Background

In human cortical neural progenitor cells, we investigated the effects of propofol on calcium homeostasis in both the ryanodine and inositol 1,4,5-trisphosphate calcium release channels. We also studied propofol-mediated effects on autophagy, cell survival, and neuro- and gliogenesis.

Methods

The dose–response relationship between propofol concentration and duration was studied in neural progenitor cells. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase release assays. The effects of propofol on cytosolic calcium concentration were evaluated using Fura-2, and autophagy activity was determined by LC3II expression levels with Western blot. Proliferation and differentiation were evaluated by bromodeoxyuridine incorporation and immunostaining with neuronal and glial markers.

Results

Propofol dose- and time-dependently induced cell damage and elevated LC3II expression, most robustly at 200 µM for 24 h (67 ± 11% of control, n = 12 to 19) and 6 h (2.4 ± 0.5 compared with 0.6 ± 0.1 of control, n = 7), respectively. Treatment with 200 μM propofol also increased cytosolic calcium concentration (346 ± 71% of control, n = 22 to 34). Propofol at 10 µM stimulated neural progenitor cell proliferation and promoted neuronal cell fate, whereas propofol at 200 µM impaired neuronal proliferation and promoted glial cell fate (n = 12 to 20). Cotreatment with ryanodine and inositol 1,4,5-trisphosphate receptor antagonists and inhibitors, cytosolic Ca2+ chelators, or autophagy inhibitors mostly mitigated the propofol-mediated effects on survival, proliferation, and differentiation.

Conclusions

These results suggest that propofol-mediated cell survival or neurogenesis is closely associated with propofol’s effects on autophagy by activation of ryanodine and inositol 1,4,5-trisphosphate receptors.

What We Already Know about This Topic
  • In neonatal rodents, propofol has been reported to cause brain cell degeneration and learning and behavior deficits

  • In neural progenitor cell cultures, isoflurane was reported to cause changes in cell proliferation and differentiation

What This Article Tells Us That Is New
  • In neural progenitor cell cultures, only extremely high concentrations of propofol for 24 h caused cell damage and impaired neuronal proliferation

  • Propofol effects appeared related to autophagy

PROPOFOL, the most commonly used intravenous anesthetic agent, has been reported to cause brain cell degeneration,1  as well as learning and behavior deficits,2  in neonatal rodents. These studies have raised significant safety concerns over the administration of anesthetics in the pediatric population, and we propose that propofol may regulate neurogenesis when administered early in life. Neural progenitor cells (NPCs), which are plentiful in the postnatal developing rodent brain, are capable of differentiating into neurons and glial cells and provide a promising cell model to probe the underlying mechanisms governing anesthetic-induced neurotoxicity. Previous studies testing the administration of isoflurane have identified neuronal apoptosis in immature neurons3  but not in NPCs.4,5  However, changes in both proliferation and differentiation of NPCs were identified.4,5  It remains unknown whether propofol has the same effect on NPCs as isoflurane. The aim of this study is to determine propofol’s effects on and the role of autophagy activity in cortical-derived NPC viability, proliferation, and differentiation.

NPC Cultures

ReNcell CX cells, an immortalized human NPC line obtained from human fetal cortex (Millipore, USA), were cultured following the manufacturer’s protocol. For all experiments, cells frozen between passages 6 and 15 were thawed and resuspended in laminin-coated (Sigma-Aldrich, USA) T75-cm2 tissue culture flasks in ReNcell NSC maintenance medium (Millipore). To ensure that the cells remained in a proliferative state, 20 ng/ml of basic fibroblast growth factor (Sigma-Aldrich) and epidermal growth factor (Millipore) were added to the medium. The cell cultures were maintained in an incubator at 37°C, 95% humidity, and 5% CO2. Culture medium was replaced every 24 h. Differentiation was induced by withdrawal of both growth factors (basic fibroblast growth factor and epidermal growth factor) at a confluence of approximately 70%.

Determination of Cytotoxicity

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma-Aldrich) reduction assays were used to measure the cellular redox activity, a relatively early cell damage indicator. The assay measured the activity of mitochondrial dehydrogenase, which reduces MTT to formazan. MTT (0.5 mg/ml) was added to the growth medium in 96-well plates and incubated with NPCs for 4 h at 37°C. Formazan was solubilized from the medium in 150 μl of dimethyl sulfoxide, and the optical density was measured at 540 nm (Synergy™ H1 microplate reader; BioTek, USA). A lactate dehydrogenase (LDH) release assay (Thermo Scientific, USA) was used to quantify disruption of membrane integrity, an indicator of later stage cell damage, as described previously,6  by measuring lactate dehydrogenase released by the cells into the medium. Briefly, 50 μl of the medium was added to 96-well plates with the reaction mixture for 30 min at room temperature. The reaction was stopped, and the mixture solution was measured at 490 and 680 nm (Synergy™ H1 microplate reader; BioTek). Background signal of the medium was deducted from control signals. The mean signal was determined from 6 to 10 wells per condition from three or four separate cultures for each condition (n = 18). The data were presented as percentages of vehicle control.

Cell Proliferation Assays

ReNcell CX cells were plated onto laminin-coated coverslips for 4 h in the proliferation medium containing the growth factors (see NPC Cultures). 5-Bromodeoxyuridine (Invitrogen, USA) at a concentration of 10 μM was mixed with the medium 24 h before the end of the propofol treatment. Then the cells were fixed (4% paraformaldehyde) and permeabilized (0.1% Triton X-100 in phosphate-buffered saline [PBS]). The cells then underwent acid treatment (1 N HCL on ice for 10 min followed by 2 N HCL at room temperature for 10 min), which separated the DNA into single strands so that the primary antibody could detect the 5-bromodeoxyuridine. After incubation with blocking serum (1% bovine serum and 10% goat serum in PBS), the cells were incubated overnight with rat monoclonal anti-5-bromodeoxyuridine primary antibody (1:100) (Santa Cruz Biotechnology, USA) at 4°C. The cells were washed (0.1% Triton X-100 in PBS), and 5-bromodeoxyuridine was detected with fluorescently labeled secondary antibody conjugated with anti-rat IgG (1:1,000 for 2 h) (Invitrogen). The immunostained cells were mounted on microscope slides with Prolong Gold Antifade Reagent containing 4′,6-diamidino-2-phenylindole (DAPI; Invitrogen) for visualization of cell nuclei. The cells were imaged on an Olympus BX41TF fluorescence microscope (200×; Olympus, USA) equipped with iVision v10.10.5 software (Biovision Technologies, USA). The number of DAPI-labeled cells and the number of 5-bromodeoxyuridine-labeled cells were counted, and the mean number of cells was calculated from 5 random areas of each coverslip, with 5 to 10 coverslips per condition, from 3 to 4 different cultures. The experimental n equals the number of coverslips. The data are expressed as the percentages of the number of 5-bromodeoxyuridine-positive cells to the total number of cells.

Cell Differentiation Assays

ReNcell CX cells were initially cultured with the same proliferation medium containing growth factors (see NPC Cultures) until just before the differentiation experiments, when the medium was replaced with medium devoid of growth factors. After the propofol treatment, the cells were allowed to differentiate for an additional 3 days, fixed (4% formaldehyde), and processed for immunocytochemistry overnight at 4°C using mouse monoclonal antibody reactive to the neuronal marker Tuj1 (1:200) (Covance, USA) or the rabbit polyclonal antibody reactive to astrocytes, glial fibrillary acidic protein (GFAP; 1:1,500) (Millipore). Thereafter, cells were incubated with Alexa 594 goat anti-mouse (1:1,000) and Alexa 488 goat anti-rabbit IgG antibodies (1:1,000) (Invitrogen) at room temperature for 1 h to visualize the primary antibody signal. The slides with immunostained cells were coverslipped with Prolong Gold antifade reagent containing DAPI to visualize cell nuclei, examined on an Olympus BX41TF fluorescence microscope (200×; Olympus), and the images were acquired using iVision v10.10.5 software (Biovision Technologies). Tuj1- or GFAP-positive cells overlapping with DAPI signal were analyzed at 10 random locations on each slide. The number of DAPI-labeled cells and the number of Tuj1- or GFAP-positive cells were counted, the mean number of Tuj1- or GFAP-positive cells were calculated for each slide, and the mean as determined for three slides per condition. The n equals the number of slides. The data are expressed as the percentages of the number of Tuj1- or GFAP-positive cells relative to the total number of cells.

Western Blot Analysis

A 6-well cell culture plate on ice was washed once with ice-cold PBS. After aspiration of PBS, 100 μl of ice-cold lysis buffer (1% Triton X-100, 150 mM NaCl, and 50 mM Tris-HCl) was added to each well and maintained on ice for 5 min. The homogenate was collected with a cell scraper and then centrifuged at 4°C in a microcentrifuge at 10,000g for 30 min. The supernatant was gently collected and preserved at −70°C for future use. Bicinchoninic acid assay determined the protein concentration with a bicinchoninic acid kit (Thermo Scientific). For electrophoresis, 25 to 30 μg of protein from different samples were loaded on 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis gels and run with a constant current, and then the protein was transferred to nitrocellulose membranes (Bio-Rad, USA) using a wet transfer system (Bio-Rad). The membranes were blocked with 5% nonfat dry milk dissolved in PBS with 0.2% Tween 20 for 1 h at room temperature and incubated overnight at 4°C with the primary rabbit monoclonal antibody against LC3 (1:1,000) (Cell Signaling Technology, USA) or primary mouse monoclonal antibody against β-actin (1:3,000) (Santa Cruz Biotechnology). This was followed by a wash with secondary antibody conjugated with anti-rabbit and anti-mouse IgG conjugated with horseradish peroxidase (1:10,000) (Bio-Rad) at room temperature for 1 h. The protein on the membranes was detected in a Kodak Image Station 4,000 MM Pro (Kodak, USA) ECL Prime Western blotting detection reagent (GE Healthcare, UK), and images were acquired with Carestream imaging software v5.3.4 (Carestream Health, USA). Signal intensity was quantitatively analyzed with ImageJ v1.49 software (https://imagej.nih.gov/ij/download.html), and the β-actin loading control was used for normalization. The n equals the number of wells for each condition.

Measurement Cytosolic Ca2+ Concentration

The NPC cytosolic Ca2+ concentrations ([Ca2+]c) were measured by Fura-2 acetoxy methylester (AM) fluorescence (Invitrogen) on an Olympus IX70 inverted system microscope (Olympus) equipped with a photometer and IPLab v3.71 software (Scientific Instrument Company, USA), as previously described.6  Briefly, NPCs were plated onto 35-mm laminin-coated culture dishes for 4 h, washed three times in Ca2+-free Dulbecco’s modified Eagle’s medium (Gibco, USA), loaded with 2.5 μM Fura-2 AM in the same buffer at 37°C for 30 min, then washed, and incubated in Ca2+-free Dulbecco’s modified Eagle’s medium for another 30 min at 37°C. The cells were exposed to propofol (200 μM) in minimum essential medium (Gibco), and fluorescence intensities were determined by recording at 340- and 380-nm excitation and at 510-nm emission for up to 5 min for each treatment. The data are presented as ratios of 340/380 nm of fluorescence intensity normalized to baseline. After each imaging experiment, the trypan blue exclusion assay was routinely used to ensure that the cells for [Ca2+]c measurements were healthy and living.

Dose Selection of the Pharmacologic Agents

The doses of dantrolene and xestospongin C used in this experiment were the same as those used in our previous study with the same cell line.6  In a pilot study, we conducted dose responses for bis-N,N,N',N'-tetraacetic acid-acetoxy methylester (BAPTA-AM) and bafilomycin (data not shown). The maximum concentrations that did not induce cytotoxicity (5 μM and 400 nM, respectively) were chosen for this study. The doses of lithium used (2 and 10 mM) were based on previous studies.7,8 

Statistical Analysis

The statistical analyses were performed, and graphs were created using GraphPad Prism 6 software (GraphPad Software, Inc., USA). The sample sizes we used were based on previous publications.6,9,10  One set of MTT and LDH data for the dose response was excluded for suspicious contamination. Parametric variables are expressed as the means ± SD with a Gaussian distribution. The data were analyzed using one-way ANOVA followed by Tukey multiple comparisons testing or two-way ANOVA using propofol concentration and exposure duration as the between-group factors followed by the Bonferroni multiple comparisons test, as detailed in the figure legends. The results were considered statistically significant with P < 0.05, based on two-tailed analysis.

Propofol Induces NPC Cytotoxicity through Excessive Autophagy by Overactivation of Inositol 1,4,5-Trisphosphate and/or Ryanodine Receptors

ReNcell CX NPCs were exposed to different concentrations of propofol (1 to 300 μM) for 2, 6, or 24 h. MTT assays determined that treatment with 10 μM propofol, a clinically relevant concentration, increased cell viability after the 6- and 24-h exposures. On the contrary, propofol at 200 or 300 μM for the same duration (6 h and 24 h) decreased cell viability, whereas 100 μM propofol had no effect on cell survival throughout the experiment (fig. 1A). However, LDH assays showed that significant cytotoxicity was only observed with the 200 μM propofol treatment for 24 h, indicating late-stage cell death (fig. 1B). Our results suggested that the concentration and exposure duration of propofol were the determining factors for the survival of ReNcell CX NPCs.

Fig. 1.

Propofol induces neural progenitor cell cytotoxicity in a time- and dose-dependent manner. ReNcell CX human neural progenitor cells were exposed to different concentrations of propofol (1 to 300 μM) for 2, 6, or 24 h. (A) The 3-(4,5-dimethyithiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) reduction assay was used to determine relatively early cell damage. Treatment with 10 μM propofol increased, whereas 200 μM propofol decreased cell viability after 6- and 24-h exposures. 300 μM propofol induced remarkable cell damage at all three time points relative to vehicle control. #P < 0.05, ##P < 0.01, ####P < 0.0001 compared with treatment for 2 h with the same concentration; &&&P < 0.001 compared with treatment for 6 h; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 compared with vehicle controls at corresponding time points (n = 19 at 2 h; n = 12 at 6 h; n = 17 at 24 h). (B) Relatively late cell damage was determined by lactate dehydrogenase (LDH) release assay. Only 200 μM propofol treatment for 24 h exhibited significant cell damage. ****P < 0.0001 compared with vehicle controls (n = 18). All data are expressed as the means ± SD from at least three separate experiments in duplicate or triplicate and analyzed by two-way ANOVA followed by Tukey multiple comparison tests.

Fig. 1.

Propofol induces neural progenitor cell cytotoxicity in a time- and dose-dependent manner. ReNcell CX human neural progenitor cells were exposed to different concentrations of propofol (1 to 300 μM) for 2, 6, or 24 h. (A) The 3-(4,5-dimethyithiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) reduction assay was used to determine relatively early cell damage. Treatment with 10 μM propofol increased, whereas 200 μM propofol decreased cell viability after 6- and 24-h exposures. 300 μM propofol induced remarkable cell damage at all three time points relative to vehicle control. #P < 0.05, ##P < 0.01, ####P < 0.0001 compared with treatment for 2 h with the same concentration; &&&P < 0.001 compared with treatment for 6 h; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 compared with vehicle controls at corresponding time points (n = 19 at 2 h; n = 12 at 6 h; n = 17 at 24 h). (B) Relatively late cell damage was determined by lactate dehydrogenase (LDH) release assay. Only 200 μM propofol treatment for 24 h exhibited significant cell damage. ****P < 0.0001 compared with vehicle controls (n = 18). All data are expressed as the means ± SD from at least three separate experiments in duplicate or triplicate and analyzed by two-way ANOVA followed by Tukey multiple comparison tests.

Close modal

Previous studies suggested that propofol impacted cellular biology through an autophagy-involved regulatory mechanism.11,12  In this study, we proposed that autophagy plays a role in the cytotoxicity induced by propofol. Western blots showed a time- and concentration-dependent increase in the level of the autophagy biomarker LC3II during propofol treatment, peaking at 24 h with 100 μM and at 6 h with 200 μM propofol (fig. 2A). However, exposure to 10 μM propofol, a clinically relevant concentration, did not show a significant difference in LC3II (fig. 2A) in ReNcell CX NPCs. To differentiate between the induction of autophagy from the impairment of autophagy flux, bafilomycin was added to the culture medium at 22 h of the 24-h propofol exposure period. Further increases in the LC3II level after the bafilomycin treatment indicated the induction of autophagy by propofol (fig. 2B).

Fig. 2.

Propofol induced autophagy in neural progenitor cells. ReNcell CX human neural progenitor cells were exposed to propofol concentrations ranging from 1 to 200 μM for 2, 6, or 24 h. (A) Quantitative analysis of Western blots of the autophagy biomarker, LC3II. The LC3-II/β-actin ratio showed that 200 μM propofol increased the levels of LC3-II at exposure durations at all time points (n = 7). (B) A representative immunoblot of LC3II from three separate experiments from neural progenitor cells cultured with 10 or 200 μM propofol for 24 h and the densitometric analysis of the LC3II/β-actin ratio. Further increases in the LC3II level after bafilomycin (Baf-A1; 400 nM) treatment indicated induction of autophagy by propofol (n = 6). All bar graphs are presented as means ± SD from three separate experiments with duplicate samples and analyzed by two-way ANOVA followed by Tukey multiple comparison tests. *P < 0.05, ***P < 0.001, ****P < 0.0001 compared with corresponding time control or as indicated.

Fig. 2.

Propofol induced autophagy in neural progenitor cells. ReNcell CX human neural progenitor cells were exposed to propofol concentrations ranging from 1 to 200 μM for 2, 6, or 24 h. (A) Quantitative analysis of Western blots of the autophagy biomarker, LC3II. The LC3-II/β-actin ratio showed that 200 μM propofol increased the levels of LC3-II at exposure durations at all time points (n = 7). (B) A representative immunoblot of LC3II from three separate experiments from neural progenitor cells cultured with 10 or 200 μM propofol for 24 h and the densitometric analysis of the LC3II/β-actin ratio. Further increases in the LC3II level after bafilomycin (Baf-A1; 400 nM) treatment indicated induction of autophagy by propofol (n = 6). All bar graphs are presented as means ± SD from three separate experiments with duplicate samples and analyzed by two-way ANOVA followed by Tukey multiple comparison tests. *P < 0.05, ***P < 0.001, ****P < 0.0001 compared with corresponding time control or as indicated.

Close modal

Previously, we detected that isoflurane induced ReNcell CX cytotoxicity by Ca2+ release from the endoplasmic reticulum (ER) through inositol 1,4,5-trisphosphate (InsP3) and/or ryanodine receptors (RyRs).6  To evaluate the role that Ca2+ release from InsP3 and RyRs may have in the propofol-mediated cytotoxicity, the intracellular Ca2+ concentrations were measured after propofol treatments in the presence of the inositol 1,4,5-trisphosphate receptor (InsP3R) antagonist xestospongin C, the ryanodine receptor antagonist dantrolene, the intracellular Ca2+ chelator BAPTA-AM, or the InsP3 production inhibitor lithium. Propofol alone induced a greater elevation in the cytosolic Ca2+ concentration ([Ca2+]c) than in the presence of xestospongin C, dantrolene, or BAPTA-AM (fig. 3, A and B). Pretreatment with 2 or 10 mM lithium significantly decreased the peak amplitudes and the area under curve of propofol-evoked [Ca2+]c signals (fig. 3, C and D). These results suggested that propofol possibly increased the [Ca2+]c by Ca2+ release from the InsP3 or RyRs into the ER. Thus, we propose that the cytotoxicity induced by propofol was mediated by these ER membrane receptors’ excessive release of calcium into the cytosol. The MTT reduction assays showed that cotreatment with xestospongin C, dantrolene, BAPTA-AM, or lithium inhibited ReNcell CX cell damage induced by 200 μM propofol for 24 h (fig. 3, E–G).

Fig. 3.

Propofol induces neural progenitor cell (NPC) damage through excessive autophagy by inositol 1,4,5-trisphosphate (InsP3) and/or ryanodine receptors. (A) Average calcium response to 200 μM propofol (Pfl, arrow) in NPCs cultured in normal medium or medium pretreated with 200 nm of the inositol 1,4,5-trisphosphate receptor antagonist, xestospongin C (Xc), 20 μM of the ryanodine receptor antagonist, dantrolene (Dan), or 5 μM of the intracellular calcium chelator, bis-N,N,N',N'-tetraacetic acid-acetoxy methylester (BAPTA-AM). Changes in Fura-2 AM intensities from at least 15 single cells in three separate experiments were measured to assess the F340/F380 ratio, a representative indicator for the cytosolic calcium concentration ([Ca2+]c). The F340/F380 ratios were normalized to baseline. (B) Pretreatment with Xc, Dan, or BAPTA-AM decreased the peak amplitudes and the areas under the curve (AUC) of the propofol-evoked [Ca2+]c signals. ****P < 0.0001 compared with propofol administration alone. (C) Average calcium response to 200 μM propofol (arrow) in NPCs cultured in normal medium or medium pretreated with inositol 1,4,5-trisphosphate production inhibitor lithium (Li; 2 or 10 mM). (D) Pretreatment of 2 or 10 mM Li significantly decreased the peak amplitudes and AUC of propofol-evoked [Ca2+]c signals. ****P < 0.0001 compared with propofol administration alone. (E–G) Cell viability, determined with the 3-(4,5-dimethyithiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) reduction assay, detected inhibition of cell damage induced by 200 μM propofol for 24 h after cotreatment with Xc and Dan (E), pretreatment with Li for 3 days (F), and cotreatment with BAPTA-AM (G). The data in (E, F) are expressed as the means ± SD from at least three separate experiments and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. **P < 0.01, ***P < 0.001, ****P < 0.0001. (H) A representative immunoblot of LC3II of three separate experiments from cells cultured with 200 μM propofol for 24 h with or without Xc. (I) Dan and the quantitative analysis of LC3II/β-actin ratio (means ± SD). Cotreatment with Xc or Dan alleviated the propofol-induced elevation in the autophagy biomarker, LC3II. (J) Similarly, the NPC cytotoxicity induced by 200 μM propofol for 24 h was mitigated by cotreatment with 5 nM of the autophagy inhibitor 3-methyladenine (3-MA). (K, L) Cotreatment with (K) autophagy inducer rapamycin (Rap; 100 nM) or (L) autophagy flux inhibitor bafilomycin (Baf; 1 nM) promoted NPC damage induced by 200 μM propofol for 24 h. The data in (I, J) are expressed as the means ± SD from at least three separate experiments and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. **P < 0.01, ****P < 0.0001.

Fig. 3.

Propofol induces neural progenitor cell (NPC) damage through excessive autophagy by inositol 1,4,5-trisphosphate (InsP3) and/or ryanodine receptors. (A) Average calcium response to 200 μM propofol (Pfl, arrow) in NPCs cultured in normal medium or medium pretreated with 200 nm of the inositol 1,4,5-trisphosphate receptor antagonist, xestospongin C (Xc), 20 μM of the ryanodine receptor antagonist, dantrolene (Dan), or 5 μM of the intracellular calcium chelator, bis-N,N,N',N'-tetraacetic acid-acetoxy methylester (BAPTA-AM). Changes in Fura-2 AM intensities from at least 15 single cells in three separate experiments were measured to assess the F340/F380 ratio, a representative indicator for the cytosolic calcium concentration ([Ca2+]c). The F340/F380 ratios were normalized to baseline. (B) Pretreatment with Xc, Dan, or BAPTA-AM decreased the peak amplitudes and the areas under the curve (AUC) of the propofol-evoked [Ca2+]c signals. ****P < 0.0001 compared with propofol administration alone. (C) Average calcium response to 200 μM propofol (arrow) in NPCs cultured in normal medium or medium pretreated with inositol 1,4,5-trisphosphate production inhibitor lithium (Li; 2 or 10 mM). (D) Pretreatment of 2 or 10 mM Li significantly decreased the peak amplitudes and AUC of propofol-evoked [Ca2+]c signals. ****P < 0.0001 compared with propofol administration alone. (E–G) Cell viability, determined with the 3-(4,5-dimethyithiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) reduction assay, detected inhibition of cell damage induced by 200 μM propofol for 24 h after cotreatment with Xc and Dan (E), pretreatment with Li for 3 days (F), and cotreatment with BAPTA-AM (G). The data in (E, F) are expressed as the means ± SD from at least three separate experiments and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. **P < 0.01, ***P < 0.001, ****P < 0.0001. (H) A representative immunoblot of LC3II of three separate experiments from cells cultured with 200 μM propofol for 24 h with or without Xc. (I) Dan and the quantitative analysis of LC3II/β-actin ratio (means ± SD). Cotreatment with Xc or Dan alleviated the propofol-induced elevation in the autophagy biomarker, LC3II. (J) Similarly, the NPC cytotoxicity induced by 200 μM propofol for 24 h was mitigated by cotreatment with 5 nM of the autophagy inhibitor 3-methyladenine (3-MA). (K, L) Cotreatment with (K) autophagy inducer rapamycin (Rap; 100 nM) or (L) autophagy flux inhibitor bafilomycin (Baf; 1 nM) promoted NPC damage induced by 200 μM propofol for 24 h. The data in (I, J) are expressed as the means ± SD from at least three separate experiments and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. **P < 0.01, ****P < 0.0001.

Close modal

We further questioned whether InsP3Rs and/or RyRs were also involved in the induction of autophagy flux stimulated by propofol. Blockage of InsP3Rs or RyRs significantly alleviated propofol-induced elevation of the autophagy biomarker LC3II (fig. 3, H and I). ReNcell CX cell damage induced by 200 μM propofol for 24 h was mitigated by cotreatment with autophagy inhibitor 3-methyladenine and promoted by cotreatment with the autophagy inducer rapamycin or the autophagy flux inhibitor bafilomycin (fig. 3, J–L), meaning that excessive autophagy activity was associated with propofol-induced cell damage, which may have resulted in type II autophagic cell death.13 

Propofol Affects Proliferation in NPCs and Regulation of Autophagy

Because autophagy is involved in differentiation and development,14,15  we investigated whether autophagy plays a role in the effects of propofol on proliferation. The impact of propofol on the proliferation of ReNcell NPCs was assessed at various concentrations and exposure times. Compared with control cells, exposure to the clinical concentration of propofol (10 μM) for 24 h promoted cell proliferation, whereas 200 μM propofol treatment significantly decreased proliferation (fig. 4, A and B). Interestingly, cotreatment with xestospongin C or dantrolene alleviated the dual effects of propofol on NPC proliferation, except for the dantrolene treatment with 10 μM propofol (fig. 4, C and D). These results suggest that the dual effects of propofol on cell proliferation is mediated by calcium released from the ER, whereas the distinctly opposing impact of differing concentration may be due to the amount of calcium released. Propofol at 200 µM for 24 h significantly impaired proliferation, but this could be inhibited by the autophagy inhibitor 3-methyladenine (3-MA) and potentiated by the autophagy inducer rapamycin (fig. 4, E and F). However, cotreatment with 3-MA or rapamycin had no effect on the 10 μM propofol-induced proliferation increase (fig. 4G). These results suggest that suppression of proliferation induced by propofol at 200 µM for 24 h was related to increased autophagy activity, whereas the clinical concentration of propofol accelerated cell proliferation via a nonautophagy mechanism.

Fig. 4.

Propofol affects neural progenitor cell proliferation and regulation of autophagy. (A) Representative images showing the double immunostaining of cell nuclei with 4′,6-diamino-2-phenylindole (DAPI, blue, arrows) and 5-bromodeoxyuridine (BrdU, red, arrows) in ReNcell CX neural progenitor cells (NPCs) with or without propofol (10, 200 μM) for 24 h. Scale bar = 50 μm. (B) Quantitative analysis of the effect of a 24-h exposure to 10 or 200 μM propofol on the proliferation of BrdU-positive NPCs. Exposure to 10 μM propofol increased the percentage of BrdU-positive cells, whereas 200 μM propofol resulted in significantly decreased proliferation (n = 30). (C, D) Quantitative analysis of the effect of a 24-h exposure to 10 or 200 μM propofol on the proliferation of BrdU-positive NPCs, with or without the inositol 1,4,5-trisphosphate receptor antagonist, xestospongin C (Xc, 50 nm) (C) or the ryanodine receptor antagonist, dantrolene (Dan, 1 μM) (D). Cotreatment with xestospongin C (n = 20) or dantrolene (n = 14) alleviated the dual effects of propofol on NPC proliferation. (E) Quantitative analysis of the effect of a 24-h exposure to 10 or 200 μM propofol on the proliferation of BrdU-positive NPCs, with or without the autophagy inhibitor 3-methyladenine (3-MA, 5 mM) or the autophagy inducer rapamycin (Rap; 100 nM). (F) Propofol significantly impaired proliferation at 200 µM for 24 h, which was inhibited by 3-MA (n = 15) and potentiated by Rap (n = 15). (G) Cotreatment with 3-MA or Rap had no effect on 10 μM propofol-induced increase of proliferation (n = 17). The above data are expressed as the means ± SD from at least three separate experiments and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. *P < 0.05, **P < 0.01, ***P < 0.001, or ****P < 0.0001, compared with vehicle control or as indicated.

Fig. 4.

Propofol affects neural progenitor cell proliferation and regulation of autophagy. (A) Representative images showing the double immunostaining of cell nuclei with 4′,6-diamino-2-phenylindole (DAPI, blue, arrows) and 5-bromodeoxyuridine (BrdU, red, arrows) in ReNcell CX neural progenitor cells (NPCs) with or without propofol (10, 200 μM) for 24 h. Scale bar = 50 μm. (B) Quantitative analysis of the effect of a 24-h exposure to 10 or 200 μM propofol on the proliferation of BrdU-positive NPCs. Exposure to 10 μM propofol increased the percentage of BrdU-positive cells, whereas 200 μM propofol resulted in significantly decreased proliferation (n = 30). (C, D) Quantitative analysis of the effect of a 24-h exposure to 10 or 200 μM propofol on the proliferation of BrdU-positive NPCs, with or without the inositol 1,4,5-trisphosphate receptor antagonist, xestospongin C (Xc, 50 nm) (C) or the ryanodine receptor antagonist, dantrolene (Dan, 1 μM) (D). Cotreatment with xestospongin C (n = 20) or dantrolene (n = 14) alleviated the dual effects of propofol on NPC proliferation. (E) Quantitative analysis of the effect of a 24-h exposure to 10 or 200 μM propofol on the proliferation of BrdU-positive NPCs, with or without the autophagy inhibitor 3-methyladenine (3-MA, 5 mM) or the autophagy inducer rapamycin (Rap; 100 nM). (F) Propofol significantly impaired proliferation at 200 µM for 24 h, which was inhibited by 3-MA (n = 15) and potentiated by Rap (n = 15). (G) Cotreatment with 3-MA or Rap had no effect on 10 μM propofol-induced increase of proliferation (n = 17). The above data are expressed as the means ± SD from at least three separate experiments and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. *P < 0.05, **P < 0.01, ***P < 0.001, or ****P < 0.0001, compared with vehicle control or as indicated.

Close modal

Propofol Affects NPC Differentiation via Activation of InsP3Rs

Exposure to 10 μM propofol for 24 h promoted neuronal fate as measured by the increase in Tuj1-positive cells and suppressed glial fate as determined by the decrease of GFAP-positive cells (fig. 5, A and B). On the contrary, 200 μM propofol decreased the percentage of Tuj1-positive cells and increased GFAP-positive cells, indicating an enhancement of glial fate. Consistent with propofol’s effects on cytotoxicity and proliferation, cotreatment with the InsP3R antagonist xestospongin C mitigates these dual effects of propofol, suggesting the prominent role of InsP3Rs in these processes (fig. 5, C and D).

Fig. 5.

Propofol affects neural progenitor cell differentiation via inositol 1,4,5-trisphosphate receptors. (A) Representative micrographs showing immunostaining for Tuj1, neuronal class III β-tubulin (red), glial fibrillary acidic protein (GFAP; green), and 4′,6-diamino-2-phenylindole (DAPI; blue) of ReNcell neural progenitor cells cultured in differentiation medium for 3 days. (B) Representative micrographs showing immunostaining of Tuj1 (red), GFAP (green), and DAPI (blue) of neural progenitor cells cultured with 10 or 200 μM propofol, with or without the inositol 1,4,5-trisphosphate receptor antagonist, xestospongin C (Xc; 50 nm), followed by differentiation medium for 3 days. (C, D) Tuj1-positive (C) or GFAP-positive (D) cells were counted, and the data expressed are as percentages of controls. Propofol at 10 μM increased Tuj1-positive neuronal cells but decreased the GFAP-positive glial cells. Propofol at 200 μM had the opposite effect of deceasing Tuj1-positive cells and increasing GFAP-positive cells. These effects could be mitigated by cotreatment with Xc. Scale bar = 50 μm. The data are expressed as the mean ± SD and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. **P < 0.01, ***P < 0.001, ****P < 0.0001.

Fig. 5.

Propofol affects neural progenitor cell differentiation via inositol 1,4,5-trisphosphate receptors. (A) Representative micrographs showing immunostaining for Tuj1, neuronal class III β-tubulin (red), glial fibrillary acidic protein (GFAP; green), and 4′,6-diamino-2-phenylindole (DAPI; blue) of ReNcell neural progenitor cells cultured in differentiation medium for 3 days. (B) Representative micrographs showing immunostaining of Tuj1 (red), GFAP (green), and DAPI (blue) of neural progenitor cells cultured with 10 or 200 μM propofol, with or without the inositol 1,4,5-trisphosphate receptor antagonist, xestospongin C (Xc; 50 nm), followed by differentiation medium for 3 days. (C, D) Tuj1-positive (C) or GFAP-positive (D) cells were counted, and the data expressed are as percentages of controls. Propofol at 10 μM increased Tuj1-positive neuronal cells but decreased the GFAP-positive glial cells. Propofol at 200 μM had the opposite effect of deceasing Tuj1-positive cells and increasing GFAP-positive cells. These effects could be mitigated by cotreatment with Xc. Scale bar = 50 μm. The data are expressed as the mean ± SD and analyzed by one-way ANOVA followed by Tukey multiple comparison tests. **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

This study examined the effects of propofol on human cortex-derived NPCs and the role of calcium-mediated autophagy pathway in neurogenesis and neurodegeneration. We found that propofol induced Ca2+ release from the endoplasmic reticulum of NPCs by activation of the InsP3R and/or RyR. A clinically relevant dose of propofol promoted cell proliferation and favored neuronal cell fate via adequate activation of the InsP3R and adequate autophagy. However, a high dose of propofol induced cell damage and favored glial cell fate through excessive autophagy by overactivation of InsP3Rs and RyRs.

Previous studies have shown that propofol can induce both excitotoxic and apoptotic cell death16  in cell cultures17,18  and in animal models,19,20  although the mechanism is not clear. Most of the previous studies have focused on the anesthetic-induced type 1 cell death (apoptosis),21,22  but cells can also die from type 2 (autophagic cell death) or type 3 cell death (necrosis). Palanisamy et al.23  recently demonstrated that propofol, at approximately 15 and 30 µM for a 4-h exposure, did not induce rat primary NPC necrotic cell death and apoptosis. However, prolonged use of propofol for 24 h transiently impaired proliferation. Consistent with these results, we detected significant NPC cytotoxicity at an extremely high pharmacologic concentration (200 µM) but not at clinically relevant concentrations (1, 10 µM). In contrast, in this study, propofol at 10 µM for 24 h promoted NPC proliferation, whereas in the previous study,23  propofol at about 15 µM (2.5 µg/ml) transiently impaired rat primary NPC proliferation. The discrepancy between these two studies is likely due to the different types of cells, primary versus immortalized NPCs. Future studies in rat primary NPCs at clinically relevant concentrations will be needed to sort out these differences.

There is a consensus that autophagy is required for cell survival and is involved in various physiologic events, including immune responses, cancer, aging, and neurodegeneration,24,25  as well as brain development and differentiation.14,15,26  A recent study has shown that knockdown of the of the autophagy-related gene Atg5 inhibited cortical NPC differentiation but increased proliferation during embryonic brain development.27  The exact role of autophagy during neurogenesis, as well as the consequences of pharmacologic stresses, such as anesthetics, on autophagy, are unclear.

Like most anesthetics including pentobarbital, ketamine, and isoflurane, propofol exhibited up-regulation of autophagy in skeletal muscles28  and human umbilical vein endothelial cells.11  This dose-dependent stimulatory effect of propofol on autophagy flux was also observed in our experiments. To implicate autophagy as a possible contribution to neurogenesis, an autophagy inducer or inhibitor was applied, together with propofol. As confirmed by MTT and proliferation assays, cell damage or decreased proliferation correlated with excessive induction of autophagy flux by propofol exposure. Consistent with our results, autophagy is believed to be a double-edged sword in ischemia and preconditioning.29  Physiologic autophagy eliminates harmful protein aggregates and damaged organelles in cells and thus limits the transmission of harmful signaling; excessive or pathologic autophagy, however, induces type II autophagy cell death and causes irreversible injury.

We also explored changes in cytosolic calcium concentrations [Ca2+]c as a possible mechanism for our observed effects. Previously, propofol has been reported to increased [Ca2+]c in glia, neurons,30  and neural stem/progenitor cells31  and that an increase in [Ca2+]c within the physiologic range promotes cell proliferation, differentiation, synthesis, and catabolism.32  Similarly, we found that [Ca2+]c almost tripled after propofol treatment, and we posit that propofol initiated a Ca2+-dependent signaling pathway.

The γ-aminobutyric acid type A receptor is one of the likely targets for propofol. Previous research has shown that activation of γ-aminobutyric acid receptor type A receptors by isoflurane during brain development may cause cell membrane depolarization, Ca2+ influx through voltage-dependent Ca2+ channels, and increased [Ca2+]c.33  Recently, InsP3Rs have been implicated as a molecular target for isoflurane and to be involved in anesthetic-mediated neurotoxicity.10  The InsP3Rs and RyRs are the main channels through which Ca2+ leaves the ER, although a small amount of Ca2+ leaves the ER through the so-called leak channel. The increase in the [Ca2+]c after isoflurane administration in mature neurons was caused primarily by calcium release from intracellular stores, most likely from the ER. Therefore, we propose that the mechanism of propofol’s impact on neurogenesis on NPCs is InsP3R activation, followed by a rise in the [Ca2+]c, and finally either the induction or an overactivation of autophagy, depending on the amount of Ca2+ released (fig. 6). Indeed, we found that InsP3R inhibition prevented most of the effects of propofol on cell viability, autophagy, proliferation, and differentiation. Although the mechanisms underlying these observed results by propofol in human cortical NPCs are not fully apparent, our results suggest the involvement of InsP3Rs and RyRs. Because the mitochondria and the ER have intracellular communication sites, they may also regulate Ca2+ homeostasis by uptake of calcium released by the ER,34  especially when intracellular calcium concentrations are high, such as after exposures to anesthetics. A previous study35  demonstrated that mitochondrial Ca2+ concentrations are increased after isoflurane-mediated Ca2+ release from the ER via activation of InsP3R, followed by transfer into mitochondria. It is important that future studies investigate whether propofol has similar effects, because mitochondria play important roles in ATP production and autophagy regulation.36 

Fig. 6.

Summary of the effects of propofol on neuronal progenitor cell (NPC) proliferation and differentiation. Exposure to a clinically relevant concentration of propofol induces calcium release from the endoplasmic reticulum (ER) through the activation of the calcium channels, inositol 1,4,5-trisphosphate receptors (InsP3R), and/or ryanodine receptors (RyR). This results in a moderate increase in the concentration of cytosolic calcium that stimulates NPC proliferation and neuronal and glial cell differentiation. These effects involve a nonautophagy mechanism. On the other hand, a prolonged exposure to a high concentration of propofol induces the overactivation of InsP3Rs and/or RyRs on the ER membrane, thereby allowing the buildup of intracellular Ca2+. This elevated concentration of cytosolic calcium leads to excessive autophagy and the inhibition of NPC proliferation, which contributes to the propofol-mediated neurotoxicity. BAPTA-AM = bis-N,N,N',N'-tetraacetic acid-acetoxy methylester; IMP = inositol monophosphatase; IP3 = inositol 1,4,5-trisphosphate; 3-MA = 3-methyladenine.

Fig. 6.

Summary of the effects of propofol on neuronal progenitor cell (NPC) proliferation and differentiation. Exposure to a clinically relevant concentration of propofol induces calcium release from the endoplasmic reticulum (ER) through the activation of the calcium channels, inositol 1,4,5-trisphosphate receptors (InsP3R), and/or ryanodine receptors (RyR). This results in a moderate increase in the concentration of cytosolic calcium that stimulates NPC proliferation and neuronal and glial cell differentiation. These effects involve a nonautophagy mechanism. On the other hand, a prolonged exposure to a high concentration of propofol induces the overactivation of InsP3Rs and/or RyRs on the ER membrane, thereby allowing the buildup of intracellular Ca2+. This elevated concentration of cytosolic calcium leads to excessive autophagy and the inhibition of NPC proliferation, which contributes to the propofol-mediated neurotoxicity. BAPTA-AM = bis-N,N,N',N'-tetraacetic acid-acetoxy methylester; IMP = inositol monophosphatase; IP3 = inositol 1,4,5-trisphosphate; 3-MA = 3-methyladenine.

Close modal

The outcome of either physiologic autophagy or pathologic autophagy may be explained by InsP3Rs and intracellular Ca2+ signaling. Although apparently contradictory, both autophagic flux37  and inhibition38  result from autolysosomal formation from an increase in autophagosomes to lysosomes. Our experimental data provide compelling evidence for both the propofol-mediated dose-dependent increase in [Ca2+]c and the subsequent induction of autophagy. The different outcomes of autophagy are possibly due to a divergent role of the InsP3R with respect to basal versus stress-induced autophagy and the different spatiotemporal characteristics of Ca2+ signals that can be generated, each having distinct impacts on different steps in the autophagy pathway.

There is evidence, from in vitro39  and in vivo40  studies, that lithium, a common drug for the treatment of neuropsychiatric disorders, has facilitated neurogenesis and reduced the neurotoxicity induced by certain stressors, including anesthetics.41  It has been proposed that the mechanism for the neuroprotective properties of lithium result from its interactions with cell survival and cell death pathways. Lithium can affect intracellular Ca2+ homeostasis by inhibiting N-methyl-d-aspartate receptor-mediated Ca2+ entry into the cell, as well as reducing InsP3 production by inhibiting inositol monophosphatase. Lithium can also affect autophagy via its regulation of intracellular Ca2+ homeostasis,42  inhibition of GSK-3β, and activation of the inhibitory regulator mammalian target of rapamycin.43  Thus, the effects of lithium observed in our experiment may be explained by its effects on the regulation of calcium homeostasis and the associated autophagy process. In addition, lithium may affect propofol’s effects due to their mutual interaction at the voltage-gated Na+ channel, because both have been demonstrated to affect these Na+ channels.44  Thus, the effects of lithium appear to outweigh the effects of propofol at lower concentrations than at higher anesthetic concentrations.

Propofol exposure during the period of rapid brain development in neonatal animals has negatively impacted hippocampal structure and function, resulting in subsequent cognitive impairment.2,45  The mechanisms for propofol’s deleterious effects during development are conflicting, including interference with n eural stem cell function, neurogenesis, and synapse formation.4,46  Such controversy may be due to confounding variables in the use of animal models.47,48  In an attempt to reduce such variables, we chose to study NPCs in vitro. Our results suggested that at clinically relevant concentrations of propofol, proliferation contributed more to the increased cell viability than differentiation. Although the increased cell viability may also be due to suppressed apoptosis, a previous study indicated that apoptosis was not a major factor associated with improved viability when neural stem cells were treated with propofol.9  Propofol administration is generally considered to be excitotoxic neurodegeneration, which is fundamentally different, both in type and sequence, from that of the apoptotic phenomenon.16  Propofol toxicity was only observed at concentrations that exceeded clinically relevant concentrations. Corresponding with previous results, propofol at clinical concentrations increased NPC differentiation into neurons,49  whereas differentiation into astrocytes was decreased.50 

When interpreting the data presented in our study, some limitations must be considered. Our investigations into the mechanisms of the propofol-induced changes on calcium homeostasis and autophagy, and thus NPC fate, were not exhaustive. Recent studies indicated that enhanced cAMP response element binding protein (CREB) phosphorylation may play a role in the Ca2+-mediated pathway9  and in both proliferation and differentiation of neural stem cells.51  Furthermore, the protective effect of xenon preconditioning for asphyxia is thought to involve pCREB-regulated protein synthesis in rat brain.52  Downstream signaling after CREB activation may be the key to direct stem cell fate, and further study is required to elucidate the mechanisms.

In conclusion, only extreme, high pharmacologic concentrations of propofol adversely affect NPC cell viability in vitro by excessive autophagy through a Ca2+-mediated pathway. Clinically relevant doses of propofol enhance proliferation of NPCs and increase neuronal differentiation by a Ca2+-related nonautophagic mechanism.

We appreciate the valuable discussions and support from Roderic G. Eckenhoff, M.D., Maryellen F. Eckenhoff, Ph.D., and Lee A. Fleisher, M.D., Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.

Supported by grant Nos. K08-GM073224, R01GM084979, 3R01GM084979-02S1, and 2R01GM084979-06A1 from the National Institute of General Medical Science, National Institutes of Health, Baltimore, Maryland (to Dr. Wei); research grant No. 12-FY08-167 from the March of Dimes Birth Defects Foundation, White Plains, New York (to Dr. Wei); and a bridging fund from the Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.

The authors declare no competing interests.

1.
Milanovic
D
,
Popic
J
,
Pesic
V
,
Loncarevic-Vasiljkovic
N
,
Kanazir
S
,
Jevtovic-Todorovic
V
,
Ruzdijic
S
:
Regional and temporal profiles of calpain and caspase-3 activities in postnatal rat brain following repeated propofol administration.
Dev Neurosci
2010
;
32
:
288
301
2.
Karen
T
,
Schlager
GW
,
Bendix
I
,
Sifringer
M
,
Herrmann
R
,
Pantazis
C
,
Enot
D
,
Keller
M
,
Kerner
T
,
Felderhoff-Mueser
U
:
Effect of propofol in the immature rat brain on short- and long-term neurodevelopmental outcome.
PLoS One
2013
;
8
:
e64480
3.
Istaphanous
GK
,
Howard
J
,
Nan
X
,
Hughes
EA
,
McCann
JC
,
McAuliffe
JJ
,
Danzer
SC
,
Loepke
AW
:
Comparison of the neuroapoptotic properties of equipotent anesthetic concentrations of desflurane, isoflurane, or sevoflurane in neonatal mice.
Anesthesiology
2011
;
114
:
578
87
4.
Sall
JW
,
Stratmann
G
,
Leong
J
,
McKleroy
W
,
Mason
D
,
Shenoy
S
,
Pleasure
SJ
,
Bickler
PE
:
Isoflurane inhibits growth but does not cause cell death in hippocampal neural precursor cells grown in culture.
Anesthesiology
2009
;
110
:
826
33
5.
Culley
DJ
,
Boyd
JD
,
Palanisamy
A
,
Xie
Z
,
Kojima
K
,
Vacanti
CA
,
Tanzi
RE
,
Crosby
G
:
Isoflurane decreases self-renewal capacity of rat cultured neural stem cells.
Anesthesiology
2011
;
115
:
754
63
6.
Zhao
X
,
Yang
Z
,
Liang
G
,
Wu
Z
,
Peng
Y
,
Joseph
DJ
,
Inan
S
,
Wei
H
:
Dual effects of isoflurane on proliferation, differentiation, and survival in human neuroprogenitor cells.
Anesthesiology
2013
;
118
:
537
49
7.
Bosche
B
,
Schäfer
M
,
Graf
R
,
Härtel
FV
,
Schäfer
U
,
Noll
T
:
Lithium prevents early cytosolic calcium increase and secondary injurious calcium overload in glycolytically inhibited endothelial cells.
Biochem Biophys Res Commun
2013
;
434
:
268
72
8.
De-Paula
Vde J
,
Kerr
DS
,
de Carvalho
MP
,
Schaeffer
EL
,
Talib
LL
,
Gattaz
WF
,
Forlenza
OV
:
Long-term lithium treatment increases cPLA2 and iPLA2 activity in cultured cortical and hippocampal neurons.
Molecules
2015
;
20
:
19878
85
9.
Tao
T
,
Zhao
Z
,
Hao
L
,
Gu
M
,
Chen
L
,
Tang
J
:
Propofol promotes proliferation of cultured adult rat hippocampal neural stem cells.
J Neurosurg Anesthesiol
2013
;
25
:
299
305
10.
Joseph
JD
,
Peng
Y
,
Mak
DO
,
Cheung
KH
,
Vais
H
,
Foskett
JK
,
Wei
H
:
General anesthetic isoflurane modulates inositol 1,4,5-trisphosphate receptor calcium channel opening.
Anesthesiology
2014
;
121
:
528
37
11.
Chang
CY
,
Chen
PH
,
Lu
SC
,
Hsieh
MC
,
Lin
CW
,
Lee
HM
,
Jawan
B
,
Kao
YH
:
Propofol-enhanced autophagy increases motility and angiogenic capacity of cultured human umbilical vascular endothelial cells.
Life Sci
2015
;
142
:
49
59
12.
Cui
DR
,
Wang
L
,
Jiang
W
,
Qi
AH
,
Zhou
QH
,
Zhang
XL
:
Propofol prevents cerebral ischemia-triggered autophagy activation and cell death in the rat hippocampus through the NF-κB/p53 signaling pathway.
Neuroscience
2013
;
246
:
117
32
13.
Green
DR
,
Llambi
F
:
Cell death signaling.
Cold Spring Harb Perspect Biol
2015
;
7
:
a006080
14.
Zhao
Y
,
Huang
Q
,
Yang
J
,
Lou
M
,
Wang
A
,
Dong
J
,
Qin
Z
,
Zhang
T
:
Autophagy impairment inhibits differentiation of glioma stem/progenitor cells.
Brain Res
2010
;
1313
:
250
8
15.
Vázquez
P
,
Arroba
AI
,
Cecconi
F
,
de la Rosa
EJ
,
Boya
P
,
de Pablo
F
:
Atg5 and Ambra1 differentially modulate neurogenesis in neural stem cells.
Autophagy
2012
;
8
:
187
99
16.
Ishimaru
MJ
,
Ikonomidou
C
,
Tenkova
TI
,
Der
TC
,
Dikranian
K
,
Sesma
MA
,
Olney
JW
:
Distinguishing excitotoxic from apoptotic neurodegeneration in the developing rat brain.
J Comp Neurol
1999
;
408
:
461
76
17.
Luo
T
,
Wu
J
,
Kabadi
SV
,
Sabirzhanov
B
,
Guanciale
K
,
Hanscom
M
,
Faden
J
,
Cardiff
K
,
Bengson
CJ
,
Faden
AI
:
Propofol limits microglial activation after experimental brain trauma through inhibition of nicotinamide adenine dinucleotide phosphate oxidase.
Anesthesiology
2013
;
119
:
1370
88
18.
Pearn
ML
,
Hu
Y
,
Niesman
IR
,
Patel
HH
,
Drummond
JC
,
Roth
DM
,
Akassoglou
K
,
Patel
PM
,
Head
BP
:
Propofol neurotoxicity is mediated by p75 neurotrophin receptor activation.
Anesthesiology
2012
;
116
:
352
61
19.
Yang
B
,
Liang
G
,
Khojasteh
S
,
Wu
Z
,
Yang
W
,
Joseph
D
,
Wei
H
:
Comparison of neurodegeneration and cognitive impairment in neonatal mice exposed to propofol or isoflurane.
PLoS One
2014
;
9
:
e99171
20.
Creeley
C
,
Dikranian
K
,
Dissen
G
,
Martin
L
,
Olney
J
,
Brambrink
A
:
Propofol-induced apoptosis of neurones and oligodendrocytes in fetal and neonatal rhesus macaque brain.
Br J Anaesth
2013
;
110
(
suppl 1
):
i29
38
21.
Peng
J
,
Drobish
JK
,
Liang
G
,
Wu
Z
,
Liu
C
,
Joseph
DJ
,
Abdou
H
,
Eckenhoff
MF
,
Wei
H
:
Anesthetic preconditioning inhibits isoflurane-mediated apoptosis in the developing rat brain.
Anesth Analg
2014
;
119
:
939
46
22.
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
23.
Palanisamy
A
,
Friese
MB
,
Cotran
E
,
Moller
L
,
Boyd
JD
,
Crosby
G
,
Culley
DJ
:
Prolonged treatment with propofol transiently impairs proliferation but not survival of rat neural progenitor cells in vitro.
PLoS One
2016
;
11
:
e0158058
24.
Levine
B
,
Kroemer
G
:
Autophagy in the pathogenesis of disease.
Cell
2008
;
132
:
27
42
25.
Mizushima
N
,
Levine
B
:
Autophagy in mammalian development and differentiation.
Nat Cell Biol
2010
;
12
:
823
30
26.
Boya
P
,
Mellén
MA
,
de la Rosa
EJ
:
How autophagy is related to programmed cell death during the development of the nervous system.
Biochem Soc Trans
2008
;
36
:
813
7
27.
Lv
X
,
Jiang
H
,
Li
B
,
Liang
Q
,
Wang
S
,
Zhao
Q
,
Jiao
J
:
The crucial role of Atg5 in cortical neurogenesis during early brain development.
Sci Rep
2014
;
4
:
6010
28.
Kashiwagi
A
,
Hosokawa
S
,
Maeyama
Y
,
Ueki
R
,
Kaneki
M
,
Martyn
JA
,
Yasuhara
S
:
Anesthesia with disuse leads to autophagy up-regulation in the skeletal muscle.
Anesthesiology
2015
;
122
:
1075
83
29.
Sheng
R
,
Qin
ZH
:
The divergent roles of autophagy in ischemia and preconditioning.
Acta Pharmacol Sin
2015
;
36
:
411
20
30.
Kahraman
S
,
Zup
SL
,
McCarthy
MM
,
Fiskum
G
:
GABAergic mechanism of propofol toxicity in immature neurons.
J Neurosurg Anesthesiol
2008
;
20
:
233
40
31.
Tozuka
Y
,
Fukuda
S
,
Namba
T
,
Seki
T
,
Hisatsune
T
:
GABAergic excitation promotes neuronal differentiation in adult hippocampal progenitor cells.
Neuron
2005
;
47
:
803
15
32.
Rushton
DJ
,
Mattis
VB
,
Svendsen
CN
,
Allen
ND
,
Kemp
PJ
:
Stimulation of GABA-induced Ca2+ influx enhances maturation of human induced pluripotent stem cell-derived neurons.
PLoS One
2013
;
8
:
e81031
33.
Zhao
YL
,
Xiang
Q
,
Shi
QY
,
Li
SY
,
Tan
L
,
Wang
JT
,
Jin
XG
,
Luo
AL
:
GABAergic excitotoxicity injury of the immature hippocampal pyramidal neurons’ exposure to isoflurane.
Anesth Analg
2011
;
113
:
1152
60
34.
De Stefani
D
,
Raffaello
A
,
Teardo
E
,
Szabò
I
,
Rizzuto
R
:
A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter.
Nature
2011
;
476
:
336
40
35.
Yang
H
,
Liang
G
,
Hawkins
BJ
,
Madesh
M
,
Pierwola
A
,
Wei
H
:
Inhalational anesthetics induce cell damage by disruption of intracellular calcium homeostasis with different potencies.
Anesthesiology
2008
;
109
:
243
50
36.
Cárdenas
C
,
Miller
RA
,
Smith
I
,
Bui
T
,
Molgó
J
,
Müller
M
,
Vais
H
,
Cheung
KH
,
Yang
J
,
Parker
I
,
Thompson
CB
,
Birnbaum
MJ
,
Hallows
KR
,
Foskett
JK
:
Essential regulation of cell bioenergetics by constitutive InsP3 receptor Ca2+ transfer to mitochondria.
Cell
2010
;
142
:
270
83
37.
Decuypere
JP
,
Parys
JB
,
Bultynck
G
:
ITPRs/inositol 1,4,5-trisphosphate receptors in autophagy: From enemy to ally.
Autophagy
2015
;
11
:
1944
8
38.
Decuypere
JP
,
Bultynck
G
,
Parys
JB
:
A dual role for Ca2+ in autophagy regulation.
Cell Calcium
2011
;
50
:
242
50
39.
Hashimoto
R
,
Senatorov
V
,
Kanai
H
,
Leeds
P
,
Chuang
DM
:
Lithium stimulates progenitor proliferation in cultured brain neurons.
Neuroscience
2003
;
117
:
55
61
40.
Bianchi
P
,
Ciani
E
,
Contestabile
A
,
Guidi
S
,
Bartesaghi
R
:
Lithium restores neurogenesis in the subventricular zone of the Ts65Dn mouse, a model for Down syndrome.
Brain Pathol
2010
;
20
:
106
18
41.
Straiko
MM
,
Young
C
,
Cattano
D
,
Creeley
CE
,
Wang
H
,
Smith
DJ
,
Johnson
SA
,
Li
ES
,
Olney
JW
:
Lithium protects against anesthesia-induced developmental neuroapoptosis.
Anesthesiology
2009
;
110
:
862
8
42.
Chiu
CT
,
Chuang
DM
:
Molecular actions and therapeutic potential of lithium in preclinical and clinical studies of CNS disorders.
Pharmacol Ther
2010
;
128
:
281
304
43.
Sarkar
S
,
Perlstein
EO
,
Imarisio
S
,
Pineau
S
,
Cordenier
A
,
Maglathlin
RL
,
Webster
JA
,
Lewis
TA
,
O’Kane
CJ
,
Schreiber
SL
,
Rubinsztein
DC
:
Small molecules enhance autophagy and reduce toxicity in Huntington’s disease models.
Nat Chem Biol
2007
;
3
:
331
8
44.
Sibarov
DA
,
Abushik
PA
,
Poguzhelskaya
EE
,
Bolshakov
KV
,
Antonov
SM
:
Inhibition of plasma membrane Na/Ca-exchanger by KB-R7943 or lithium reveals its role in Ca-dependent N-methyl-d-aspartate receptor inactivation.
J Pharmacol Exp Ther
2015
;
355
:
484
95
45.
Kargaran
P
,
Lenglet
S
,
Montecucco
F
,
Mach
F
,
Copin
JC
,
Vutskits
L
:
Impact of propofol anaesthesia on cytokine expression profiles in the developing rat brain: A randomised placebo-controlled experimental in-vivo study.
Eur J Anaesthesiol
2015
;
32
:
336
45
46.
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
47.
Lasarzik
I
,
Winkelheide
U
,
Stallmann
S
,
Orth
C
,
Schneider
A
,
Tresch
A
,
Werner
C
,
Engelhard
K
:
Assessment of postischemic neurogenesis in rats with cerebral ischemia and propofol anesthesia.
Anesthesiology
2009
;
110
:
529
37
48.
Huang
J
,
Jing
S
,
Chen
X
,
Bao
X
,
Du
Z
,
Li
H
,
Yang
T
,
Fan
X
:
Propofol administration during early postnatal life suppresses hippocampal neurogenesis.
Mol Neurobiol
2016
;
53
:
1031
44
49.
Sall
JW
,
Stratmann
G
,
Leong
J
,
Woodward
E
,
Bickler
PE
:
Propofol at clinically relevant concentrations increases neuronal differentiation but is not toxic to hippocampal neural precursor cells in vitro.
Anesthesiology
2012
;
117
:
1080
90
50.
Li
Q
,
Lu
J
,
Wang
X
:
Propofol and remifentanil at moderate and high concentrations affect proliferation and differentiation of neural stem/progenitor cells.
Neural Regen Res
2014
;
9
:
2002
7
51.
Miyamoto
N
,
Tanaka
R
,
Zhang
N
,
Shimura
H
,
Onodera
M
,
Mochizuki
H
,
Hattori
N
,
Urabe
T
:
Crucial role for Ser133-phosphorylated form of cyclic AMP-responsive element binding protein signaling in the differentiation and survival of neural progenitors under chronic cerebral hypoperfusion.
Neuroscience
2009
;
162
:
525
36
52.
Ma
D
,
Hossain
M
,
Pettet
GK
,
Luo
Y
,
Lim
T
,
Akimov
S
,
Sanders
RD
,
Franks
NP
,
Maze
M
:
Xenon preconditioning reduces brain damage from neonatal asphyxia in rats.
J Cereb Blood Flow Metab
2006
;
26
:
199
208