Background

Propofol exposure to neurons during synaptogenesis results in apoptosis, leading to cognitive dysfunction in adulthood. Previous work from our laboratory showed that isoflurane neurotoxicity occurs through p75 neurotrophin receptor (p75(NTR)) and subsequent cytoskeleton depolymerization. Given that isoflurane and propofol both suppress neuronal activity, we hypothesized that propofol also induces apoptosis in developing neurons through p75(NTR).

Methods

Days in vitro 5-7 neurons were exposed to propofol (3 μM) for 6 h and apoptosis was assessed by cleaved caspase-3 (Cl-Csp3) immunoblot and immunofluorescence microscopy. Primary neurons from p75(NTR-/-) mice or wild-type neurons were treated with propofol, with or without pretreatment with TAT-Pep5 (10 μM, 15 min), a specific p75(NTR) inhibitor. P75(NTR-/-) neurons were transfected for 72 h with a lentiviral vector containing the synapsin-driven p75(NTR) gene (Syn-p75(NTR)) or control vector (Syn-green fluorescent protein) before propofol. To confirm our in vitro findings, wild-type mice and p75(NTR-/-) mice (PND5) were pretreated with either TAT-Pep5 or TAT-ctrl followed by propofol for 6 h.

Results

Neurons exposed to propofol showed a significant increase in Cl-Csp3, an effect attenuated by TAT-Pep5 and hydroxyfasudil. Apoptosis was significantly attenuated in p75(NTR-/-) neurons. In p75(NTR-/-) neurons transfected with Syn-p75(NTR), propofol significantly increased Cl-Csp3 in comparison with Syn-green fluorescent protein-transfected p75(NTR-/-) neurons. Wild-type mice exposed to propofol exhibited increased Cl-Csp3 in the hippocampus, an effect attenuated by TAT-Pep5. By contrast, propofol did not induce apoptosis in p75(NTR-/-) mice.

Conclusion

These results demonstrate that propofol induces apoptosis in developing neurons in vivo and in vitro and implicate a role for p75(NTR) and the downstream effector RhoA kinase.

  • Isoflurane leads to reductions in the activity-dependent processing of brain-derived neurotrophic factor (proBDNF) to the survival-promoting mature form BDNF in immature brain

  • The authors hypothesized that this mechanism applies to other general anesthetics that reduce neuronal activity

  • Propofol induced apoptosis in immature mouse neurons both in vitro  and in vivo  through alterations in BDNF signaling

  • This provides a potential therapeutic target for prevention of developmental neurotoxicity

DURING synaptogenesis, on postnatal day 5–7, anesthetics lead to neurodegeneration.1,,3Many anesthetics cause neurotoxicity, which include midazolam and nitrous oxide, isoflurane, sevoflurane, propofol, thiopental, and ketamine.4,,11In addition, isoflurane does not induce neuronal apoptosis on postnatal day 15, but does alter synaptic plasticity; these changes persist for at least 4 weeks postexposure.12Of significant concern is that neonatal exposure to anesthetics results in neurocognitive and behavioral abnormalities during adolescence and adulthood.2,3,13,14Although the mechanism by which this toxicity occurs is not clear, γ-aminobutyric acid (GABAA) agonism and N-methyl- D-aspartate receptor antagonism play a central role.

Recently we demonstrated that proBDNF-p75NTRsignaling mediates isoflurane neurotoxicity in developing neurons in vivo  and in vitro . Brain-derived neurotrophic factor (BDNF) is important to both prosurvival and proapoptotic signaling pathways. BDNF is stored as a proneurotrophin (proBDNF) within synaptic vesicles and is proteolytically cleaved to mature BDNF (mBDNF) in the synaptic cleft by plasmin, a protease activated by tissue plasminogen activator (tPA).15,,18Prosurvival signaling is triggered by mBDNF agonism of tropomyosin receptor kinase B, which leads to neurite outgrowth and synapse maturation and stabilization.15,17,19In contrast, noncleaved proBDNF binds to the p75 neurotrophin receptor (p75NTR) and activates RhoA, a small GTPase that regulates actin cytoskeleton polymerization resulting in inhibition of axonal elongation, growth cone collapse, and apoptosis.16,20,,22Neuronal stimulation is important in this process because proBDNF is constitutively secreted while tPA release is regulated; without neuronal depolarization, conversion of proBDNF to mBDNF may be blunted, which then leads to preferential signaling through p75NTR. Upon neuronal excitation, tPA release results in plasmin production and subsequent generation of mBDNF-tropomyosin receptor kinase B activation, leading to neuronal survival, neurite sprouting, and synaptogenesis.15,,17,22 

Head et al.  showed that isoflurane induces apoptosis in DIV5 neurons, a finding that did not occur in DIV14 or DIV21 neurons.22This effect was attenuated by TAT-Pep5, a p75NTRintracellular domain inhibitor, suggesting a role for p75NTRin isoflurane-mediated neurotoxicity. Apoptosis was also attenuated by pretreatment with tPA or plasmin, suggesting that isoflurane-mediated neurotoxicity is caused in part by suppressing tPA release and preventing proBDNF conversion to mBDNF, thus leading to preferential p75NTRsignaling, RhoA activation, actin cytoskeleton depolymerization, and subsequent neuronal apoptosis. Lemkuil et al.  extended these findings by showing that isoflurane exposure increases p75NTR-RhoA activation in parallel with apoptosis, and that inhibition of RhoA activation or cytoskeleton stabilization attenuates the isoflurane-mediated neurotoxic effects.23 

Although these data support the premise that proBDNF-p75NTRsignaling plays a significant role in neonatal neurotoxicity, a number of questions remain. If this mechanism is important to anesthetic neurotoxicity, then it should also be involved in toxicity mediated by other anesthetics that activate GABAAreceptor (e.g. , propofol). In the studies of Head et al.  and Lemkuil et al. , p75NTRinhibition was achieved pharmacologically. In addition to concerns about nonspecific effects of pharmacologic agents, complete suppression of RhoA activation was not achieved; residual RhoA activity may have influenced the results.24To address these concerns, we investigated the role of p75NTR-RhoA- RhoA kinase (ROCK) pathway in propofol-induced neurotoxicity.

Preparation of Neuronal Cell Cultures

All studies performed on animals were approved by Veteran Affairs San Diego Institutional Animal Care and Use Committee (San Diego, California) and conform to the guidelines of Public Health Service Policy on Human Care and Use of Laboratory Animals.

Neonatal mouse neurons (BALB/c; The Jackson Laboratory, Bar Harbor, ME) were isolated using a papain dissociation kit (Worthington Biochemical, Lakewood, NJ) as previously described.22p75NTRknockout BALBc mice were provided by Don Pizzo, Ph.D. (Project Scientist, University of California, San Diego, Department of Pathology), at the Veterans Affairs San Diego Healthcare System (originally generated by Dr. Kuo-Fen Lee25). Neurons were isolated from 1- or 2-day-old pups (postnatal day 1 or 2) and grown in culture for 4 to 7 days in vitro . Neurons were cultured in Neuobasal A media supplemented with B27 (2%), 250 mM GLUTMax1, and penicillin/streptomycin (1%). Neurons were cultured on poly-D-lysine/laminin (2 g/cm2) coated plates or coverslips at 37°C in 5% CO2for 4–7 days before experiments. Lentiviral (LV) vectors driven by neuronal specific synapsin promoters expressing p75NTRor green fluorescent protein (GFP) (LV-syn-p75NTRand LV-syn-GFP, respectively) were generated by and obtained from Atsushi Miyanohara, Ph.D. (Assistant Professor, Gene Therapy Program, University of California, San Diego, La Jolla, California), at the University of California, San Diego Viral Vector Core. Cleaved-caspase 3 (Cl-Csp3) (Cell Signaling, Danvers, MA) and drebrin (Abcam, Cambridge, MA) were used to detect apoptosis and the F-actin cytoskeleton, respectively, via  immunoblot or immunofluorescence deconvolution or confocal microscopy. Cl-Csp3 and drebrin were normalized to the nuclear stain 4′,6-diamidino-2-phenylindole (Molecular Probes/Invitrogen, Carlsbad, CA). Cl-Csp3 immunoblots were quantified by densitometry and normalized to total caspase-3 (T-Csp3). p75NTRantibody was obtained from Abcam. The cell-permeable peptide, TAT-Pep5 [H-YGRKKRRQRRR-CFFRGGFFNHNPRYC-OH], which blocks the intracellular association of p75NTRwith RhoGDI, thus blocking its ability to activate RhoA and hydroxyfasudil (HA 1100 hydrochloride), were purchased from CalBiochem (Gibbstown, NJ) and Tocris (Ellsville MO), respectively.

Anesthetic Neurotoxicity Model

In vitro , primary neuronal cultures were placed within an incubator and exposed to propofol 3 μM for 6 h in a gas mixture of 5% CO2, 21% O2, balance nitrogen at a flow rate of 2 l/min. The temperature in the incubator was maintained at 37°C. Neurons were harvested for analysis 2 h postexposure. In vivo , neonatal mice on postnatal day 5 were given a single intraperitoneal injection of propofol (100 mg/kg). Mice were sacrificed 6 h postexposure and brains were prepared by perfusion fixation for immunofluorescence confocal microscopy. Mice were administered TAT-Pep5 intraperitoneal (10 μM) 15 min before propofol injections. All mice were kept in room air and on a warming pad maintained at 37°C.

Lentiviral Vector Transfection

Primary neuronal cultures were transfected with a lentiviral vector that expresses p75NTRdriven by a neuronal specific synapsin-promoter (LV-syn-p75NTR) for 72 h, and then exposed to propofol. A lentiviral vector that expresses GFP driven by a neuronal specific synapsin-promoter (LV-syn-GFP) served as control.

Protein Extraction and Western Blot Analysis

Proteins in cell lysates were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis using 10% acrylamide gels (Invitrogen, Carlsbad, CA) and transferred to polyvinylidene difluoride membranes (Millipore, Billerica, MA) by electroelution. Membranes were blocked in 20 mM phosphate-buffered saline Tween (1%) containing 4% bovine serum albumin and incubated with primary antibody overnight at 4°C as previously described.22,23Primary antibodies were visualized using secondary antibodies conjugated to horseradish peroxidase (Santa Cruz Biotech, Santa Cruz, CA) and chemo luminescent reagent (Amersham Pharmacia Biotech, Piscataway, NJ). All displayed bands are expected to migrate to the appropriate size and were determined by comparison with molecular weight standards. We performed an oversaturation analysis with the UVP Imaging software. Red pixilation is assigned to the bands as an indicator of oversaturation. Image J (National Institutes of Health, Bethesda, MD) was used for densitometric analysis of immunoblots with normalization of cleaved caspase-3 to total caspase-3.

Immunofluorescence Confocal Microscopy

Neurons were prepared for immunofluorescence microscopy as previously described.22,23Primary neurons were fixed with 4% paraformaldehyde in phosphate-buffered saline for 10 min at room temperature, incubated with 100 mM glycine (pH 7.4) for 10 min to quench aldehyde groups, permeabilized in buffered Triton X-100 (0.1%) for 10 min, blocked with 1% bovine serum albumin/PBS/Tween (0.05%) for 20 min, and then incubated with primary antibodies in 1% bovine serum albumin/PBS/Tween (0.05%) for 24–48 h at 4°C. Excess antibody was removed by washing with PBS/Tween (0.1%) for 15 min followed by incubation with fluorescein isothiocyanate or Alexa-conjugated secondary antibody (1:250) for 1 h. To remove excess secondary antibody, tissue or cells were washed six times at 5-min intervals with PBS/Tween (0.1%) and incubated for 20 min with the nuclear stain 4′,6-diamidino-2-phenylindole (1:5,000) diluted in phosphate-buffered saline. Cells were washed for 10 min with phosphate-buffered saline and mounted in gelvatol for microscopic imaging. Confocal images were captured with an Olympus confocal microscope system (Applied Precision, Inc., Issaquah, WA.) that included a Photometrics CCD (Photometrics, Tucson, AZ) mounted on a Nikon TE-200 (Nikon, Melville, NY) inverted epi-fluorescence microscope. Between 30 and 80 optical sections spaced by approximately 0.1–0.3 μm were captured. Exposure times were set such that the camera response was in the linear range for each fluorophore. Maximal projection volume views or single optical sections were visualized. Pixels were assessed quantitatively by CoLocalizer Pro 1.0 software (Colocalization Research Software, Japan and Switzerland). Statistical analysis was performed using Prism 4 (GraphPad Software, La Jolla, CA).

Apoptosis Quantification

The cleaved caspase-3 pixels (red, Alexa 594) were normalized to nuclear stained pixels (blue, 405).22,23Cleaved caspase-3 is an executioner and marker of apoptosis. Ten visual fields at 40× magnification were counted per experimental condition.

Cytoskeletal Depolymerization Quantification

The drebrin pixels (green, Alexa 488) were normalized to nuclear stained pixels (blue, 405).22,23Drebrin is a filamentous F-actin binding protein that stabilizes the actin cytoskeleton within neuritic processes. Pixel values were obtained after subtracting background through normalized threshold values in Colocalizer Pro as previously described.26,27A reduction in neuritic processses is indicated by decreased drebrin protein expression. Sample size (n) equals number of neurons counted per experimental condition.

Statistical Analysis

All parametric data were analyzed by either two-tailed unpaired t  tests (fig. 1,,5) or by one-way ANOVA with Bonferroni correction (fig. 6). Significance was set at P < 0.05. Statistical analysis was performed using Prism 4 (GraphPad Software). Sample size (n) represents the amount of times the experiments were repeated on separate neuronal cell culture preparations derived from 12–20 postnatal day 1–3 pups.

Fig. 1. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were then exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Apoptosis was evaluated after propofol exposure by cleaved caspase-3 immunoblot. (A ) Immunoblot analysis shows an increase in apoptosis marker cleaved caspase-3, with propofol exposure. (B ) Quantitation of the data are represented in the graph (n = 3; #P = 0.002). Sample size is indicated above the error bars ± SEM. These data demonstrate that, in vitro , primary neurons from neonatal rodents exposed to propofol exhibit increased apoptosis. Cl-Csp3 = cleaved caspase-3; T-Csp3 = total caspase-3.

Fig. 1. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were then exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Apoptosis was evaluated after propofol exposure by cleaved caspase-3 immunoblot. (A ) Immunoblot analysis shows an increase in apoptosis marker cleaved caspase-3, with propofol exposure. (B ) Quantitation of the data are represented in the graph (n = 3; #P = 0.002). Sample size is indicated above the error bars ± SEM. These data demonstrate that, in vitro , primary neurons from neonatal rodents exposed to propofol exhibit increased apoptosis. Cl-Csp3 = cleaved caspase-3; T-Csp3 = total caspase-3.

Close modal

Propofol Exposure Increases Apoptosis in Primary Mouse Neurons (Days In Vitro  5–7)

Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Propofol (3 μM, 6 h) exposure resulted in a significantly increased (n = 3; P = 0.002) expression of Cl-Csp3 compared with control (fig. 1).

Propofol Exposure Decreases Neuritic Processes in Primary Mouse Neurons (Days In Vitro  5–7)

Based on our previous data on isoflurane-mediated neurotoxicity, a key mechanistic tenet of injury is preferential activation of p75NTRsignaling pathway leading to actin cytoskeleton destabilization and subsequent apoptosis.22,23Because we hypothesize that propofol mediates neuronal apoptosis through a similar mechanism to that of isoflurane, we investigated the effects of propofol exposure on formation of neuritic processes as measured by drebrin, a neuronal F-actin binding protein and marker of dendritic filopodial spines.22,23Primary neurons were isolated from neonatal rodent pup brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Propofol (3 μM, 6 h) exposure resulted in a significantly decreased (n = 5; P = 0.008) expression of drebrin compared with control (fig. 2).

Fig. 2. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. On day in vitro  7 neurons were exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Neuritic processes were evaluated after propofol exposure by drebrin immunofluorescence microscopy. Nucleus was stained with DAPI (4′,6-diamidino-2-phenylindole). (A ) Immunofluorescence microscopy shows a decrease in drebrin in neurons exposed to propofol versus  control. (B ) Quantitation of the data are represented in the graph (n = 5; #P = 0.008). Sample size is indicated above the error bars ± SEM. These data demonstrate that, in vitro , primary neurons from neonatal rodents exposed to propofol exhibit a reduction in neuritic processes. DAPI = 4′,6- diamidino-2-phenylindole.

Fig. 2. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. On day in vitro  7 neurons were exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Neuritic processes were evaluated after propofol exposure by drebrin immunofluorescence microscopy. Nucleus was stained with DAPI (4′,6-diamidino-2-phenylindole). (A ) Immunofluorescence microscopy shows a decrease in drebrin in neurons exposed to propofol versus  control. (B ) Quantitation of the data are represented in the graph (n = 5; #P = 0.008). Sample size is indicated above the error bars ± SEM. These data demonstrate that, in vitro , primary neurons from neonatal rodents exposed to propofol exhibit a reduction in neuritic processes. DAPI = 4′,6- diamidino-2-phenylindole.

Close modal

TAT-Pep5 Attenuates Propofol-induced Cleaved Caspase-3 Activation in Primary Mouse Neurons (Days In Vitro  5–7)

Previous work from our laboratory has demonstrated that isoflurane-mediated neurotoxicity in developing primary neurons (days in vitro  5–7) is mediated through neuronal suppression and subsequent proBDNF activation of p75NTR.22,23Moreover, using TAT-Pep5, we showed that signaling from p75NTRto ρ was mediating by isoflurane neurotoxicity.23We tested whether propofol exposure to primary neurons (days in vitro  5–7) also causes cellular injury in a pattern similar to isoflurane (i.e. , propofol induces neuronal apoptosis through p75NTRactivation). TAT-Pep5 (10 μM, 15 min) treatment of primary mouse neurons (days in vitro  5–7) before propofol exposure (3 μM, 6 h) significantly attenuated Cl-Csp3 expression (n = 3; P = 0.046) compared with propofol exposure without TAT-Pep5 pretreatment (fig. 3).

Fig. 3. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were pretreated with TAT-Pep5 (10 μM; 15 min), a p75NTRintracellular domain antagonist, before propofol exposure. After pretreatment, neurons were exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Apoptosis was evaluated after propofol exposure by cleaved caspase-3 immunoblot. (A ) Immunoblot analysis shows a decrease in apoptosis marker, cleaved caspase-3, with TAT-Pep5 pretreatment. (B ) Quantitation of the data are represented in the graph (n = 3; #P = 0.046). Sample size is indicated above the error bars ± SEM. Cl-Csp3 = cleaved caspase-3; T-Csp3 = total caspase-3.

Fig. 3. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were pretreated with TAT-Pep5 (10 μM; 15 min), a p75NTRintracellular domain antagonist, before propofol exposure. After pretreatment, neurons were exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Apoptosis was evaluated after propofol exposure by cleaved caspase-3 immunoblot. (A ) Immunoblot analysis shows a decrease in apoptosis marker, cleaved caspase-3, with TAT-Pep5 pretreatment. (B ) Quantitation of the data are represented in the graph (n = 3; #P = 0.046). Sample size is indicated above the error bars ± SEM. Cl-Csp3 = cleaved caspase-3; T-Csp3 = total caspase-3.

Close modal

Hydroxyfasudil Attenuates Propofol-induced Cleaved Caspase-3 Activation in Primary Mouse Neurons (Days In Vitro  5–7)

RhoA is known to regulate actin cytoskeleton dynamics in neurons and cause growth cone collapse, leading to apoptosis.28,,30RhoA is activated by p75NTRand mediates its effects through downstream activation of ROCK.31Because ROCK is indirectly activated by p75NTRand inhibition of p75NTRwith TAT-Pep5 attenuates propofol-mediated apoptosis in developing neurons (days in vitro  5–7), we hypothesized that inhibition of ROCK would attenuate propofol-mediated apoptosis. Primary mouse neurons (days in vitro  5–7) were pretreated with a ROCK inhibitor, hydroxyfasudil (10 μM, 15 min), before propofol exposure (3 μM, 6 h); hydroxyfasudil significantly attenuated Cl-Csp3 activation (n = 3; P = 0.007) compared with propofol exposure (with hydroxyfasudil vehicle) in the absence of hydroxyfasudil pretreatment (fig. 4).

Fig. 4. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were pretreated with hydroxyfasudil (10 μM; 15 min), a ρ kinase (ROCK) inhibitor, before propofol exposure. After pretreatment, neurons were exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Apoptosis was evaluated after propofol exposure by cleaved caspase-3 immunoblot. (A ) Immunoblot analysis shows a decrease in apoptosis marker, cleaved caspase-3, with hydroxyfasudil pretreatment. (B ) Quantitation of the data are represented in the graph (n = 3; #P = 0.007). Sample size is indicated above the error bars ± SEM. Cl-Csp3 = cleaved caspase-3; ROCK inh = hydroxyfasudil; T-Csp3 = total caspase-3.

Fig. 4. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were pretreated with hydroxyfasudil (10 μM; 15 min), a ρ kinase (ROCK) inhibitor, before propofol exposure. After pretreatment, neurons were exposed to propofol 3.0 μM for 6 h in 5% CO2in air. Apoptosis was evaluated after propofol exposure by cleaved caspase-3 immunoblot. (A ) Immunoblot analysis shows a decrease in apoptosis marker, cleaved caspase-3, with hydroxyfasudil pretreatment. (B ) Quantitation of the data are represented in the graph (n = 3; #P = 0.007). Sample size is indicated above the error bars ± SEM. Cl-Csp3 = cleaved caspase-3; ROCK inh = hydroxyfasudil; T-Csp3 = total caspase-3.

Close modal

Primary Neurons Transfected with a Lentiviral Vector that Expresses p75NTRDriven by a Neuronal-specific Synapsin Promoter (LV-syn-p75NTR) Increases p75NTRExpression

Because our results from figures 3and 4demonstrate that inhibition of p75NTRsignaling attenuates propofol-mediated apoptosis in developing primary neurons, we hypothesized that primary neurons from p75NTRknockout (p75NTR−/−) mice would be less susceptible to propofol-mediated apoptosis, and that reexpression of p75NTRin p75NTR−/−neurons would reestablish anesthetic vulnerability. Primary neurons were isolated from p75NTR−/−mice at postnatal day 1–3 and grown in vitro  for 5–7 days. On day 4 in vitro , neurons were transfected with a lentiviral vector that expresses p75NTRdriven by a neuron-specific synapsin promoter (LV-syn-p75NTR) or GFP control vector (LV-syn-GFP) for 72 h. Immunoblot analysis demonstrated a dose-dependent increase in p75NTRprotein expression after transfection with LV-syn-p75NTR(figs. 5A and B). On day 7 in vitro , neurons were exposed to propofol (3 μM, 6 h) and subjected to immunofluroscence confocal microscopy. Fixed neurons were incubated with antibodies to drebrin, and Cl-Csp3, and the nuclear marker 4′,6-diamidino-2-phenylindole. Propofol administration to p75NTR−/−neurons (as confirmed by polymerase chain reaction, fig. 5C) on day 7 in vitro  transfected with LV-syn-GFP did not result in neuronal apoptosis as indicated by Cl-Csp3 immunofluorescence (fig. 5Di), suggesting that p75NTRis necessary for propofol-induced neurotoxicity. By contrast, propofol administration to p75NTR−/−neurons on day 7 in vitro  transfected with LV-syn-p75NTRsignificantly increased (n = 10; P = 0.0002) levels of Cl-Csp3 (fig. 5Dii).

Fig. 5. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were then transfected for 72 h with increasing doses of a lentiviral (LV) vector that expresses p75NTRdriven by a neuronal-specific synapsin promoter (LV-syn-p75NTR). (A ) Immunoblot analysis shows an increase in p75NTRexpression with increasing doses of LV-syn-p75NTR. (B ) Quantitation of the data are represented in the graph. These data demonstrate that, in vitro , primary neurons from neonatal rodents transfected for 72 h with LV-syn-p75NTRexhibits a dose-dependent increase in p75NTRexpression. (C ) Polymerase chain reactor confirmed that rodents used for primary neuronal cultures were p75NTRknockout genotype (-/-). On day in vitro  4, p75NTRknockout neurons were transfected with LV-syn-p75NTRfor 72 h. LV-syn-GFP served as a control. On day in vitro  7 neurons were exposed to propofol 3.0 μM for 6 h and apoptosis was evaluated by cleaved caspase-3 immunofluorescence microscopy. (D ) Immunofluorescence microscopy shows an increase in cleaved caspase-3 in p75NTRknockout neurons transfected with LV-syn-p75NTRand exposed to propofol versus  LV-syn-GFP transfected neurons exposed to propofol. (E ) Quantitation of the data are represented in the graph (n = 10; P = 0.0002). Sample size is indicated above the error bars ± SEM. These data demonstrate that, in vitro , reexpression of p75NTRin p75NTRknockout neurons reestablishes propofol-mediated apoptosis. Cl-Csp3 = cleaved caspase-3; DAPI = 4′,6-diamidino-2-phenylindole; GAPDH = glyceraldehyde 3-phosphate dehydrogenase; GFP = green fluorescent protein; LV-syn-p75NTR= a neuronal-specific synapsin promoter.

Fig. 5. Primary neurons were isolated from neonatal rodent brains at postnatal day 1–3 and grown in vitro  for 5–7 days. Neurons were then transfected for 72 h with increasing doses of a lentiviral (LV) vector that expresses p75NTRdriven by a neuronal-specific synapsin promoter (LV-syn-p75NTR). (A ) Immunoblot analysis shows an increase in p75NTRexpression with increasing doses of LV-syn-p75NTR. (B ) Quantitation of the data are represented in the graph. These data demonstrate that, in vitro , primary neurons from neonatal rodents transfected for 72 h with LV-syn-p75NTRexhibits a dose-dependent increase in p75NTRexpression. (C ) Polymerase chain reactor confirmed that rodents used for primary neuronal cultures were p75NTRknockout genotype (-/-). On day in vitro  4, p75NTRknockout neurons were transfected with LV-syn-p75NTRfor 72 h. LV-syn-GFP served as a control. On day in vitro  7 neurons were exposed to propofol 3.0 μM for 6 h and apoptosis was evaluated by cleaved caspase-3 immunofluorescence microscopy. (D ) Immunofluorescence microscopy shows an increase in cleaved caspase-3 in p75NTRknockout neurons transfected with LV-syn-p75NTRand exposed to propofol versus  LV-syn-GFP transfected neurons exposed to propofol. (E ) Quantitation of the data are represented in the graph (n = 10; P = 0.0002). Sample size is indicated above the error bars ± SEM. These data demonstrate that, in vitro , reexpression of p75NTRin p75NTRknockout neurons reestablishes propofol-mediated apoptosis. Cl-Csp3 = cleaved caspase-3; DAPI = 4′,6-diamidino-2-phenylindole; GAPDH = glyceraldehyde 3-phosphate dehydrogenase; GFP = green fluorescent protein; LV-syn-p75NTR= a neuronal-specific synapsin promoter.

Close modal

TAT-Pep5 Attenuates Propofol-mediated Apoptosis in the Hippocampus of Wild-type Mice In Vivo 

To further confirm our in vitro  findings, wild type mice were pretreated with either TAT-Pep5 (10 μM) or TAT-ctrl (10 μM) intraperitoneal followed by exposure to propofol with for 6 h in vivo . In addition, p75NTR−/−mice were exposed to propofol for 6 h. Wild-type mice exhibited a significant increase in Cl-Csp3 in the dentate gyrus (P < 0.0001), CA3 (P < 0.0001), and CA1 (P < 0.001) regions of the hippocampus, compared with intralipid-treated controls (n = 4 or 5, fig. 6A). TAT-Pep5 significantly attenuated propofol-mediated apoptosis (P < 0.001, dentate gyrus; P < 0.001, CA3; P < 0.001, dentate gyrus), suggesting that activation of ρ by p75NTRis involved in propofol neurotoxicity. There was no observed apoptosis in propofol-treated p75NTR−/−mice (n = 4, fig. 6B), suggesting that p75NTRexpression mediates propofol-mediated neuronal cell death in vivo .

Fig. 6. Postnatal day 5 wild-type or p75NTRknockout mice were given an intraperitoneal (intraperitoneal) injection of propofol (100 mg/kg) or intralipid for 6 h and apoptosis was evaluated by cleaved caspase-3 immunofluorescence. (A ) Dentate gyrus, CA3, and CA1 regions of the hippocampus are indicated on image. Basal TAT-CTRL, Basal TAT-Pep5, propofol TAT-CTRL, and propofol TAT-Pep5 are indicated on the image. Immunofluorescence microscopic analysis shows that wild-type mice exhibited a significant increase in cleaved caspase-3 in the dentate gyrus (*P = 0.002), CA3 (*P = 0.008), and CA1 (*P = 0.007) regions of the hippocampus compared with intralipid-treated controls (n = 4 or 5). TAT-Pep5 (10 μM, 15 min) significantly attenuated propofol-mediated apoptosis (*P = 0.005, CA1; *P = 0.002, CA3; *P = 0.004, dentate gyrus). (B ) Additional immunofluorescence microscopic analysis shows that p75NTRknockout mice exhibited no increase in cleaved caspase-3 in the dentate gyrus, CA3, or CA1 following propofol exposure compared to wild-type (#P = 0.0008, dentate gyrus; #P = 0.03, CA3; #P = 0.002, CA1). Quantitation of the data are represented in the graphs. Sample size is indicated above the error bars ± SEM. Scale bar = 20 μm. Cl-Csp3 = cleaved caspase-3; DAPI = 4′,6-diamidino-2-phenylindole; DG = dentate gyrus; KO = knockout.

Fig. 6. Postnatal day 5 wild-type or p75NTRknockout mice were given an intraperitoneal (intraperitoneal) injection of propofol (100 mg/kg) or intralipid for 6 h and apoptosis was evaluated by cleaved caspase-3 immunofluorescence. (A ) Dentate gyrus, CA3, and CA1 regions of the hippocampus are indicated on image. Basal TAT-CTRL, Basal TAT-Pep5, propofol TAT-CTRL, and propofol TAT-Pep5 are indicated on the image. Immunofluorescence microscopic analysis shows that wild-type mice exhibited a significant increase in cleaved caspase-3 in the dentate gyrus (*P = 0.002), CA3 (*P = 0.008), and CA1 (*P = 0.007) regions of the hippocampus compared with intralipid-treated controls (n = 4 or 5). TAT-Pep5 (10 μM, 15 min) significantly attenuated propofol-mediated apoptosis (*P = 0.005, CA1; *P = 0.002, CA3; *P = 0.004, dentate gyrus). (B ) Additional immunofluorescence microscopic analysis shows that p75NTRknockout mice exhibited no increase in cleaved caspase-3 in the dentate gyrus, CA3, or CA1 following propofol exposure compared to wild-type (#P = 0.0008, dentate gyrus; #P = 0.03, CA3; #P = 0.002, CA1). Quantitation of the data are represented in the graphs. Sample size is indicated above the error bars ± SEM. Scale bar = 20 μm. Cl-Csp3 = cleaved caspase-3; DAPI = 4′,6-diamidino-2-phenylindole; DG = dentate gyrus; KO = knockout.

Close modal

Fig. 6. Postnatal day 5 wild-type or p75NTRknockout mice were given an intraperitoneal (intraperitoneal) injection of propofol (100 mg/kg) or intralipid for 6 h and apoptosis was evaluated by cleaved caspase-3 immunofluorescence. (A ) Dentate gyrus, CA3, and CA1 regions of the hippocampus are indicated on image. Basal TAT-CTRL, Basal TAT-Pep5, propofol TAT-CTRL, and propofol TAT-Pep5 are indicated on the image. Immunofluorescence microscopic analysis shows that wild-type mice exhibited a significant increase in cleaved caspase-3 in the dentate gyrus (*P = 0.002), CA3 (*P = 0.008), and CA1 (*P = 0.007) regions of the hippocampus compared with intralipid-treated controls (n = 4 or 5). TAT-Pep5 (10 μM, 15 min) significantly attenuated propofol-mediated apoptosis (*P = 0.005, CA1; *P = 0.002, CA3; *P = 0.004, dentate gyrus). (B ) Additional immunofluorescence microscopic analysis shows that p75NTRknockout mice exhibited no increase in cleaved caspase-3 in the dentate gyrus, CA3, or CA1 following propofol exposure compared to wild-type (#P = 0.0008, dentate gyrus; #P = 0.03, CA3; #P = 0.002, CA1). Quantitation of the data are represented in the graphs. Sample size is indicated above the error bars ± SEM. Scale bar = 20 μm. Cl-Csp3 = cleaved caspase-3; DAPI = 4′,6-diamidino-2-phenylindole; DG = dentate gyrus; KO = knockout.

Fig. 6. Postnatal day 5 wild-type or p75NTRknockout mice were given an intraperitoneal (intraperitoneal) injection of propofol (100 mg/kg) or intralipid for 6 h and apoptosis was evaluated by cleaved caspase-3 immunofluorescence. (A ) Dentate gyrus, CA3, and CA1 regions of the hippocampus are indicated on image. Basal TAT-CTRL, Basal TAT-Pep5, propofol TAT-CTRL, and propofol TAT-Pep5 are indicated on the image. Immunofluorescence microscopic analysis shows that wild-type mice exhibited a significant increase in cleaved caspase-3 in the dentate gyrus (*P = 0.002), CA3 (*P = 0.008), and CA1 (*P = 0.007) regions of the hippocampus compared with intralipid-treated controls (n = 4 or 5). TAT-Pep5 (10 μM, 15 min) significantly attenuated propofol-mediated apoptosis (*P = 0.005, CA1; *P = 0.002, CA3; *P = 0.004, dentate gyrus). (B ) Additional immunofluorescence microscopic analysis shows that p75NTRknockout mice exhibited no increase in cleaved caspase-3 in the dentate gyrus, CA3, or CA1 following propofol exposure compared to wild-type (#P = 0.0008, dentate gyrus; #P = 0.03, CA3; #P = 0.002, CA1). Quantitation of the data are represented in the graphs. Sample size is indicated above the error bars ± SEM. Scale bar = 20 μm. Cl-Csp3 = cleaved caspase-3; DAPI = 4′,6-diamidino-2-phenylindole; DG = dentate gyrus; KO = knockout.

Close modal

Anesthetic agents inhibit neuronal activity in part by potentiating GABAAreceptors, inhibiting N-methyl-D-aspartate channels, or activating two-pore potassium channels.32We have recently shown that reduced neuronal activity during the critical period of synaptogenesis leads to neuronal apoptosis by preferential signaling of proBDNF via  p75NTR.22,23The mechanism by which p75NTRactivation leads to neurodegeneration is mediated in part through RhoA activation, actin cytoskeleton destabilization, and subsequent apoptosis.23Specifically, these findings pertained to studies conducted with isoflurane. Previous work has shown that propofol exposure during the neonatal period leads to a similar pattern of injury as seen with isoflurane.33Because propofol and isoflurane depress neuronal activity and are GABAAreceptor agonists, the present study was conducted to determine whether the mechanisms involved in isoflurane-mediated neurotoxicty are also the key mediators in propofol-induced neuronal cell death. The present study is the first to demonstrate that propofol exposure to developing neurons induces apoptosis through the p75NTR-RhoA-ROCK pathway.

Exposure of developing neurons (days in vitro  5–7) to propofol results in decreased dendritic spines and increased apoptosis, and both these effects were attenuated by either p75NTRor ROCK inhibition. Apoptosis was not observed in p75NTR−/−neurons in vitro  or p75NTR−/−mice in vivo . Moreover, reexpression of p75NTRin p75NTR−/−neurons using a neuron-specific promoter (synapsin) reestablished the neurotoxic effects from propofol. These results strongly suggest a role for p75NTRin mediating anesthetic-induced neuronal apoptosis. While our data are in support of proBDNF-p75NTRsignaling being involved in propofol-mediated neurotoxicity, it still remains to be answered whether this is the sole or dominant mechanism. Other studies have reported that GABAAactivation contributes to anesthetic mediated neurotoxicity in immature neurons, and that this toxicity might be mediated by GABA mediated excitation in neonatal neurons.34,35However, our results show that propofol exposure does not induce apoptosis in p75NTR−/−neurons even though GABAAactivation would be expected. This suggests that GABAAsignaling may be disrupted in p75NTR−/−neurons, or that GABAAactivation per se  does not play a major role in propofol-mediated neurotoxicity. Although we did not directly measure GABAAactivation, unpublished findings from our group show GABA protein expression in p75NTR−/−is similar to wild-type, and that onset of, recovery from, and sensitivity to propofol anesthesia in p75NTR−/−mice pups parallel that in wild-type mice pups. Despite clinical evidence of deep anesthesia, neuronal apoptosis was not observed in the p75NTR−/−. Other work from our laboratory demonstrated that treatment of day in vitro  4 neurons with 4-aminopyridine, an agent that induces synaptic release of neurotransmitters, prevented anesthetic-mediated neurotoxicity. Given that propofol and isoflurane are modulators of the GABA channel and increase Clcurrent significantly only in the presence of GABA, one would have expected an increase in neurotoxicity with synaptic release of GABA. To the contrary, in our in vitro  model, 4-aminopyridine reduced death in neurons exposed to anesthetic. In addition, in our original paper in which we put forward the proBDNF-p75NTRhypothesis, we measured tPA levels with and without anesthetic administration in vitro  and found that tPA levels were substantially reduced by isoflurane. If GABA-mediated excitation had occurred, we would have expected increased tPA levels; what we observed was in fact the opposite. In aggregate, it is our belief that our data support the premise that a reduction in neuronal activity is one of the causes of anesthetic neurotoxicity; confirmation of this awaits results of electrophysiologic studies. Therefore, a reasonable argument can be made that in our in vitro  and in vivo  model system, propofol-mediated neuronal death does not appear to be primarily mediated by GABAAactivation but rather though activation of p75NTR.

Anesthetic agents have pleiotrophic effects and it is therefore unsurprising that a number of other mechanisms by which they induce toxicity may be operative. Straiko et al.  have shown that treatment of postnatal day 5 mice with lithium counteracted propofol- and ketamine-mediated suppression of extracellular-regulated kinase phosphorylation and subsequent neurapoptosis.36Lithium has also been shown to be an inhibitor of GSK3β37; given the salutary effect of lithium, it is possible that GSK3β may also contribute to anesthetic neurotoxicity. Work by Wang et al.  showed that antisense knockdown of N-methyl-D-aspartate receptor subunit NR1 or NR2A, but not NR2B, reduced phencyclidine-induced neurapoptosis.38These authors suggested that blockade of N-methyl-D-aspartate receptors leads to an upregulation of N-methyl-D-aspartate receptor in neurons. These neurons may subsequently be more vulnerable to excitotoxic injury. A more recent finding demonstrated that ketamine exposure results in aberrant cell cycle reentry of neurons and subsequent apoptosis in the developing rat brain.39What remains to be defined is whether anesthetic neurotoxicity is a function of a dominant mechanism or a combination of mechanisms. In addition, the possibility that the underlying mechanisms of toxicity might be dependent upon the class of anesthetic agents (predominantly GABAAagonists or N-methyl-D-aspartate receptor antagonists) remains to be explored.

Actin cytoskeleton organization and dynamics play a critical role for development and maturation of neurons.40RhoA and its effector ρ-associated kinase, ROCK, are key mediators of actin rearrangement and formation. RhoA is a small GTPase that is activated by p75NTRsignaling; RhoA activation leads to actin cytoskeleton depolymerization and subsequent apoptosis.28,,31,41,,46The RhoA effects are through activation of ROCK, a serine/threonine kinase.47,48In the present study pretreatment with a RhoA-signaling pathway inhibitor before propofol exposure significantly attenuated apoptosis. This strongly supports proBDNF-p75NTRsignaling as a common mechanism of isoflurane and propofol-mediated neurotoxicity.22,23A limitation to the present study is that we have not investigated the effects of anesthetics on other small GTPases, such as Rac1. Rac1 is a small GTPase known to be promote dendritic spine development through activation of N-methyl-D-aspartate receptors.49,50Anesthetics antagonize N-methyl-D-aspartate receptors and therefore may promote actin cytoskeleton depolymerization through decreased Rac1 activity. This may contribute to the decrease in neuritic processes seen with propofol exposure in neonatal neurons (fig. 2), a notion worthy of further investigation. Although we investigated RhoA and actin depolymerization specifically in neurons, others have shown that anesthetics also affect RhoA/myosin light-chain-signaling cascade and lead to alterations in actin organization in astroglia, a finding that may shed light on how anesthetics affect the interplay between astroglia and neurons during development or following injury.51 

Collectively, these results demonstrate that propofol induces apoptosis in developing neurons during the critical period of synaptogenesis and mechanistically links activation of p75NTRand the downstream effector ROCK in this neurotoxic process. As such, the results provide a mechanistic framework upon which novel therapeutic approaches for the prevention of anesthetic neurotoxicity can be developed.

The authors thank the University of California, San Diego Cancer Center Digital Imaging Shared Resource, in particular James Feramisco, Ph.D. (Professor of Medicine, University of California, San Diego, La Jolla, California), and Kersi Pestonjamasp, Ph.D. (Junior Faculty, University of California, San Diego). For technical support, the authors also thank Michael Kidd, B.S. (Technician, University of California, San Diego), Ana Moreno, B.S. (Technician, University of California, San Diego), and Yue Hu (Technician). In some cases, three-dimensional perspective views were made at the VisLab in the San Diego Supercomputer Center using National Partnership for Advanced Computational Infrastructure Scalable Visualization Tools.

1.
Jevtovic-Todorovic V, Wozniak DF, Benshoff ND, Olney JW: A comparative evaluation of the neurotoxic properties of ketamine and nitrous oxide. Brain Res 2001; 895:264–7
2.
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
3.
Fredriksson A, Pontén E, Gordh T, Eriksson P: Neonatal exposure to a combination of N-methyl-D-aspartate and γ-aminobutyric acid type A receptor anesthetic agents potentiates apoptotic neurodegeneration and persistent rehavioral deficits. ANESTHESIOLOGY 2007; 107:427–36
4.
Honegger P, Matthieu JM: Selective toxicity of the general anesthetic propofol for GABAergic neurons in rat brain cell cultures. J Neurosci Res 1996; 45:631–6
5.
Olney JW, Ishimaru MJ, Bittigau P, Ikonomidou C: Ethanol-induced apoptotic neurodegeneration in the developing brain. Apoptosis 2000; 5:515–21
6.
Spahr-Schopfer I, Vutskits L, Toni N, Buchs PA, Parisi L, Muller D: Differential neurotoxic effects of propofol on dissociated cortical cells and organotypic hippocampal cultures. ANESTHESIOLOGY 2000; 92:1408–17
7.
Young C, Jevtovic-Todorovic V, Qin YQ, Tenkova T, Wang H, Labruyere J, Olney JW: Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the infant mouse brain. Br J Pharmacol 2005; 146:189–97
8.
Vutskits L, Gascon E, Tassonyi E, Kiss JZ: Effect of ketamine on dendritic arbor development and survival of immature GABAergic neurons in vitro . Toxicol Sci 2006; 91:540–9
9.
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
10.
Wang C, Sadovova N, Patterson TA, Zou X, Fu X, Hanig JP, Paule MG, Ali SF, Zhang X, Slikker W Jr: Protective effects of 7-nitroindazole on ketamine-induced neurotoxicity in rat forebrain culture. Neurotoxicology 2008; 29:613–20
11.
Shi Q, Guo L, Patterson TA, Dial S, Li Q, Sadovova N, Zhang X, Hanig JP, Paule MG, Slikker W Jr., Wang C: Gene expression profiling in the developing rat brain exposed to ketamine. Neuroscience 2010; 166:852–63
12.
Briner A, De Roo M, Dayer A, Muller D, Habre W, Vutskits L: Volatile anesthetics rapidly increase dendritic spine density in the rat medial prefrontal cortex during synaptogenesis. ANESTHESIOLOGY 2010; 112:546–56
13.
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
14.
Paule MG, Li M, Allen RR, Liu F, Zou X, Hotchkiss C, Hanig JP, Patterson TA, Slikker W Jr., Wang C: Ketamine anesthesia during the first week of life can cause long-lasting cognitive deficits in rhesus monkeys. Neurotoxicol Teratol 2011; 33:220–30
15.
Lee R, Kermani P, Teng KK, Hempstead BL: Regulation of cell survival by secreted proneurotrophins. Science 2001; 294:1945–8
16.
Lu B: Pro-region of neurotrophins: Role in synaptic modulation. Neuron 2003; 39:735–8
17.
Pang PT, Teng HK, Zaitsev E, Woo NT, Sakata K, Zhen S, Teng KK, Yung WH, Hempstead BL, Lu B: Cleavage of proBDNF by tPA/plasmin is essential for long-term hippocampal plasticity. Science 2004; 306:487–91
18.
Lu LX, Yon JH, Carter LB, Jevtovic-Todorovic V: General anesthesia activates BDNF-dependent neuroapoptosis in the developing rat brain. Apoptosis 2006; 11:1603–15
19.
Lu B, Pang PT, Woo NH: The yin and yang of neurotrophin action. Nat Rev Neurosci 2005; 6:603–14
20.
Dubreuil CI, Winton MJ, McKerracher L: Rho activation patterns after spinal cord injury and the role of activated Rho in apoptosis in the central nervous system. J Cell Biol 2003; 162:233–43
21.
Yamauchi J, Chan JR, Shooter EM: Neurotrophins regulate Schwann cell migration by activating divergent signaling pathways dependent on Rho GTPases. Proc Natl Acad Sci U S A 2004; 101:8774–9
22.
Head BP, Patel HH, Niesman IR, Drummond JC, Roth DM, Patel PM: Inhibition of p75 neurotrophin receptor attenuates isoflurane-mediated neuronal apoptosis in the neonatal central nervous system. ANESTHESIOLOGY 2009; 110:813–25
23.
Lemkuil BP, Head BP, Pearn ML, Patel HH, Drummond JC, Patel PM: Isoflurane neurotoxicity is mediated by p75NTR-RhoA activation and actin depolymerization. ANESTHESIOLOGY 2011; 114:49–57
24.
Morgan P, Sedensky M: A new phase in anesthetic-induced neurotoxicity research. ANESTHESIOLOGY 2011; 114:10–11
25.
Lee KF, Li E, Huber LJ, Landis SC, Sharpe AH, Chao MV, Jaenisch R: Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system. Cell 1992; 69:737–49
26.
Zinchuk O, Fukushima A, Hangstefer E: Dynamics of PAF-induced conjunctivitis reveals differential expression of PAF receptor by macrophages and eosinophils in the rat. Cell Tissue Res 2004; 317:265–77
27.
Head BP, Patel HH, Roth DM, Murray F, Swaney JS, Niesman IR, Farquhar MG, Insel PA: Microtubules and actin microfilaments regulate lipid raft/caveolae localization of adenylyl cyclase signaling components. J Biol Chem 2006; 281:26391–9
28.
Schubert V, Dotti CG: Transmitting on actin: Synaptic control of dendritic architecture. J Cell Sci 2007; 120:205–12
29.
Davies AM: Neurotrophins: Neurotrophic modulation of neurite growth. Curr Biol 2000; 10:R198–200
30.
Schmidt A, Hall A: Guanine nucleotide exchange factors for Rho GTPases: Turning on the switch. Genes Dev 2002; 16:1587–609
31.
Yamashita T, Tohyama M: The p75 receptor acts as a displacement factor that releases Rho from Rho-GDI. Nat Neurosci 2003; 6:461–7
32.
Franks NP, Lieb WR: Molecular and cellular mechanisms of general anaesthesia. Nature 1994; 367:607–14
33.
Cattano D, Young C, Straiko MM, Olney JW: Subanesthetic doses of propofol induce neuroapoptosis in the infant mouse brain. Anesth Analg 2008; 106:1712–4
34.
Ben-Ari Y: Excitatory actions of gaba during development: The nature of the nurture. Nat Rev Neurosci 2002; 3:728–39
35.
Kahraman S, Zup SL, McCarthy MM, Fiskum G: GABAergic mechanism of propofol toxicity in immature neurons. J Neurosurg Anesthesiol 2008; 20:233–40
36.
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
37.
Xu CM, Wang J, Wu P, Xue YX, Zhu WL, Li QQ, Zhai HF, Shi J, Lu L: Glycogen synthase kinase 3β in the nucleus accumbens core is critical for methamphetamine-induced behavioral sensitization. J Neurochem 2011; 118:126–39
38.
Wang C, Fridley J, Johnson KM: The role of NMDA receptor upregulation in phencyclidine-induced cortical apoptosis in organotypic culture. Biochem Pharmacol 2005; 69:1373–83
39.
Soriano SG, Liu Q, Li J, Liu JR, Han XH, Kanter JL, Bajic D, Ibla JC: Ketamine activates cell cycle signaling and apoptosis in the neonatal rat brain. ANESTHESIOLOGY 2010; 112:1155–63
40.
Sekino Y, Kojima N, Shirao T: Role of actin cytoskeleton in dendritic spine morphogenesis. Neurochem Int 2007; 51:92–104
41.
Yamashita T, Tucker KL, Barde YA: Neurotrophin binding to the p75 receptor modulates Rho activity and axonal outgrowth. Neuron 1999; 24:585–93
42.
Luo L: Rho GTPases in neuronal morphogenesis. Nat Rev Neurosci 2000; 1:173–80
43.
Yamashita T, Higuchi H, Tohyama M: The p75 receptor transduces the signal from myelin-associated glycoprotein to Rho. J Cell Biol 2002; 157:565–70
44.
Hable WE, Miller NR, Kropf DL: Polarity establishment requires dynamic actin in fucoid zygotes. Protoplasma 2003; 221:193–204
45.
Vallotton P, Gupton SL, Waterman-Storer CM, Danuser G: Simultaneous mapping of filamentous actin flow and turnover in migrating cells by quantitative fluorescent speckle microscopy. Proc Natl Acad Sci U S A 2004; 101:9660–5
46.
Ridley AJ: Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol 2006; 16:522–9
47.
Coleman ML, Olson MF: Rho GTPase signalling pathways in the morphological changes associated with apoptosis. Cell Death Differ 2002; 9:493–504
48.
Etienne-Manneville S, Hall A: Rho GTPases in cell biology. Nature 2002; 420:629–35
49.
Tolias KF, Bikoff JB, Burette A, Paradis S, Harrar D, Tavazoie S, Weinberg RJ, Greenberg ME: The Rac1-GEF Tiam1 couples the NMDA receptor to the activity-dependent development of dendritic arbors and spines. Neuron 2005; 45:525–38
50.
Tolias KF, Bikoff JB, Kane CG, Tolias CS, Hu L, Greenberg ME: The Rac1 guanine nucleotide exchange factor Tiam1 mediates EphB receptor-dependent dendritic spine development. Proc Natl Acad Sci U S A 2007; 104:7265–70
51.
Lunardi N, Hucklenbruch C, Latham JR, Scarpa J, Jevtovic-Todorovic V: Isoflurane impairs immature astroglia development in vitro : The role of actin cytoskeleton. J Neuropathol Exp Neurol 2011; 70:281–91