevoflurane on the interaction of PSD-95 and MDM2 or proteasome by using synaptosomes obtained from mice with different ages.

There was an interaction of the sevoflurane and MG132 on the levels of ubiquitinated PSD-95 (fig. 4). The original hypothesis was that sevoflurane would increase the ubiquitinated PSD-95 levels by promoting the ubiquitination of PSD-95. Interestingly, we found that sevoflurane decreased the ubiquitinated PSD-95 levels in the hippocampus neurons. These data suggest that sevoflurane may have stronger effects on stimulating proteasome-associated degradation of ubiquitinated PSD-95 than on promoting MDM2-regulated generation of ubiquitinated PSD-95 in a time-dependent manner. This hypothesis was supported by the observation that proteasome inhibitor MG132 blocked the sevoflurane-induced changes in ubiquitinated PSD-95 levels (fig. 4). Moreover, these findings are consistent with the results obtained from the previous studies that treatment with NMDA induces time-dependent changes in ubiquitinated PSD-95 levels.36  Specifically, there was a reduction and then an increase in ubiquitinated PSD-95 levels at 5 and 10 min after the treatment of NMDA, respectively, in hippocampus neurons.36  Future studies should test whether sevoflurane increases ubiquitinated PSD-95 levels at shorter time and decreases its levels at longer time after the treatment.

Moreover, sevoflurane promoted the interaction of PSD-95 and MDM2 (top panel in fig. 5) in neurons and decreased PSD-95 levels in total cell extract (bottom panel in fig. 5). These data suggest that sevoflurane can promote the binding and interaction of MDM2 with PSD-95, leading to ubiquitination of PSD-95. These findings further suggest that sevoflurane may act on both MDM2 and other parts in the ubiquitination–proteasome pathway.

Finally, both MG132 and Nutlin-3 ameliorated the sevoflurane-induced cognitive impairment in young mice (fig. 6), as well as the sevoflurane-induced changes in PSD-95 levels. Taken together, these findings suggest that the sevoflurane-induced reduction in PSD-95 may lead to cognitive impairment in young mice.

PSD-95 is a major scaffolding protein in excitatory postsynaptic density25  that enhances maturation of the presynaptic terminal, increases the number and size of dendritic spines,58  and contributes to synaptic organization.59  Experiments demonstrate that PSD-95 can orchestrate synaptic development and play an important role in synapse stabilization and plasticity.60  Our previous studies reported that sevoflurane reduced the levels of PSD-95 in vitro and in vivo.12,18  The findings from current studies reveal that sevoflurane may decrease PSD-95 levels by activating the ubiquitination–proteasome pathway.

Earlier studies have demonstrated that PSD-95 can be degraded by the ubiquitination–proteasome pathway.36,37  Specifically, Colledge et al.36  showed that treatment with NMDA induced a rapid ubiquitination of PSD-95, which reached peak levels at 10 min after the treatment, leading to reduction in PSD-95 levels in the neurons. Tsai et al.37  reported that activation of the activity-dependent transcription factor myocyte enhancer factor 2 in the nucleus promoted the ubiquitination of PSD-95, which consequently decreased the levels of PSD-95 and synapse in hippocampus neurons.

Consistently, sevoflurane decreased PSD-95 levels by promoting PSD-95 ubiquitination in neurons, synaptosomes, and brain tissues of mice. However, the current studies are different from the previous studies in revealing: (1) the ubiquitination–proteasome pathway is one of the underlying mechanisms of the cognitive impairment in young mice; (2) sevoflurane induces cognitive impairment in young mice through stimulation of the ubiquitination–proteasome pathway and promoting interaction of MDM2 and PSD-95; (3) the sevoflurane-induced reduction in PSD-95 levels contributes, at least partially, to the sevoflurane-induced cognitive impairment; and (4) MG132 and Nutlin-3 may prevent and treat the sevoflurane-induced neurotoxicity and neurobehavioral deficits in young mice. Collectively, these findings could help us to further understand cognitive impairment and anesthesia neurotoxicity in the developing brain.

It is possible that different mechanisms may contribute to the in vitro and in vivo findings. However, in the present studies, the in vitro and in vivo findings likely share, at least partially, the same mechanism: the ubiquitination-proteasome degradation pathway. First, the effects of MG132 and Nutlin-3 on attenuating the sevoflurane-induced reduction in PSD-95 levels were similar among neurons and synaptosomes (in vitro studies) and brain tissues (in vivo studies). Second, the control conditions in the in vitro studies, which did not affect PSD-95 levels, had the same direct physical interference of airflow rate as compared to the anesthesia condition.

The study has several limitations. First, we only assessed the effects of sevoflurane on the levels of PSD-95, but not the number of synapse or dendritic spine, in hippocampus of young mice. This is because previous studies have reported that reductions in PSD-95 levels are associated with reductions in synapse numbers.37  Future studies should determine whether sevoflurane can reduce the number of synapses and dendritic spines in the hippocampi of mice. Second, we only performed the interaction of PSD-95 and MDM2 studies in neurons, but not in synaptosomes or brain tissues, due to technical limitations of immunoprecipitation experiments in synaptosomes. Third, the anesthesia used in the current studies was 3% sevoflurane 2 h daily for 3 days on P6, P7, and P8 in the young mice. This is because our previous studies showed that anesthesia with 3% sevoflurane 2 h daily for 3 days (P6, P7, and P8), but not for 1 day, in young mice was able to induce cognitive impairment at P31 to P37 in the mice.11,12  This anesthesia may not be exactly relevant to pediatric anesthesia practice. In addition, the anesthetic exposure on 3 consecutive days could represent a severe form of toxicity. However, this sevoflurane anesthesia did not increase mortality or induce abnormal appearance in the mice, e.g., ruffled fur guarding, loss of appetite, reluctance to move, mutilation, restlessness, recumbence, and vocalization, as compared to the control group at P31. Moreover, anesthesia with 9% desflurane 2 h daily for 3 days (P6, P7, and P8) in the young mice did not induce cognitive impairment tested from P31 to P37 in the mice.11  These data suggest that the 3 consecutive days of anesthesia may not cause a severe form of toxicity. Finally, the findings that anesthesia with 3% sevoflurane 2 h daily for nonconsecutive days (P6, P8, and P10) in young mice still induced cognitive impairment (fig. 8) suggest that the sevoflurane-induced cognitive impairment in young mice may not be dependent on the 3 consecutive day anesthesia exposures. Fourth, we did not specifically compare the effects of the anesthesia in male and female mice in the present studies. This is because our previous studies demonstrated that sevoflurane was able to induce cognitive impairment in both male and female young mice, and the objective of the present study was to use the established system to determine the upstream ubiquitination–proteasome mechanism by which the sevoflurane anesthesia decreases the level of PSD-95 and the downstream cognitive consequence of such reduction in PSD-95 levels. Finally, there were dam’s rearing behaviors in the young mice because the mice were separated from mothers.61  However, the control mice also had the same time of separation from the mother. Moreover, the separation only occurred 2 h daily for 3 days.

In conclusion, we found that sevoflurane reduced levels of PSD-95 in vitro (neurons and synaptosomes) and in vivo (brain tissues of young mice) by activating the ubiquitination–proteasome pathway. Specifically, sevoflurane stimulated the proteasome and promoted interaction of MDM2 and PSD-95, which caused ubiquitination of PSD-95. The ubiquitinated PSD-95 then was degraded by proteasome, leading to reduction in PSD-95 levels. MG132, a proteasome inhibitor of the ubiquitination–proteasome pathway and Nutlin-3, an inhibitor of MDM2, mitigated these effects. These results illustrated the potential underlying mechanisms of neurotoxicity and neurobehavioral deficits induced by anesthesia or other external stressors in developing brain. These findings should promote further investigation of cognitive impairment in young mice, which will ultimately improve the safety of anesthesia care and lead to better outcomes of anesthesia and surgery in children.

Sevoflurane was generously provided by the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School (Boston, Massachusetts).

Supported by National Institutes of Health (Bethesda, Maryland) grant Nos. HD086977, GM088801, and AG041274 (to Dr. Xie) and in part by the Chinese National Natural Science Foundation (Beijing, China) grant Nos. 81102513 (to Dr. Lu) and 81373492 (to Dr. Yu).

The authors declare no competing interests.

1.
Wilder
RT
,
Flick
RP
,
Sprung
J
,
Katusic
SK
,
Barbaresi
WJ
,
Mickelson
C
,
Gleich
SJ
,
Schroeder
DR
,
Weaver
AL
,
Warner
DO
:
Early exposure to anesthesia and learning disabilities in a population-based birth cohort.
Anesthesiology
2009
;
110
:
796
804
2.
Flick
RP
,
Katusic
SK
,
Colligan
RC
,
Wilder
RT
,
Voigt
RG
,
Olson
MD
,
Sprung
J
,
Weaver
AL
,
Schroeder
DR
,
Warner
DO
:
Cognitive and behavioral outcomes after early exposure to anesthesia and surgery.
Pediatrics
2011
;
128
:
e1053
61
3.
Backeljauw
B
,
Holland
SK
,
Altaye
M
,
Loepke
AW
:
Cognition and brain structure following early childhood surgery with anesthesia.
Pediatrics
2015
;
136
:
e1
12
4.
Sun
L
:
Early childhood general anaesthesia exposure and neurocognitive development.
Br J Anaesth
2010
;
105
(
suppl 1
):
i61
8
5.
Vutskits
L
,
Xie
Z
:
Lasting impact of general anaesthesia on the brain: Mechanisms and relevance.
Nat Rev Neurosci
2016
;
17
:
705
17
6.
Davidson
AJ
,
Disma
N
,
de Graaff
JC
,
Withington
DE
,
Dorris
L
,
Bell
G
,
Stargatt
R
,
Bellinger
DC
,
Schuster
T
,
Arnup
SJ
,
Hardy
P
,
Hunt
RW
,
Takagi
MJ
,
Giribaldi
G
,
Hartmann
PL
,
Salvo
I
,
Morton
NS
,
von Ungern Sternberg
BS
,
Locatelli
BG
,
Wilton
N
,
Lynn
A
,
Thomas
JJ
,
Polaner
D
,
Bagshaw
O
,
Szmuk
P
,
Absalom
AR
,
Frawley
G
,
Berde
C
,
Ormond
GD
,
Marmor
J
,
McCann
ME
;
GAS Consortium
:
Neurodevelopmental outcome at 2 years of age after general anaesthesia and awake-regional anaesthesia in infancy (GAS): An international multicentre, randomised controlled trial.
Lancet
2016
;
387
:
239
50
7.
Sun
LS
,
Li
G
,
Miller
TL
,
Salorio
C
,
Byrne
MW
,
Bellinger
DC
,
Ing
C
,
Park
R
,
Radcliffe
J
,
Hays
SR
,
DiMaggio
CJ
,
Cooper
TJ
,
Rauh
V
,
Maxwell
LG
,
Youn
A
,
McGowan
FX
:
Association between a single general anesthesia exposure before age 36 months and neurocognitive outcomes in later childhood.
JAMA
2016
;
315
:
2312
20
8.
Jevtovic-Todorovic
V
,
Hartman
RE
,
Izumi
Y
,
Benshoff
ND
,
Dikranian
K
,
Zorumski
CF
,
Olney
JW
,
Wozniak
DF
:
Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits.
J Neurosci
2003
;
23
:
876
82
9.
Stratmann
G
,
Sall
JW
,
May
LD
,
Bell
JS
,
Magnusson
KR
,
Rau
V
,
Visrodia
KH
,
Alvi
RS
,
Ku
B
,
Lee
MT
,
Dai
R
:
Isoflurane differentially affects neurogenesis and long-term neurocognitive function in 60-day-old and 7-day-old rats.
Anesthesiology
2009
;
110
:
834
48
10.
Satomoto
M
,
Satoh
Y
,
Terui
K
,
Miyao
H
,
Takishima
K
,
Ito
M
,
Imaki
J
:
Neonatal exposure to sevoflurane induces abnormal social behaviors and deficits in fear conditioning in mice.
Anesthesiology
2009
;
110
:
628
37
11.
Shen
X
,
Dong
Y
,
Xu
Z
,
Wang
H
,
Miao
C
,
Soriano
SG
,
Sun
D
,
Baxter
MG
,
Zhang
Y
,
Xie
Z
:
Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment.
Anesthesiology
2013
;
118
:
502
15
12.
Tao
G
,
Zhang
J
,
Zhang
L
,
Dong
Y
,
Yu
B
,
Crosby
G
,
Culley
DJ
,
Zhang
Y
,
Xie
Z
:
Sevoflurane induces tau phosphorylation and glycogen synthase kinase 3β activation in young mice.
Anesthesiology
2014
;
121
:
510
27
13.
Brambrink
AM
,
Evers
AS
,
Avidan
MS
,
Farber
NB
,
Smith
DJ
,
Zhang
X
,
Dissen
GA
,
Creeley
CE
,
Olney
JW
:
Isoflurane-induced neuroapoptosis in the neonatal rhesus macaque brain.
Anesthesiology
2010
;
112
:
834
41
14.
Brambrink
AM
,
Back
SA
,
Riddle
A
,
Gong
X
,
Moravec
MD
,
Dissen
GA
,
Creeley
CE
,
Dikranian
KT
,
Olney
JW
:
Isoflurane-induced apoptosis of oligodendrocytes in the neonatal primate brain.
Ann Neurol
2012
;
72
:
525
35
15.
Liu
F
,
Rainosek
SW
,
Frisch-Daiello
JL
,
Patterson
TA
,
Paule
MG
,
Slikker
W
Jr
,
Wang
C
,
Han
X
:
Potential adverse effects of prolonged sevoflurane exposure on developing monkey brain: From abnormal lipid metabolism to neuronal damage.
Toxicol Sci
2015
;
147
:
562
72
16.
Zou
X
,
Liu
F
,
Zhang
X
,
Patterson
TA
,
Callicott
R
,
Liu
S
,
Hanig
JP
,
Paule
MG
,
Slikker
W
Jr
,
Wang
C
:
Inhalation anesthetic-induced neuronal damage in the developing rhesus monkey.
Neurotoxicol Teratol
2011
;
33
:
592
7
17.
Slikker
W
Jr
,
Zou
X
,
Hotchkiss
CE
,
Divine
RL
,
Sadovova
N
,
Twaddle
NC
,
Doerge
DR
,
Scallet
AC
,
Patterson
TA
,
Hanig
JP
,
Paule
MG
,
Wang
C
:
Ketamine-induced neuronal cell death in the perinatal rhesus monkey.
Toxicol Sci
2007
;
98
:
145
58
18.
Zhang
J
,
Dong
Y
,
Zhou
C
,
Zhang
Y
,
Xie
Z
:
Anesthetic sevoflurane reduces levels of hippocalcin and postsynaptic density protein 95.
Mol Neurobiol
2015
;
51
:
853
63
19.
Masliah
E
,
Mallory
M
,
Hansen
L
,
DeTeresa
R
,
Alford
M
,
Terry
R
:
Synaptic and neuritic alterations during the progression of Alzheimer’s disease.
Neurosci Lett
1994
;
174
:
67
72
20.
Love
S
,
Siew
LK
,
Dawbarn
D
,
Wilcock
GK
,
Ben-Shlomo
Y
,
Allen
SJ
:
Premorbid effects of APOE on synaptic proteins in human temporal neocortex.
Neurobiol Aging
2006
;
27
:
797
803
21.
Sultana
R
,
Banks
WA
,
Butterfield
DA
:
Decreased levels of PSD95 and two associated proteins and increased levels of BCl2 and caspase 3 in hippocampus from subjects with amnestic mild cognitive impairment: Insights into their potential roles for loss of synapses and memory, accumulation of Aβ, and neurodegeneration in a prodromal stage of Alzheimer’s disease.
J Neurosci Res
2010
;
88
:
469
77
22.
Takeuchi
M
,
Hata
Y
,
Hirao
K
,
Toyoda
A
,
Irie
M
,
Takai
Y
:
SAPAPs. A family of PSD-95/SAP90-associated proteins localized at postsynaptic density.
J Biol Chem
1997
;
272
:
11943
51
23.
Liu
Q
,
Trotter
J
,
Zhang
J
,
Peters
MM
,
Cheng
H
,
Bao
J
,
Han
X
,
Weeber
EJ
,
Bu
G
:
Neuronal LRP1 knockout in adult mice leads to impaired brain lipid metabolism and progressive, age-dependent synapse loss and neurodegeneration.
J Neurosci
2010
;
30
:
17068
78
24.
Yuki
D
,
Sugiura
Y
,
Zaima
N
,
Akatsu
H
,
Takei
S
,
Yao
I
,
Maesako
M
,
Kinoshita
A
,
Yamamoto
T
,
Kon
R
,
Sugiyama
K
,
Setou
M
:
DHA-PC and PSD-95 decrease after loss of synaptophysin and before neuronal loss in patients with Alzheimer’s disease.
Sci Rep
2014
;
4
:
7130
25.
Chen
X
,
Nelson
CD
,
Li
X
,
Winters
CA
,
Azzam
R
,
Sousa
AA
,
Leapman
RD
,
Gainer
H
,
Sheng
M
,
Reese
TS
:
PSD-95 is required to sustain the molecular organization of the postsynaptic density.
J Neurosci
2011
;
31
:
6329
38
26.
Xu
W
:
PSD-95-like membrane associated guanylate kinases (PSD-MAGUKs) and synaptic plasticity.
Curr Opin Neurobiol
2011
;
21
:
306
12
27.
Swulius
MT
,
Kubota
Y
,
Forest
A
,
Waxham
MN
:
Structure and composition of the postsynaptic density during development.
J Comp Neurol
2010
;
518
:
4243
60
28.
Fan
J
,
Vasuta
OC
,
Zhang
LY
,
Wang
L
,
George
A
,
Raymond
LA
:
N-Methyl-d-aspartate receptor subunit- and neuronal-type dependence of excitotoxic signaling through post-synaptic density 95.
J Neurochem
2010
;
115
:
1045
56
29.
Bhattacharyya
S
,
Biou
V
,
Xu
W
,
Schlüter
O
,
Malenka
RC
:
A critical role for PSD-95/AKAP interactions in endocytosis of synaptic AMPA receptors.
Nat Neurosci
2009
;
12
:
172
81
30.
Delint-Ramírez
I
,
Salcedo-Tello
P
,
Bermudez-Rattoni
F
:
Spatial memory formation induces recruitment of NMDA receptor and PSD-95 to synaptic lipid rafts.
J Neurochem
2008
;
106
:
1658
68
31.
Sornarajah
L
,
Vasuta
OC
,
Zhang
L
,
Sutton
C
,
Li
B
,
El-Husseini
A
,
Raymond
LA
:
NMDA receptor desensitization regulated by direct binding to PDZ1-2 domains of PSD-95.
J Neurophysiol
2008
;
99
:
3052
62
32.
Cousins
SL
,
Papadakis
M
,
Rutter
AR
,
Stephenson
FA
:
Differential interaction of NMDA receptor subtypes with the post-synaptic density-95 family of membrane associated guanylate kinase proteins.
J Neurochem
2008
;
104
:
903
13
33.
Béïque
JC
,
Lin
DT
,
Kang
MG
,
Aizawa
H
,
Takamiya
K
,
Huganir
RL
:
Synapse-specific regulation of AMPA receptor function by PSD-95.
Proc Natl Acad Sci USA
2006
;
103
:
19535
40
34.
Opazo
P
,
Sainlos
M
,
Choquet
D
:
Regulation of AMPA receptor surface diffusion by PSD-95 slots.
Curr Opin Neurobiol
2012
;
22
:
453
60
35.
Ehlers
MD
:
Activity level controls postsynaptic composition and signaling via the ubiquitin–proteasome system.
Nat Neurosci
2003
;
6
:
231
42
36.
Colledge
M
,
Snyder
EM
,
Crozier
RA
,
Soderling
JA
,
Jin
Y
,
Langeberg
LK
,
Lu
H
,
Bear
MF
,
Scott
JD
:
Ubiquitination regulates PSD-95 degradation and AMPA receptor surface expression.
Neuron
2003
;
40
:
595
607
37.
Tsai
NP
,
Wilkerson
JR
,
Guo
W
,
Maksimova
MA
,
DeMartino
GN
,
Cowan
CW
,
Huber
KM
:
Multiple autism-linked genes mediate synapse elimination via proteasomal degradation of a synaptic scaffold PSD-95.
Cell
2012
;
151
:
1581
94
38.
Joazeiro
CA
,
Hunter
T
:
Biochemistry: Ubiquitination–more than two to tango.
Science
2000
;
289
:
2061
2
39.
Lee
DH
,
Goldberg
AL
:
Proteasome inhibitors: Valuable new tools for cell biologists.
Trends Cell Biol
1998
;
8
:
397
403
40.
Secchiero
P
,
Corallini
F
,
Gonelli
A
,
Dell’Eva
R
,
Vitale
M
,
Capitani
S
,
Albini
A
,
Zauli
G
:
Antiangiogenic activity of the MDM2 antagonist Nutlin-3.
Circ Res
2007
;
100
:
61
9
41.
Clancy
B
,
Kersh
B
,
Hyde
J
,
Darlington
RB
,
Anand
KJ
,
Finlay
BL
:
Web-based method for translating neurodevelopment from laboratory species to humans.
Neuroinformatics
2007
;
5
:
79
94
42.
Dobbing
J
,
Sands
J
:
Comparative aspects of the brain growth spurt.
Early Hum Dev
1979
;
3
:
79
83
43.
Semple
BD
,
Blomgren
K
,
Gimlin
K
,
Ferriero
DM
,
Noble-Haeusslein
LJ
:
Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species.
Prog Neurobiol
2013
;
106–7
:
1
16
44.
Lu
Y
,
Wu
X
,
Dong
Y
,
Xu
Z
,
Zhang
Y
,
Xie
Z
:
Anesthetic sevoflurane causes neurotoxicity differently in neonatal naïve and Alzheimer disease transgenic mice.
Anesthesiology
2010
;
112
:
1404
16
45.
Li
Q
,
Zhu
XL
,
Jin
AP
,
Liu
XY
,
Zhao
YX
:
Inhibition of synaptophysin ubiquitination may improve the intelligent drop due to high glucose and hypoxia.
Int J Clin Exp Med
2014
;
7
:
5021
30
46.
Ma
XZ
,
Bartczak
A
,
Zhang
J
,
Khattar
R
,
Chen
L
,
Liu
MF
,
Edwards
A
,
Levy
G
,
McGilvray
ID
:
Proteasome inhibition in vivo promotes survival in a lethal murine model of severe acute respiratory syndrome.
J Virol
2010
;
84
:
12419
28
47.
Li
Y
,
Stockton
ME
,
Bhuiyan
I
,
Eisinger
BE
,
Gao
Y
,
Miller
JL
,
Bhattacharyya
A
,
Zhao
X
:
MDM2 inhibition rescues neurogenic and cognitive deficits in a mouse model of fragile X syndrome.
Sci Transl Med
2016
;
8
:
336ra61
48.
Zhang
Y
,
Dong
Y
,
Wu
X
,
Lu
Y
,
Xu
Z
,
Knapp
A
,
Yue
Y
,
Xu
T
,
Xie
Z
:
The mitochondrial pathway of anesthetic isoflurane-induced apoptosis.
J Biol Chem
2010
;
285
:
4025
37
49.
Dong
Y
,
Zhang
G
,
Zhang
B
,
Moir
RD
,
Xia
W
,
Marcantonio
ER
,
Culley
DJ
,
Crosby
G
,
Tanzi
RE
,
Xie
Z
:
The common inhalational anesthetic sevoflurane induces apoptosis and increases β-amyloid protein levels.
Arch Neurol
2009
;
66
:
620
31
50.
Ma
J
,
Lindquist
S
:
Conversion of PrP to a self-perpetuating PrPSc-like conformation in the cytosol.
Science
2002
;
298
:
1785
8
51.
Lu
D
,
Lin
W
,
Gao
X
,
Wu
S
,
Cheng
C
,
Avila
J
,
Heese
A
,
Devarenne
TP
,
He
P
,
Shan
L
:
Direct ubiquitination of pattern recognition receptor FLS2 attenuates plant innate immunity.
Science
2011
;
332
:
1439
42
52.
Vassilev
LT
,
Vu
BT
,
Graves
B
,
Carvajal
D
,
Podlaski
F
,
Filipovic
Z
,
Kong
N
,
Kammlott
U
,
Lukacs
C
,
Klein
C
,
Fotouhi
N
,
Liu
EA
:
In vivo activation of the p53 pathway by small-molecule antagonists of MDM2.
Science
2004
;
303
:
844
8
53.
Tovar
C
,
Rosinski
J
,
Filipovic
Z
,
Higgins
B
,
Kolinsky
K
,
Hilton
H
,
Zhao
X
,
Vu
BT
,
Qing
W
,
Packman
K
,
Myklebost
O
,
Heimbrook
DC
,
Vassilev
LT
:
Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy.
Proc Natl Acad Sci USA
2006
;
103
:
1888
93
54.
Kim
E
,
Sheng
M
:
PDZ domain proteins of synapses.
Nat Rev Neurosci
2004
;
5
:
771
81
55.
Ehrlich
I
,
Malinow
R
:
Postsynaptic density 95 controls AMPA receptor incorporation during long-term potentiation and experience-driven synaptic plasticity.
J Neurosci
2004
;
24
:
916
27
56.
Stein
V
,
House
DR
,
Bredt
DS
,
Nicoll
RA
:
Postsynaptic density-95 mimics and occludes hippocampal long-term potentiation and enhances long-term depression.
J Neurosci
2003
;
23
:
5503
6
57.
Weissman
AM
:
Themes and variations on ubiquitylation.
Nat Rev Mol Cell Biol
2001
;
2
:
169
78
58.
El-Husseini
AE
,
Schnell
E
,
Chetkovich
DM
,
Nicoll
RA
,
Bredt
DS
:
PSD-95 involvement in maturation of excitatory synapses.
Science
2000
;
290
:
1364
8
59.
Aarts
M
,
Liu
Y
,
Liu
L
,
Besshoh
S
,
Arundine
M
,
Gurd
JW
,
Wang
YT
,
Salter
MW
,
Tymianski
M
:
Treatment of ischemic brain damage by perturbing NMDA receptor- PSD-95 protein interactions.
Science
2002
;
298
:
846
50
60.
Maletic-Savatic
M
,
Malinow
R
,
Svoboda
K
:
Rapid dendritic morphogenesis in CA1 hippocampal dendrites induced by synaptic activity.
Science
1999
;
283
:
1923
7
61.
Liu
D
,
Diorio
J
,
Day
JC
,
Francis
DD
,
Meaney
MJ
:
Maternal care, hippocampal synaptogenesis and cognitive development in rats.
Nat Neurosci
2000
;
3
:
799
806