Open Access

Overexpression of phosphoprotein enriched in astrocytes 15 reverses the damage induced by propofol in hippocampal neurons

  • Authors:
    • Feng Xian
    • Qifang Li
    • Zuping Chen
  • View Affiliations

  • Published online on: June 21, 2019     https://doi.org/10.3892/mmr.2019.10412
  • Pages: 1583-1592
  • Copyright: © Xian et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Propofol is a general anesthetic used in surgical operations. Phosphoprotein enriched in astrocytes 15(PEA15) was initially identified in astrocytes. The present study examined the role of PEA15 in the damage induced by propofol in hippocampal neurons. A model of hippocampal neuron damage was established using 50 µmol/l propofol. Cell viability, proliferation and apoptosis of hippocampal neurons were tested by Cell Counting Kit‑8 and flow cytometry. Western blotting and reverse transcription‑quantitative polymerase chain reaction analysis were performed to measure the expression levels of PEA15, and additional factors involved in apoptosis or in the signaling pathway downstream of PEA15. The present results suggested that propofol significantly decreased PEA15 expression levels in hippocampal neurons. Furthermore, overexpression of PEA15 significantly increased the cell viability and cell proliferation of cells treated with propofol. Additionally, PEA15 overexpression decreased apoptosis, which was promoted by propofol. Treatment with propofol significantly decreased the protein expression levels of pro‑caspase‑3, B‑cell lymphoma-2, phosphorylated extracellular signal‑regulated kinases (ERK)1/2, ribosomal S6 kinase 2 (RSK2) and phosphorylated cAMP responsive element binding protein 1 (CREB1). However, propofol upregulated active caspase‑3 and Bax expression levels. Notably, PEA15 overexpression was able to reverse the effects of propofol. Collectively, overexpression of PEA15 was able to attenuate the neurotoxicity of propofol in rat hippocampal neurons by increasing proliferation and repressing apoptosis via upregulation of the ERK‑CREB‑RSK2 signaling pathway.

Introduction

Annually, in the USA, approximately six million children require general anesthesia (1,2). Furthermore, in China, the number of children requiring anesthetics is increasing (3). The human brain develops rapidly between the embryonic stage and 2 years after birth. During this period the number of neurons increases, and large quantities of axons, dendrites and synapses are formed (46). Previous clinical studies have demonstrated that anesthesia and surgery may lead to cognitive impairment (7,8). Therefore, decreasing the brain neuronal damage caused by narcotic drugs is becoming an increasingly popular research topic.

A number of previous studies identified that inhalable narcotic drugs, including midazolam, propofol, isoflurane and sevoflurane, are used in newborns (912). Propofol is a general anesthetic used in surgical operations. Previous studies demonstrated that propofol may induce neurotoxicity in the brains of newborn animals (1316). In addition, previous studies identified that propofol-induced apoptosis in numerous types of cells associated with neurons (1719). Therefore, it is crucial to identify novel effective strategies to decrease the neurotoxicity of propofol in the developing brain.

Phosphoprotein enriched in astrocytes 15 (PEA15) is a phosphoprotein that was initially identified in astrocytes (20). PEA15 is known to mediate the signal transduction pathway; PEA15 serves as a signal connection point, thus possessing the potential to directly regulate cell behavior (21). Previous studies demonstrated that PEA15 serves roles in numerous processes, including tumor progression, diabetes, metabolic disorders and nervous system diseases (2226). However, at present, to the best of the authors' knowledge, the effect of PEA15 on propofol-induced hippocampal nerve injury has not been investigated.

In the present study, propofol was used to damage rat hippocampal neurons. Furthermore, the effect and potential molecular mechanism of PEA15 on propofol-induced hippocampal neuronal cell damage were examined by Cell Counting Kit-8 (CCK-8), flow cytometry, western blotting and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis.

Materials and methods

Animals

Pregnant Sprague-Dawley (SD) rats (3 months old; embryonic age E18; weight: 200–230 g) were purchased from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China). The rats were housed in cages at 22±1°C. The animals had free access to food and water, with a constant humidity (50±10%) and with a 12 h light/dark cycle. The animal experimental research was approved by the Ethics Committee of The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University (Changzhou, China).

Isolation of hippocampal neurons

The extraction and cultivation methods of the neurons were performed as previously described (27,28). In total, 5 newborn SD rats (3 male and 2 female; weight, 5.0–6.0 g) were sacrificed by cervical dislocation, within 24 h following birth. A solution of 75% ethanol was used to disinfect the fetal rats in sterile trays. Subsequently, the rats were guillotined. The brain was removed using bent tweezers and placed in a precooled Hank's balanced salt solution and the hippocampus was separated from the brain. Subsequently, the hippocampus was digested using 2% trypsin for 15 min at 37°C in a cell incubator with 5% CO2. The digestion was stopped by adding neurobasal medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (Cellbio, Shanghai, China). The hippocampal neurons were obtained and maintained in an incubator at 37°C with 5% CO2. The morphology of the cells was observed daily using a TMM-220 inverted light microscope (magnification, ×200; Shanghai Tianxing Instrument Co., Ltd., Shanghai, China).

Cell treatment and transfection

Propofol was purchased from Fresenius Kabi Austria GmbH (Graz, Austria). As previously described (29,30), the hippocampal neurons were exposed to 50 µmol/l propofol for 0, 6, 12 and 24 h. Subsequently, cells were transfected with 50 nM PEA15 cloned in pcDNA3.1 or empty pcDNA3.1 vectors (Invitrogen; Thermo Fisher Scientific, Inc.) using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) and cultured for 48 h at 37°C. A total of six experimental groups were considered in the present study: Control group (0.1% PBS), empty-vector group (transfected with pcDNA3.1 vector), PEA15 group (transfected with PEA15), propofol group (treated with 50 µmol/l propofol), empty-vector + propofol group (transfected with pcDNA3.1 empty vector and treated with 50 µmol/l propofol), and PEA15+propofol group (transfected with PEA15 and treated with 50 µmol/l propofol).

Cell viability assay

A CCK-8 (Beyotime Institute of Biotechnology, Haimen, China) assay was conducted to detect the cell viability. A total of 2×103 cells/ml in logarithmic phase were inoculated in 96-well plates and maintained in an incubator at 37°C with 5% CO2 for 24 h. Subsequently, cells were treated with propofol at 12.5, 25 and 50 µmol/l for 6 h at 37°C and divided into the aforementioned experimental groups, and transferred into the incubator for 6, 12 and 24 h. CCK-8 reagent was added into each well. Cells were cultured in the incubator at 37°C with 5% CO2 for 2.5 h. An HBS-1096A microplate reader (Nanjing Detie Experimental Equipment Co., Ltd., Nanjing, China) was used to measure the absorbance at 450 nm.

Flow cytometry assay

Cell apoptosis was determined using an Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptosis detection kit [Hangzhou Multi Sciences (Lianke) Biotech Co., Ltd., Hangzhou, China]. The cells were digested with 0.25% trypsin (Beyotime Institute of Biotechnology). The supernatant was removed and cells were resuspended in an incubation buffer at a density of 2×105 cells/ml. Annexin V-FITC and PI were added to the cell suspension in the dark at room temperature for 20 min. The CytoFLEX flow cytometer (Beckman Coulter, Inc., Brea, CA, USA) was used to detect cell apoptosis. Cell proliferation was assessed using the CellTrace™ carboxyfluorescein succinimidyl ester (CFSE) cell proliferation kit (Invitrogen; Thermo Fisher Scientific, Inc.). The cells were digested with 0.25% trypsin (Beyotime Institute of Biotechnology). The supernatant was removed and cells were resuspended in an incubation buffer at a density of 2×105 cells/ml. The CFSE solution was added to the cell suspension and incubated for 15 min at room temperature. A CytoFLEX flow cytometer (CytExpert software v1.2; Beckman Coulter, Inc.) was used to measure the cell proliferation.

Western blotting

Subsequent to being treated, cells were lysed with radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology). The protein content was analyzed using the Bradford method. A 10% SDS-PAGE was used to separate (20 µg/lane) proteins, that were transferred onto a polyvinylidene difluoride (PVDF) membrane. The PVDF membrane was sealed and blocked using 5% non-fat milk at 37°C for 1.5 h. Subsequently, the PVDF membrane was incubated with primary antibodies at 4°C for 24 h. The following antibodies were used in the present study: Anti-PEA15 [1:800; Cell Signaling Technology (CST), Inc., Danvers, MA, USA; cat. no. 2780], anti-pro-caspase-3 (1:1,000; Abcam, Cambridge, UK; cat. no. ab32150), anti-active caspase-3 (1:600; Abcam; cat. no. ab13847), anti-B-cell lymphoma-2 (Bcl-2; 1:700; Abcam; cat. no. ab59348), anti-apoptosis regulator BAX (Bax; 1:1,000; Abcam; cat. no. ab32503), anti-phosphorylated (p)-extracellular signal-regulated kinases 1/2 (ERK1/2; 1:1,000; CST, Inc.; cat. no. 4370), anti-ERK1/2 (1:600; CST, Inc.; cat. no. 9102), anti-ribosomal S6 kinase 2 (RSK2; 1:500; Abcam; cat. no. ab32133), anti-p-cAMP-response element binding protein 1 (CREB1; 1:800; Abcam; cat. no. ab32096), anti-CREB1 (1:700; Abcam; cat. no. ab32515) and anti-β-actin (1:5,000; Abcam; cat. no. ab179467). The PVDF membrane was subsequently incubated with Horseradish peroxidase-conjugated secondary antibodies (goat anti-rabbit; cat. no. ab205718; 1:2,000) at room temperature for 1 h. The proteins were detected using enhanced chemiluminescent reagents (Thermo Fisher Scientific, Inc.). The density of bands was determined using the Quantity One software version 2.4 (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

RT-qPCR

Total RNA from the neurons was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). RNA was reverse transcribed to cDNA using an RT kit (HaiGene, Harbin, China) at 37°C for 15 min followed by 85°C for 5 sec. The ABI 7500 Thermocycler (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used to amplify the cDNA with AceQ qPCR SYBR-Green Master Mix (Vazyme, Piscataway, NJ, USA). qPCR conditions were set as follows: 10 min pretreatment at 95°C, 96°C for 15 sec, 63°C for 45 sec (35 cycles) and a final extension at 75°C for 10 min. β-actin was used as the reference gene. The primers used were purchased from Shanghai ShineGene Molecular Biotech, Inc. (Shanghai, China) and were the following: PEA15 (forward, 5′-CCTGACCAACAACATCACCC-3′ and reverse, 5′-GATCTTCAGCACACGGGTTC-3′), Bcl-2 (forward, 5′-GCCTTCTTTGAGTTCGGTGG-3′ and reverse), Bax (forward, 5′-GAGACACCTGAGCTGACCTT-3′ and reverse, 5′-CGTCTGCAAACATGTCAGCT-3′) and β-actin (forward, 5′-CAACATGGATGAGCGGAAGG-3′ and reverse, 5′-GCAGTGTAGCAGCATCGAAA-3′). The 2−ΔΔCq method was used to calculate gene expression levels (31).

Statistical analysis

SPSS (version 20; IBM Corp., Armonk, NY, USA) software was used for statistical analysis. The data are presented as the mean ± standard deviation. The experiments were independently repeated at least three times. The experimental data were analyzed by one-way analysis of variance with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Propofol inhibits the expression level of PEA15 in hippocampal neurons

Following hippocampal neuron isolation, the cells were cultured for 2 days. The neurons presented 2–3 neurites, and were flat and multilateral. Following 6 days of incubation, the number of neurites increased, and the cells interconnected with each other through the neurites. Notably, the majority of the cells aggregated and exhibited a visible halo surrounding the cells (Fig. 1A).

Cell viability decreased following treatment with propofol at 12.5, 25 and 50 µmol/l for 6 h. The decrease in cell viability was significant in the 50 µmol/l group (Fig. 1B). Therefore, 50 µmol/l propofol was selected as the working concentration. Western blotting and RT-qPCR assays were performed to examine the effects of propofol on the expression level of PEA15 in hippocampal neurons. The western blotting suggested that the protein expression level of PEA15 was decreased following treatment with propofol for 6, 12 and 24 h. Notably, the protein expression level of PEA15 was lower at 12 h compared with 24 h; however, the difference was not significant (Fig. 1C and D). Furthermore, propofol significantly decreased the mRNA expression level of PEA15 in a time-dependent manner (Fig. 1E).

Overexpression of PEA15 reverts the decreased viability of hippocampal neurons induced by propofol

The transfection efficiency of PEA15 in hippocampal neurons was examined by western blotting and RT-qPCR analysis. The western blotting results demonstrated that the protein expression level of PEA15 was significantly increased following transfection with PEA15 (Fig. 2A and B). Additionally, the mRNA expression level of PEA15 was consistent with the protein expression level (Fig. 2C).

CCK-8 was used to test the effects of PEA15 and propofol on the viability of hippocampal neurons. The results suggested that propofol significantly decreased cell viability, compared with the control and empty-vector groups. The cell viability was unaffected following transfection of PEA15 alone. However, the cell viability in the PEA15 + propofol group was significantly increased compared with the propofol and empty-vector + propofol groups (Fig. 2D).

Overexpression of PEA15 represses the propofol-induced apoptosis of hippocampal neurons

The flow cytometry results suggested that propofol significantly promoted cell apoptosis. However, PEA15 overexpression was able to decrease the apoptosis of cells treated with propofol compared with the propofol and empty-vector + propofol groups (Fig. 3).

Overexpression of PEA15 reverts the proliferation of hippocampal neurons inhibited by propofol

The cell proliferation assay suggested that propofol significantly decreased the proliferation of hippocampal neurons. In addition, transfection of PEA15 was sufficient to increase the cell proliferation of cells treated with propofol compared with the propofol and empty-vector + propofol groups (Fig. 4).

PEA15 and propofol regulate apoptosis-associated factors in hippocampal neurons

Western blotting and RT-qPCR analysis were performed to investigate the effects of PEA15 and propofol on apoptosis-associated factors. The western blotting results suggested that treatment with propofol promoted the protein expression levels of active caspase-3 and Bax (Fig. 5A and B), and led to downregulation of pro-caspase-3 and Bcl-2 protein expression levels (Fig. 5A and C). By analyzing the protein and the mRNA expression levels of Bax and Bcl-2, it was identified that the Bax/Bcl-2 ratio was significantly increased following treatment with propofol (Fig. 5D and E). Notably, PEA15 overexpression led to opposite effects.

PEA15 and propofol regulate ERK-CREB-RSK2 signaling in hippocampal neurons

Western blotting was conducted to determine the protein expression levels of ERK1/2, p-ERK1/2, p-CREB1, CREB1 and RSK2, in order to examine the molecular mechanisms of PEA15 and propofol on hippocampal neurons. The present results suggested that propofol decreased the protein expression levels of p-ERK1/2, p-CREB1 and RSK2 (Fig. 6A and B). Additionally, it was identified that the ratios between the phosphorylated proteins p-ERK1/2 and p-CREB1, and the respective non-phosphorylated proteins ERK1/2 and CREB1, were significantly decreased (Fig. 6C and D) when cells were treated with propofol. Furthermore, PEA15 promoted the protein expression levels of p-ERK1/2, RSK2 and p-CREB1 in the hippocampal neurons that were treated with propofol. Notably, the protein expression levels of ERK1/2 and CREB1 among groups were not significantly affected.

Discussion

The majority of cultured hippocampal neurons derive from the hippocampus of neonatal rats, as suckling mice gain an advantage in finding a location to feed (32,33). A previous study identified that hippocampal neurons cultured at day 6 are similar to neurons of a developing human brain (34). During this period, neurons are sensitive to external stimuli, including neurotoxicity caused by anesthetics (34). Therefore, in the present study, primary hippocampal neurons were extracted from neonatal rats and used as a model of developing neurons. Following 6 days of culturing, the cell morphology remained stable and the neurons were treated with propofol.

Accumulating evidence suggested that propofol was neurotoxic in vitro (3537). Monni et al (36) observed that propofol promoted cell death in rat hypoglossal motoneurons. Yan et al (37) demonstrated that propofol induced apoptosis of hippocampal neurons in newborn mice. Furthermore, numerous previous studies demonstrated that propofol is a potent inducer of neuronal damage (35,38,39). Similar to previous studies, the present results suggested that propofol significantly decreased the viability and proliferation of neuronal cells, promoting cell apoptosis. Therefore, in the present study, treatment with propofol was used as a model for damaging neuronal cells.

A recent study demonstrated that the expression of PEA15 was downregulated in diabetic SD rats, promoting middle cerebral artery occlusion (25). A previous study demonstrated that overexpression of PEA15 decreased glucose deprivation-induced cell death in neurons (40). Therefore, it was hypothesized that the overexpression of PEA15 may decrease the neurotoxicity of propofol, and that PEA15 and propofol may be associated in hippocampal neurons. As hypothesized, the experimental results suggested that propofol significantly decreased PEA15 expression. Furthermore, the CCK-8 and flow cytometry results suggested that PEA15 overexpression significantly increased the viability and proliferation of neurons, and decreased the apoptosis of neuronal cells, reverting the effects of propofol. The present results suggested that overexpression of PEA15 conferred a neuroprotective effect on propofol-induced neuronal damage by promoting proliferation and decreasing apoptosis.

In order to investigate the mechanisms underlying PEA15 and propofol in neuronal cells, the protein expression levels of apoptosis-associated factors, including pro-caspase-3, active caspase-3, Bcl-2 and Bax, were examined by western blotting and RT-qPCR analysis. Liang et al (35) demonstrated that propofol induced apoptosis of the developing neurons by upregulating the protein expression levels of cleaved-caspase-3 and Bax, and by downregulating the Bcl-2 protein expression level. Similarly, the present results demonstrated that propofol significantly increased the protein expression levels of active caspase-3 and Bax, and decreased pro-caspase-3 and Bcl-2 expression. Furthermore, Ahn et al (22) observed that PEA-15 increased Bcl-2 and pro-caspase-3 protein expression levels, and decreased Bax and cleaved-caspase-3 protein expression levels in neuroblastoma cells treated with 1-methyl-4-phenylpyridinium. The present study demonstrated that the overexpression of PEA15 in neurons treated with propofol, increased the protein expression levels of pro-caspase-3 and Bcl-2, and Bax and cleaved-caspase-3 expression levels were downregulated. The present results suggested that overexpression of PEA15 decreased the apoptotic effects induced by propofol in nerve cells by upregulating pro-caspase-3 and Bcl-2, and by downregulating active caspase-3 and Bax.

ERK-CREB signaling regulates cell growth and apoptosis of rat hippocampal neurons (4143). Furthermore, RSK2 was observed to be involved in the ERK pathway in the hippocampus (44). Kozinn et al (45) demonstrated that propofol inhibited the ERK-CREB pathway in hippocampal neurons. In the present study, it was hypothesized that the mechanisms underlying the PEA15 and propofol effects on the proliferation and apoptosis of hippocampal neurons may be associated with the ERK-CREB pathway. The present results suggested that propofol significantly repressed the phosphorylation levels of ERK1/2 and CREB1, and decreased the protein expression level of RSK2. However, overexpression of PEA15 was able to revert the protein expression levels of p-ERK1/2, p-CREB1 and RSK2, which were suppressed by propofol. The present results suggested that PEA15 overexpression promoted the ERK-CREB-RSK2 signaling pathway. Collectively, the present results suggested that the cell damage caused by propofol was attenuated by PEA15 overexpression. However, testing the effects of PEA15 knockdown may further aid the understanding of the role of PEA15 in neuronal injury.

In conclusion, the present results suggested that PEA15 ameliorated propofol-induced hippocampal nerve damage by promoting cell proliferation, and PEA15 was able to decrease cell apoptosis by upregulating the ERK-CREB-RSK2 signaling pathway. The present study suggested that PEA15 may represent a potential therapeutic agent to treat human diseases characterized by neuronal damage.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

FX provided a substantial contribution to conception and design of the present study. QL and ZC were involved in data acquisition, analysis and interpretation. ZC and FX were involved in drafting the article or critically revising it for important intellectual content. All authors gave final approval of the version to be published. All authors agreed to be accountable for all aspects of the work, and in ensuring that questions related to the accuracy or integrity of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The animal experimental research was approved by the Ethics Committee of The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University (Changzhou, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

King MR and Feldman JM: Optimal management of apparatus dead space in the anesthetized infant. Paediatr Anaesth. 27:1185–1192. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Kotwani MB and Malde AD: Comparison of maintenance, emergence and recovery characteristics of sevoflurane and desflurane in pediatric ambulatory surgery. J Anaesthesiol Clin Pharmacol. 33:503–508. 2017.PubMed/NCBI

3 

Wang CH, Luo J, Li J, Zhang JZ, Huang SY, Shao W and Ma HS: Efficacy of inhalational sevoflurane anesthesia induction on inhibiting the stress response to endotracheal intubation in children with congenital heart disease. Eur Rev Med Pharmacol Sci. 22:1113–1117. 2018.PubMed/NCBI

4 

Chimelli L, Moura Pone S, Avvad-Portari E, Farias Meira Vasconcelos Z, Araújo Zin A, Prado Cunha D, Raposo Thompson N, Lopes Moreira ME, Wiley CA and da Silva Pone MV: Persistence of zika virus after birth: Clinical, virological, neuroimaging, and neuropathological documentation in a 5-month infant with congenital zika syndrome. J Neuropathol Exp Neurol. 77:193–198. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Hannagan T, Nieder A, Viswanathan P and Dehaene S: A random-matrix theory of the number sense. Philos Trans R Soc Lond B Biol Sci. 373:201702532017. View Article : Google Scholar : PubMed/NCBI

6 

Subedi L, Huang H, Pant A, Westgate PM, Bada HS, Bauer JA, Giannone PJ and Sithisarn T: Plasma brain-derived neurotrophic factor levels in newborn infants with neonatal abstinence syndrome. Front Pediatr. 5:2382017. View Article : Google Scholar : PubMed/NCBI

7 

Backeljauw B, Holland SK, Altaye M and Loepke AW: Cognition and brain structure following early childhood surgery with anesthesia. Pediatrics. 136:e1–12. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Sun L: Early childhood general anaesthesia exposure and neurocognitive development. Br J Anaesth. 105 (Suppl 1):i61–i68. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Coleman K, Robertson ND, Dissen GA, Neuringer MD, Martin LD, Cuzon Carlson VC, Kroenke C, Fair D and Brambrink AM: Isoflurane anesthesia has long-term consequences on motor and behavioral development in infant rhesus macaques. Anesthesiology. 126:74–84. 2017. View Article : Google Scholar : PubMed/NCBI

10 

El-Dib M and Soul JS: The use of phenobarbital and other anti-seizure drugs in newborns. Semin Fetal Neonatal Med. 22:321–327. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Hu L, Pan J, Zhang S, Yu J, He K, Shu S and Wang R: Propofol in combination with remifentanil for cesarean section: Placental transfer and effect on mothers and newborns at different induction to delivery intervals. Taiwan J Obstet Gynecol. 56:521–526. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Razlevice I, Rugyte DC, Strumylaite L and Macas A: Assessment of risk factors for cerebral oxygen desaturation during neonatal and infant general anesthesia: An observational, prospective study. BMC Anesthesiol. 16:1072016. View Article : Google Scholar : PubMed/NCBI

13 

Cattano D, Young C, Straiko MM and Olney JW: Subanesthetic doses of propofol induce neuroapoptosis in the infant mouse brain. Anesth Analg. 106:1712–1714. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Creeley C, Dikranian K, Dissen G, Martin L, Olney J and Brambrink A: Propofol-induced apoptosis of neurones and oligodendrocytes in fetal and neonatal rhesus macaque brain. Br J Anaesth. 110 (Suppl 1):i29–i38. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Pearn ML, Hu Y, Niesman IR, Patel HH, Drummond JC, Roth DM, Akassoglou K, Patel PM and Head BP: Propofol neurotoxicity is mediated by p75 neurotrophin receptor activation. Anesthesiology. 116:352–361. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Yu D, Jiang Y, Gao J, Liu B and Chen P: Repeated exposure to propofol potentiates neuroapoptosis and long-term behavioral deficits in neonatal rats. Neurosci Lett. 534:41–46. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Kahraman S, Zup SL, McCarthy MM and Fiskum G: GABAergic mechanism of propofol toxicity in immature neurons. J Neurosurg Anesthesiol. 20:233–240. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Sall JW, Stratmann G, Leong J, Woodward E and Bickler PE: Propofol at clinically relevant concentrations increases neuronal differentiation but is not toxic to hippocampal neural precursor cells in vitro. Anesthesiology. 117:1080–1090. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Spahr-Schopfer I, Vutskits L, Toni N, Buchs PA, Parisi L and Muller D: Differential neurotoxic effects of propofol on dissociated cortical cells and organotypic hippocampal cultures. Anesthesiology. 92:1408–1417. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Danziger N, Yokoyama M, Jay T, Cordier J, Glowinski J and Chneiweiss H: Cellular expression, developmental regulation, and phylogenic conservation of PEA-15, the astrocytic major phosphoprotein and protein kinase C substrate. J Neurochem. 64:1016–1025. 1995. View Article : Google Scholar : PubMed/NCBI

21 

Greig FH and Nixon GF: Phosphoprotein enriched in astrocytes (PEA)-15: A potential therapeutic target in multiple disease states. Pharmacol Ther. 143:265–274. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Ahn EH, Kim DW, Shin MJ, Kim HR, Kim SM, Woo SJ, Eom SA, Jo HS, Kim DS, Cho SW, et al: PEP-1-PEA-15 protects against toxin-induced neuronal damage in a mouse model of Parkinson's disease. Biochim Biophys Acta. 1840:1686–1700. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Greig FH, Kennedy S, Gibson G, Ramos JW and Nixon GF: PEA-15 (Phosphoprotein Enriched in Astrocytes 15) is a protective mediator in the vasculature and is regulated during neointimal hyperplasia. J Am Heart Assoc. 6:e0069362017. View Article : Google Scholar : PubMed/NCBI

24 

Mohammed HN, Pickard MR and Mourtada-Maarabouni M: The protein phosphatase 4-PEA15 axis regulates the survival of breast cancer cells. Cell Signal. 28:1389–1400. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Sung JH and Koh PO: Hyperglycemia aggravates decreases of PEA-15 and its two phosphorylated forms in cerebral ischemia. J Vet Med Sci. 79:654–660. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Wei Y: On the quest of cellular functions of PEA-15 and the therapeutic opportunities. Pharmaceuticals (Basel). 8:455–473. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Huang JY, Wang K, Vermehren-Schmaedick A, Adelman JP and Cohen MS: PARP6 is a regulator of hippocampal dendritic morphogenesis. Sci Rep. 6:185122016. View Article : Google Scholar : PubMed/NCBI

28 

Tang JS, Xie BX, Bian XL, Xue Y, Wei NN, Zhou JH, Hao YC, Li G, Zhang LR and Wang KW: Identification and in vitro pharmacological characterization of a novel and selective α7 nicotinic acetylcholine receptor agonist, Br-IQ17B. Acta Pharmacol Sin. 36:800–812. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Hsu HT, Tseng YT, Hsu YY, Cheng KI, Chou SH and Lo YC: Propofol attenuates lipopolysaccharide-induced reactive oxygen species production through activation of Nrf2/GSH and suppression of NADPH oxidase in human alveolar epithelial cells. Inflammation. 38:415–423. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Xia JH, Shi XY, Xu ZD, Liu G and Wang YH: In vitro effects of propofol on apoptosis and Bax expression induced by TNF-α in mouse spinal cord neurons. Acad J Sec Military Med Univ. 27:169–172. 2006.

31 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Facci L and Skaper SD: Culture of rodent cortical, hippocampal, and striatal neurons. Methods Mol Biol. 1727:39–47. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Rivera-Carvantes MC, Jarero-Basulto JJ, Feria-Velasco AI, Beas-Zárate C, Navarro-Meza M, González-López MB, Gudiño-Cabrera G and García-Rodríguez JC: Changes in the expression level of MAPK pathway components induced by monosodium glutamate-administration produce neuronal death in the hippocampus from neonatal rats. Neuroscience. 365:57–69. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Eibl JK, Strasser BC and Ross GM: Structural, biological, and pharmacological strategies for the inhibition of nerve growth factor. Neurochem Int. 61:1266–1275. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Liang C, Du F, Cang J and Xue Z: Pink1 attenuates propofol-induced apoptosis and oxidative stress in developing neurons. J Anesth. 32:62–69. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Monni L, Ghezzi F, Corsini S and Nistri A: Neurotoxicity of propofol on rat hypoglossal motoneurons in vitro. Neurosci Lett. 655:95–100. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Yan Y, Qiao S, Kikuchi C, Zaja I, Logan S, Jiang C, Arzua T and Bai X: Propofol induces apoptosis of neurons but not astrocytes, oligodendrocytes, or neural stem cells in the neonatal mouse hippocampus. Brain Sci. 7:E1302017. View Article : Google Scholar : PubMed/NCBI

38 

Liu Y, Yan Y, Inagaki Y, Logan S, Bosnjak ZJ and Bai X: Insufficient astrocyte-derived brain-derived neurotrophic factor contributes to propofol-induced neuron death through Akt/glycogen synthase kinase 3β/mitochondrial fission pathway. Anesth Analg. 125:241–254. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Lv J, Wei Y, Chen Y, Zhang X, Gong Z, Jiang Y, Gong Q, Zhou L, Wang H and Xie Y: Dexmedetomidine attenuates propofol-induce neuroapoptosis partly via the activation of the PI3k/Akt/GSK3β pathway in the hippocampus of neonatal rats. Environ Toxicol Pharmacol. 52:121–128. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Huang Q, Voloudakis G, Ren Y, Yoon Y, Zhang E, Kajiwara Y, Shao Z, Xuan Z, Lebedev D, Georgakopoulos A and Robakis NK: Presenilin1/γ-secretase protects neurons from glucose deprivation-induced death by regulating miR-212 and PEA15. FASEB J. 32:243–253. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Shahin S, Banerjee S, Swarup V, Singh SP and Chaturvedi CM: From the cover: 2.45-GHz microwave radiation impairs hippocampal learning and spatial memory: Involvement of local stress mechanism-induced suppression of iGluR/ERK/CREB signaling. Toxicol Sci. 161:349–374. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Yi LT, Li J, Liu BB, Luo L, Liu Q and Geng D: BDNF-ERK-CREB signalling mediates the role of miR-132 in the regulation of the effects of oleanolic acid in male mice. J Psychiatry Neurosci. 39:348–359. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Zuo H, Lin T, Wang D, Peng R, Wang S, Gao Y, Xu X, Zhao L, Wang S and Su Z: RKIP regulates neural cell apoptosis induced by exposure to microwave radiation partly through the MEK/ERK/CREB pathway. Mol Neurobiol. 51:1520–1529. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Kaphzan H, Doron G and Rosenblum K: Co-application of NMDA and dopamine-induced rapid translation of RSK2 in the mature hippocampus. J Neurochem. 103:388–399. 2007.PubMed/NCBI

45 

Kozinn J, Mao L, Arora A, Yang L, Fibuch EE and Wang JQ: Inhibition of glutamatergic activation of extracellular signal-regulated protein kinases in hippocampal neurons by the intravenous anesthetic propofol. Anesthesiology. 105:1182–1191. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xian F, Li Q and Chen Z: Overexpression of phosphoprotein enriched in astrocytes 15 reverses the damage induced by propofol in hippocampal neurons. Mol Med Rep 20: 1583-1592, 2019
APA
Xian, F., Li, Q., & Chen, Z. (2019). Overexpression of phosphoprotein enriched in astrocytes 15 reverses the damage induced by propofol in hippocampal neurons. Molecular Medicine Reports, 20, 1583-1592. https://doi.org/10.3892/mmr.2019.10412
MLA
Xian, F., Li, Q., Chen, Z."Overexpression of phosphoprotein enriched in astrocytes 15 reverses the damage induced by propofol in hippocampal neurons". Molecular Medicine Reports 20.2 (2019): 1583-1592.
Chicago
Xian, F., Li, Q., Chen, Z."Overexpression of phosphoprotein enriched in astrocytes 15 reverses the damage induced by propofol in hippocampal neurons". Molecular Medicine Reports 20, no. 2 (2019): 1583-1592. https://doi.org/10.3892/mmr.2019.10412