Differential hippocampal protein expression between normal aged rats and aged rats with postoperative cognitive dysfunction: A proteomic analysis

  • Authors:
    • Yang Li
    • Saiying Wang
    • Ke Ran
    • Zhonghua Hu
    • Zhaoqian Liu
    • Kaiming Duan
  • View Affiliations

  • Published online on: April 28, 2015     https://doi.org/10.3892/mmr.2015.3697
  • Pages: 2953-2960
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the differences in the expression of hippocampal proteins between normal control aged rats and aged rats with postoperative cognitive dysfunction (POCD). A total of 24 aged rats were randomly divided into a surgery group (n=12) and a control group (n=12). The rats in the surgery group were treated with 2 h isoflurane anesthesia and splenectomy, while the rats in the control group received 40% oxygen for 2 h without surgery. The cognitive functions of the two groups were examined using a Y‑maze test. The protein expression profiles of the hippocampus of six aged rats (three rats with POCD and three from the normal control group) were assessed using two‑dimensional gel electrophoresis and matrix‑assisted laser desorption/ionization time of flight mass spectrometry. A total of three differential proteins were further confirmed between the POCD rats and normal rats using reverse transcription quantitative polymerase chain reaction (RT‑qPCR). The expression levels of 21 proteins in the rats with POCD were significantly different compared with the normal control rats. These proteins were functionally clustered to synaptic plasticity (three proteins), oxidative stress (four proteins), energy production (six proteins), neuroinflammation (three proteins) and glutamate metabolism (two proteins). In addition, three proteins (fatty acid binding protein 7, brain, glutamate dehydrogenase 1 and glutamine synthetase), associated with astrocytic function, were significantly different in the rats with POCD compared with those in the normal control (P<0.05). Similar changes in the mRNA expression levels of the three proteins in the hippocampi of POCD rats were also detected using RT‑qPCR. Neuroinflammation, glutamate toxicity and oxidative stress were possibly involved in the pathological mechanism underlying POCD in aged rats. In addition, astrocytes may also be imporant in POCD in aged rats.

Introduction

Postoperative cognitive dysfunction (POCD) is characterized by a decline of cognitive performance following surgery, particularly following surgery of aged patients (1). It has been identified to occur in 30–80% of aged patients 1 week after surgery and in 10–15% of aged patients 3 months after surgery (24). A number of risk factors for POCD have been reported in previous studies, including advanced age, educational level, mental illness, preoperative level of recognition, operative type, and postoperative infection (4,5). However, the pathological mechanisms underlying POCD remain to be elucidated. The toxicity of anesthetics was initially assessed, with simple anesthesia impairing the learning and memory of aged rats (1,68) and mice (9). Notably, the duration and type of anesthesia had no significant effects on the incidence of POCD of aged patients at the late stage (>1 week post surgery) (10). This suggested that anesthetics were not important in POCD associated with aged patients. In addition to surgery and anesthesia, another potential mechanism underlying POCD is inflammation induced by surgery. He et al (11) and Wang et al (12) reported that surgical trauma under general anesthesia causes distinct changes in systemic and central pro-inflammatory cytokines in aged rats at an early stage (<1 week post surgery), corresponding closely to the dysfunction of cognition. Open tibial fractures impair the cognitive function and induce the increase of tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1) in adult mice (4,13). Inhibiting the signals of TNF-α and IL-1 prior to surgery partly rescued the cognitive dysfunction induced by surgery (4,13). However, a similar incidence of POCD has been detected among aged patients 3 months after surgery, although the extent of the damage induced by surgery was independent of the anaesthetic and the type of surgery (14). This suggested that, in addition to the toxicity of anesthetics and inflammation induced by surgery, other unknown factors also contribute to POCD in aged patients.

The combination of mass spectrometry and two-dimensional gel electrophoresis is a useful method to examine the differential protein expression levels between different treatments. It is usually used to examine novel mechanisms underlying certain diseases (15). The hippocampus is an important structure, closely involved with learning and memory (16,17). During normal aging, the volume of the hippocampus decreases gradually, particularly the dentate gyrus and the CA3 area (18,19). During pathological aging, as observed in Alzheimer’s disease, the hippocampal volume decreases more rapidly (18,19). Accordingly, Chen et al (20) found that the hippocampal volume in aged patients with POCD is significantly smaller compared with that of aged patients without POCD (20). In addition, the hippocampal volume was negatively correlated with the score of cognitive function of aged patients. These suggested that changes in the volume of the hippocampus were closely involved in the occurrence of POCD in aged patients. In the present study, differences in the protein expression of the hippocampus were compared between aged rats with POCD and normal aged rats.

Materials and methods

Animals and grouping

A total of 24 aged male Sprague-Dawley rats (22 months old) were purchased from the Experimental Animal Nursery of Central South University (Changsha, China). The rats were maintained under temperature-controlled environmental conditions with a 12/12-h light/dark cycle and had ad libitum access to food and water. The food consisted of standardized rodent pellets. Ambient conditions remained constant at 23°C and a relative humidity of 50–60%. All procedures were approved by the Medical Ethics Committee of the Third Xiangya Hospital of Central South University and conformed to the guidelines for animal experiments of Central South University.

A total of 24 rats were handled, according to the flow chart of the study design shown in Fig. 1. The rats were divided randomly into a surgery group (n=12) and a normal control group (n=12). The rats in the surgery group were treated with isoflurane anesthesia for 2 h and splenectomy, while rats in the normal control group received 40% oxygen for 2 h without surgery (1). The cognitive functions of the two groups were examined using a Y-maze test. The upper limit of the 95% confidence interval of the mean training duration of rats in the normal control group in the Y-maze test was used to decide whether the rats in the surgery group had cognitive dysfunction (21). The rats in the surgery group were subdivided into POCD and no-POCD groups, according to previously described methods (21). The protein profile from the hippocampi of six aged rats (three rats with POCD and three rats from the normal control group) were detected using two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF-MS).

Y-maze test

The details of the Y-maze assessment (Fig. 2) was described in our previous study (1). Briefly, the voltage for the electric shock in the Y-maze was 50±5 V. The rats were acclimatized to the Y-maze with the light on for 3 min 24 h after treatment. Subsequently, the rats were placed in the stem arm and were administered an electric shock. If the rats arrived at the lit arms within 30 sec, the trial was deemed successful. The same trial was repeated 20 times/day consecutively for 3 days. During the assessment, the following parameters were recorded: The number of correct reactions, which was the total number entering the lit arm of the 20 trials each day; the total reaction duration, which rats spent completing the 20 trials each day and the number of initiative avoidance responses, which was the number of escape responses of the rats within the 5 sec prior to the arms being electrified.

Splenectomy surgery

Splenectomy surgery was performed using the method reported in our previous study (11). Briefly, the rats were initially placed in a closed box with an airflow of 3% isoflurane for 5 min. Anesthesia was then administered through a mask using 2% isoflurane. A 14-gauge catheter was inserted through the glottis. The rats were artificially ventilated, through endotracheal intubation, with room air supplemented with 2 l/min oxygen and 2% isoflurane for 2 h (22). During the anesthesia, the gas concentrations and respiratory rates (RR; breaths per min)were continuously monitored using a multi-function monitor (Datex-Ohmeda, Helsinki, Finland) (Table I). Rectal temperature was maintained at 37±0.5°C. In addition, heart rate (HR), mean arterial blood pressure (MAP) and pulse oximeter oxygen saturation (SpO2) were measured continuously through a femoral artery catheter (Table I). Under anesthesia, a small incision was made in the upper left quadrant through the skin and muscle wall. The spleen was mobilized, isolated and removed. The wound was infiltrated with 0.25% bupivacaine and then closed with sterile sutures. The rats in the normal control group received 40% oxygen for 2 h in a gas chamber without surgery.

Table I

Vital signs of the rats during and after splenectomy surgery.

Table I

Vital signs of the rats during and after splenectomy surgery.

GroupIndex10 min preoperative10 min operative20 min operative10 min postoperative
ControlRR (bpm)82.20±3.3572.06±4.6871.33±3.2978.50±2.67
HR (bpm)374.83±11.70359.83±16.49362.35±20.11365.00±16.07
MAP (mmHg)95.33±10.1783.83±10.3285.29±13.2588.67±7.23
SpO2 (%)97.17±1.4897.50±1.6497.27±1.0697.83±1.16
SurgeryRR (bpm)80.00±4.7071.50±6.7873.75±7.6975.60±8.41
HR (bpm)372.57±12.11348.28±11.33355.88±13.68361.78±12.52
MAP (mmHg)92.21±8.3283.21±9.0785.64±8.3387.14±8.37
SpO2 (%)97.93±1.0797.78±1.3197.65±1.0897.42±1.15

[i] No statistical difference was observed in the RR, MAP and SpO2 between the rats in control group and surgery group (P>0.05). RR; respiratory rate; HR, heart rate; MAP, mean arterial blood pressure; SpO2, oxygen saturation; bpm, beats per minute.

2-DE and MALDI-TOF-MS

Hippocampal tissue (250 mg) was suspended in 1 ml of 8 M urea, 2 M sulfocarbamide, 20 mM Tris-HCl, 1 mM CHAPS, 40 mM EDTA, 65 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, 0.025 mM RNase, 0.05 mM DNase and 1 mM NaF. The suspension was sonicated for ~30 sec and centrifuged at 150,000 × g for 45 min. The protein concentration in the supernatant was examined using the Coomassie blue method (23). In first dimensional isoelectric focusing, 1,000 μg of each sample was loaded into immobilized pH 3–10 nonlinear gradient strips (18 cm). After 12 h of reswelling at 30 V, voltages of 100, 500, 1,000 and 3,000 V were administered for 1 h each and 8,000 V for 6 h. The second dimensional separation was performed using 12% sodium dodecyl sulfate-polyacrylamide gels (Sangon Biotech Shanghai Co., Ltd, Shanghai, China) (24). Following staining of the gels with colloidal Coomassie blue (25), the gels were scanned using a Umax PowerLook II scanner and Adobe Photoshop 8.0 image software (Adobe Systems Inc., San Jose,. CA, USA), and the images were analyzed using ETTAN ImageMaster 2D Elite 4.01 software (GE Healthcare, Amersham, UK). This software was used for matching and quantitative analysis of the protein spots on the gels. The average gel was constructed as a representative gel for the three hippocampal samples acquired from each group of rats. The average mode of background subtraction was used for normalization of the intensity volume that indicates the protein concentration or the quantity on each spot. The average gel was then used for determination of the existence of differences in the protein expression levels between each group. A value of 2.0 indicated a 2-fold increase and 0.5 indicated a 2-fold reduction (2629). Biologically relevant differences were defined as changes >2-fold.

MALDI-TOF-MS analysis was performed, as described previously (2529). The valuable spots in the coomassie blue-stained gels were assessed. The extracts were redissolved in 1 μl extraction buffer and 1 μl matrix solution (α-acyano-4-hydroxycinnamic acid; Sangon Biotech Shanghai Co., Ltd) and targeted onto a MALDI-TOF plate. Following drying the samples completely onto the targeting plate, MALDI-TOF/MS was performed using a Voyager-DE STR mass spectrometer (Applied Biosystems, Foster City, CA, USA) equipped with delay ion extraction. Mass spectra were obtained over a mass range of 800–3,000 Da. For the identification of proteins, peptide mass fingerprinting data were used to search against the Mascot database (http://www.matrixscience.com) at the School of Life (Central South University). Peptide matching and protein identification were performed automatically, as described previously (2629). A Mascot score >58 was considered to indicate statistical significance (P<0.05).

Reverse transcription quantitative polymerase chain reaction (RT-qPCR) for astrocyte-associated proteins

To confirm the differential protein expression based on MALDI-TOF-MS, the mRNA expression levels of the proteins of interest were detected by RT-qPCR. Total RNA from the hippocampus was isolated using the RNeasy plus mini kit (Qiagen, Valencia, CA, USA), according to the manufacturer’s instructions. Prior to RT, the total RNA was treated to remove genomic DNA using a DNA-free kit (AM1906; Applied Biosystems). cDNA was synthesized using the Retroscript kit (AM1710; Applied Biosystems). RT-qPCR was performed using Power SYBR Green PCR master mix (Applied Biosystems). The rat β-actin gene was used as a homogenous standard. The running protocol extended to 40 cycles consisting of 95°C for 15 sec and 60°C for 1 min using an Applied Biosystems 7500 Fast Real-time PCR system. The primers were designed using Primer 5.0 software (Premier Biosoft International, Palo Alto, CA, USA; Table II).

Table II

Primers used for reverse transcription quantitative polymerase chain reaction.

Table II

Primers used for reverse transcription quantitative polymerase chain reaction.

GenePrimerAmplicon length(bp)
Glutamate dehydrogenaseForward: GCTCTGGACTCTTCCCAACA115
Reverse: AAATGCCACACGCCTACTTC
Glutamine synthetaseForward: CGCTCTTCGTCTCGTTCTC119
Reverse: CTGCTTGATGCCTTTGTTCA
Fatty acid binding protein 7, brainForward: GAAGGGCAAGGATGGTAGATG133
Reverse: ACCGTTGGTTTGGTCACATT
β-actinForward: GGAAATCGTGCGTGACATTA181
Reverse: GAAGGAAGGCTGGAAGAGAG

The relative quantity of the target gene present was calculated based on the expression of β-actin in the endogenous control. The mean cycle threshold (Ct) values and standard deviations were calculated. To calibrate the analysis, the value obtained from the control rats was used. The factor difference was also calculated using the standard relative quantitative method (2−ΔΔCT method) (30,31).

Statistical analysis

All summary data are reported as the mean ± standard error of the mean. The results of the present study were processed using SPSS 17.0 statistical software (SPSS, Inc., Chicago, IL, USA). The total reaction time in the Y maze was analyzed using a repeated measures analysis of variance (ANOVA) with Bonferroni’s post-hoc test. The number of correct reactions and initiative avoidance occurrences were examined using a non-parametric Mann-Whitney U test. The RR, HR and MAP values during surgery were compared using a repeated measures ANOVA with Bonferroni’s post-hoc test. The gel electrophoresis and RT-qPCR results were analyzed using Student’s t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Surgery with inhaled anesthesia impairs the cognitive function of aged rats

In the present study, a Y-maze test was used to evaluate cognitive function (1). A decrease of correct reactions and initiative avoidance number, and an increase in total reaction time implied deteriorating cognitive function. The findings demonstrated that the number of correct reactions in the surgery group were 8.90±1.09 and 9.47±2.46 (24 and 48 h, respectively) following surgery, which was significantly lower compared with the control group (P<0.05; Fig. 3A). The number of initiative avoidance occurrences in the surgery group were 1.20±0.59 and 2.07±1.01 (24 h and 48 h, respectively) following surgery, which was also significantly lower compared with the control group (P<0.05; Fig. 3B). These findings demonstrated that surgery with inhaled anesthesia impaired the cognitive functions of aged rats at an early stage.

Surgery with inhaled anesthesia alters the protein profile of the hippocampus in aged rats with POCD

The protein expression profile of the hippocampus was assessed using MALDI-TOF-MS. The levels of 21 proteins in the hippocampus of aged rats with POCD were markedly altered, compared with the normal control. These proteins included dynamin-1, superoxide dismutase, glutamine synthetase and coronin-1A (Table III, Fig. 4). Further functional clustering revealed that three proteins were closely involved in synaptic plasticity, four in oxidative stress, six in energy production three in neuroinflammation and two in glutamate metabolism. The protein expression levels were downregulated, with regard to synaptic plasticity, oxidative stress, energy production and glutamate metabolism; however, the protein expression levels associated with neuroinflammation were upregulated in the POCD rats.

Table III

Differential protein expression between the control group and postoperative cognitive dysfunction group.

Table III

Differential protein expression between the control group and postoperative cognitive dysfunction group.

Protein symbolProtein nameNumber of spots in gelChanges in expression (POCD, vs. control)Protein function
DNM1Dynamin-12Synaptic plasticity
DHPRP2 Dihydropyrimidinase-related protein 23Synaptic plasticity
GABA-TGABA transaminase19Modulating neuronal plasticity
SOD1Superoxide dismutase (Cu-Zn)8Antioxidant protective role
GSTP1Glutathione S-transferase P12Antioxidant protective role
PRDX2 Peroxiredoxin-25Antioxidant protective role
ADRAldose reductase14Oxidative stress
DLST Dihydrolipoyllysine-residue succinyltransferase4Energy production in mitochondria
ATP5DATP synthase δ chain7Energy production in mitochondria
NDUFA10NADH dehydrogenase1 α subcomplex subunit 1013Energy production in mitochondria
PGAM1Phosphoglycerate mutase 111Energy production in mitochondria
TKTTransketolase20Energy production and neurogenesis
PK-M1/M2Pyruvate kinase isozymes M1/M221Energy production in mitochondria
CORO1ACoronin-1A1Neuroinflammation marker
GMFBGlia maturation factor b6Modulating neuroinflammation
CRYABα crystallin B chain11Modulating neuroinflammation
FABP7Fatty acid-binding protein,9Regulation of astrocyte function
GSGlutamine synthetase17Modulating extracellular glutamate level
GLUD1Glutamate dehydrogenase18Modulating extracellular glutamate level and energy production of mitochondria
CCT2T-complex protein 1 subunit β23Protein folding
EF-TuElongation factor Tu16Modulating translation of DNA

[i] Expression factors were calculated, as described in the Materials and methods. Spot number of the corresponding reference gel, the full name and the function are provided. ↑ indicates increased expression, ↓ indicates decreased expression.

Astrocytes are the predominant type of cell in the hippocampus, with important roles in supporting neurons. Thus, from the identified differentially expressed proteins, three proteins associated with astrocytes were selected [fatty acid binding protein 7, brain (FABP7) (32), glutamate dehydrogenase 1 (GLUD1) (33) and glutamine synthetase (GS) (34)] to confirm the reliability of the data from the MALDI-TOF-MS assessment. It was identified that the mRNAs of FABP-7 and GLUD increased in aged rats with POCD and the mRNAs of GS decreased, closely corresponding to the changes observed in the MALDI-TOF-MS data (P<0.05; Fig. 5).

Discussion

In the present study, the protein profile of the hippocampi in aged rats with POCD and normal aged rats were screened. A total of 21 proteins were significantly altered in aged rats with POCD compared with normal aged rats. These proteins were functionally clustered in association with synaptic plasticity, oxidative stress, energy production, glutamate metabolism and neuroinflammation. These data provided important indications that the hippocampus is functionally important in the POCD of aged rats.

POCD is usually detected among aged patients following surgery. It was initially reported by Bedford in 1955 (35). In the present study, the cognitive function of aged rats was initially assessed with or without surgery using a Y-maze. The number of correct reactions and the number of initiative avoidance occurrences in the surgery group were significantly lower compared with the control group (P<0.05). These results suggested that surgery with inhaled anesthesia impaired the cognitive function of aged rats at an early stage. Based on these findings, changes in the protein expression profile of the hippocampi of aged rats with POCD was assessed. Compared with the normal control, 14 proteins increased in the aged rats with POCD and seven proteins decreased (Table II). PubMed (http://www.ncbi.nlm.nih.gov/pubmed/) was used to search for useful information using key words. Among the 21 proteins, three proteins, DNM1 (3638), DHPRP2 (39) and GABA-T (40,41) were involved in synaptic plasticity and their expression were decreased in aged rats with POCD. A total of four proteins, SOD1 (42,43), glutathione S-transferase P (44,45), peroxiredoxin 2 (46) and ADR (47) were associated with oxidative stress. In addition, six proteins, dihydrolipoyllysine-residue succinyltransferase (48), ATP synthase δ chain (49), NADH dehydrogenase 1α (50), phosphoglycerate mutase 1 (51), transketolase (52) and pyruvate kinase isozymes M1/M2 (53), were involved in energy production and their expression levels were decreased in the aged rats with POCD. A total of three proteins, coronin-1A (54), glia maturation factor b (55), α crystallin B chain (56) were involved in neuroinflammation. These findings suggested that surgery induced neuronal damage in aged rats with POCD. Neuroinflammation and oxidative stress were important in neuronal damage and the data were consistent with previously reported results (4,1113,20,57). Chen et al (20) found that aged patients with POCD had a smaller volume hippocampus and the size of the hippocampus was negatively correlated with the cognitive function of aged patients following surgery. It has been found that surgery induced increased neuroinflammation in mice and aged rats (4,11,12), corresponding to cognitive dysfunction. In addition, An et al (57) found that surgical trauma induced oxidative stress in rats with POCD (57). Notably, the expression of FABP7 (32), a modulator of astrocyte function, was higher in aged rats with POCD. At the same time, GS (58) and GLUD1 (33,5961), which are two astrocytic enzymes modulating the level of extracellular glutamate, were also changed in aged rats with POCD. These findings suggested that astrocytes are important in the mechanism underlying POCD, and astrocytes may offer a novel direction in the investigation of POCD.

The present study did not distinguish the effect of surgery in POCD from the anaesthetic agent, as our previous study demonstrated that anesthesia and surgery can result in POCD. It has been confirmed that surgery, rather than anesthetic agents lead to long-term changes in exploratory behavior (62). It is disputed whether the mechanism underlying POCD is associated with surgery and/or anesthesia, however, it is considered to be a multi-factorial process. Further investigation is required to discriminate each component of the entire process.

Acknowledgments

The authors would like to thank all those who assisted with this study, particularly Professor Jianbin Tong, who provided technical assistance and critically edited the manuscript.

References

1 

Yan XB, Ouyang W, Li G and Duan KM: Involvement of neuronal nitric oxide synthase in cognitive impairment in isoflurane-treated rats. Neurosci Lett. 506:240–244. 2012. View Article : Google Scholar

2 

Ramaiah R and Lam AM: Postoperative cognitive dysfunction in the elderly. Anesthesiol Clin. 27:485–496. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Monk TG and Price CC: Postoperative cognitive disorders. Curr Opin Crit Care. 17:376–381. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Terrando N, Monaco C, Ma D, Foxwell BM, Feldmann M and Maze M: Tumor necrosis factor-alpha triggers a cytokine cascade yielding postoperative cognitive decline. Proc Natl Acad Sci USA. 107:20518–20522. 2012. View Article : Google Scholar

5 

Rosczyk HA, Sparkman NL and Johnson RW: Neuroinflammation and cognitive function in aged mice following minor surgery. Exp Gerontol. 43:840–846. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Culley DJ, Baxter MG, Yukhananov R and Crosby G: Long-term impairment of acquisition of a spatial memory task following isoflurane-nitrous oxide anesthesia in rats. Anesthesiology. 100:309–314. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Culley DJ, Baxter M, Yukhananov R and Crosby G, Culley DJ, Baxter M, Yukhananov R and Crosby G: The memory effects of general anesthesia persist for weeks in young and aged rats. Anesth Analg. 96:1004–1009. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Culley DJ, Baxter MG, Crosby CA, Yukhananov R and Crosby G: Impaired acquisition of spatial memory 2 weeks after isoflurane and isoflurane-nitrous oxide anesthesia in aged rats. Anesth Analg. 99:1393–1397. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Hudson AE and Hemmings HC Jr: Are anaesthetics toxic to the brain? Br J Anaesth. 107:30–37. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Rasmussen LS, Johnson T, Kuipers HM, Kristensen D, Siersma VD, Vila P, Jolles J, Papaioannou A, Abildstrom H, Silverstein JH, Bonal JA, Raeder J, Nielsen IK, Korttila K, Munoz L, Dodds C, Hanning CD and Moller JT: ISPOCD2 (international study of postoperative cognitive dysfunction) investigators: does anaesthesia cause postoperative cognitive dysfunction? A randomised study of regional versus general anaesthesia in 438 elderly patients. Acta Anaesthesiol Scand. 47:260–266. 2003. View Article : Google Scholar : PubMed/NCBI

11 

He HJ, Wang Y, Le Y, Duan KM, Yan XB, Liao Q, Liao Y, Tong JB, Terrando N and Ouyang W: Surgery upregulates high mobility group box-1 and disrupts the blood-brain barrier causing cognitive dysfunction in aged rats. CNS Neurosci Ther. 18:994–1002. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Wang Y, He HJ, Li D, Zhu WJ, Duan KM, Le Y, Liao Y and Ouyang W: The role of the TLR4 signaling pathway in cognitive deficits following surgery in aged rats. Mol Med Rep. 7:1137–1142. 2013.PubMed/NCBI

13 

Terrando N, Eriksson LI, Ryu JK, Yang T, Monaco C, Feldmann M, Jonsson Fagerlund M, Charo IF, Akassoglou K and Maze M: Resolving postoperative neuroinflammation and cognitive decline. Ann Neurol. 70:986–995. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Evered L, Scott DA, Silbert B and Maruff P: Postoperative cognitive dysfunction is independent of type of surgery and anesthetic. Anesth Analg. 112:1179–1185. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Bi FF, Huang C, Tong JB, Qiu G, Huang B, Wu Qx, Li F, Xu ZS, Xia XG and Zhou HX: Reactive astrocytes secrete Lcn2 to assist neurons in degeneration. Proc Natl Acad Sci USA. 110:4069–4074. 2013. View Article : Google Scholar

16 

Dede AJ, Wixted JT, Hopkins RO and Squire LR: Hippocampal damage impairs recognition memory broadly, affecting both parameters in two prominent models of memory. Proc Natl Acad Sci USA. 1:6577–6582. 2013. View Article : Google Scholar

17 

Da Silva WC, Cardoso G, Bonini JS, Benetti F and Izquierdo I: Memory reconsolidation and its maintenance depend on L-voltage-dependent calcium channels and CaMKII functions regulating protein turnover in the hippocampus. Proc Natl Acad Sci USA. 110:6566–6570. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Buckner RL: Memory and executive function in aging and AD: multiple factors that cause decline and reserve factors that compensate. Neuron. 44:195–208. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Jagust W: Vulnerable neural systems and the borderland of brain aging and neurodegeneration. Neuron. 77:219–234. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Chen MH, Liao Y, Rong PF, Hu R, Lin GX and Ouyang W: Hippocampal volume reduction in elderly patients at risk for postoperative cognitive dysfunction. J Anesth. 27:487–492. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Chen SQ, Wang PJ, Ten GJ, Zhan W, Li MH and Zang FC: Role of myo-inositol by magnetic resonance spectroscopy in early diagnosis of Alzheimer’s disease in APP/PS1 transgenic mice. Dement Geriatr Cogn Disord. 28:558–566. 2009. View Article : Google Scholar

22 

Covasala O, Stirn SL, Albrecht S, De Col R and Messlinger K: Calcitonin gene-related peptide receptors in rat trigeminal ganglion do not control spinal trigeminal activity. J Neurophysiol. 108:431–440. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Krauspe R and Scheer A: Coomassie brilliant blue G-250 dye-binding technique for determination of autolytic protein breakdown in Euglena gracilis and comparison to other methods of autolysis measurement. Anal Biochem. 153:242–250. 1986. View Article : Google Scholar : PubMed/NCBI

24 

Oguri T, Takahata I, Katsuta K, Nomura E, Hidaka M and Inagaki N: Proteome analysis of rat hippocampal neurons by multiple large gel two-dimensional electrophoresis. Proteomics. 2:666–672. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Fountoulakis M and Gasser R: Proteomic analysis of the cell envelope fraction of Escherichia coli. Amino Acids. 24:19–41. 2003.PubMed/NCBI

26 

Opii WO, Joshi G, Head E, Milgram NW, Muggenburg BA, Klein JB, Pierce WM, Cotman CW and Butterfield DA: Proteomic identification of brain proteins in the canine model of human aging following a long-term treatment with antioxidants and a program of behavioral enrichment: relevance to Alzheimer’s disease. Neurobiol Aging. 29:51–70. 2008. View Article : Google Scholar

27 

Berndt P, Hobohm U and Langen H: Reliable automatic protein identification from matrix-assisted laser desorption/ionization mass spectrometric peptide fingerprints. Electrophoresis. 20:3521–3526. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Nikolov M, Schmidt C and Urlaub H: Quantitative mass spectrometry-based proteomics. Methods Mol Biol. 893:85–100. 2012.

29 

Nagappan A, Park HS, Park KI, Kim JA, Hong GE, Kang SR, Zhang J, Kim EH, Lee WS, Won CK and Kim GS: Proteomic analysis of differentially expressed proteins in vitamin C-treated AGS cells. BMC Biochem. 14:242013. View Article : Google Scholar : PubMed/NCBI

30 

Nolan T, Hands RE and Bustin SA: Quantification of mRNA using real-time RT-PCR. Nat Protoc. 1:1559–1582. 2006. View Article : Google Scholar

31 

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

32 

Kipp M, Clarner T, Gingele S, Pott F, Amor S, van der Valk P and Beyer C: Brain lipid binding protein (FABP7) as modulator of astrocyte function. Physiol Res. 60(Suppl 1): 49–60. 2011.

33 

Aubrecht J and Schomaker S: Serum glutamate dehydrogenase as a potential biomarker of mitochondrial dysfunction. Toxicol Sci. 134:2232013. View Article : Google Scholar : PubMed/NCBI

34 

Butterfield DA, Hensley K, Cole P, et al: Oxidatively induced structural alteration of glutamine synthetase assessed by analysis of spin label incorporation kinetics: relevance to Alzheimer’s disease. J Neurochem. 68:2451–2457. 1997. View Article : Google Scholar : PubMed/NCBI

35 

Bedford PD: Adverse cerebral effects of anaesthesia on old people. Lancet. 269:259–263. 1955. View Article : Google Scholar : PubMed/NCBI

36 

Mallozzi C, D’Amore C, Camerini S, Macchia G, Crescenzi M, Petrucci TC and Di Stasi AM: Phosphorylation and nitration of tyrosine residues affect functional properties of Synaptophysin and Dynamin I, two proteins involved in exo-endocytosis of synaptic vesicles. Biochim Biophys Acta. 1833:110–121. 2013. View Article : Google Scholar

37 

Merriam LA, Baran CN, Girard BM, Hardwick JC, May V and Parsons RL: Pituitary adenylate cyclase 1 receptor internalization and endosomal signaling mediate the pituitary adenylate cyclase activating polypeptide-induced increase in guinea pig cardiac neuron excitability. J Neurosci. 33:4614–4422. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Yamada H, Abe T, Satoh A, Okazaki N, Tago S, Kobayashi K, Yoshida Y, Oda Y, Watanabe M, Tomizawa K, Matsui H and Takei K: Stabilization of actin bundles by a dynamin 1/cortactin ring complex is necessary for growth cone filopodia. J Neurosci. 33:4514–4526. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Koide T, Aleksic B, Ito Y, Usui H, Yoshimi A, Inada T, Suzuki M, Hashimoto R, Takeda M, Iwata N and Ozaki N: A two-stage case-control association study of the dihydropyrimidinase-like 2 gene (DPYSL2) with schizophrenia in Japanese subjects. J Hum Genet. 55:469–472. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Wang QP, Jammoul F, Duboc A, Gong J, Simonutti M, Dubus E, Craft CM, Ye W, Sahel JA and Picaud S: Treatment of epilepsy: the GABA-transaminase inhibitor, vigabatrin, induces neuronal plasticity in the mouse retina. Eur J Neurosci. 27:2177–2187. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Hwang IK, Kim DW, Yoo KY, Kim DS, Kim KS, Kang JH, Choi SY, Kim YS, Kang TC and Won MH: Age-related changes of gamma-aminobutyric acid transaminase immunoreactivity in the hippocampus and dentate gyrus of the Mongolian gerbil. Brain Res. 1017:77–84. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Wootz H, Fitzsimons-Kantamneni E, Larhammar M, Rotterman TM, Enjin A, Patra K, André E, Van Zundert B, Kullander K and Alvarez FJ: Alterations in the motor neuron-renshaw cell circuit in the Sod1 (G93A) mouse model. J Comp Neurol. 521:1449–1469. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Kang SH, Li Y, Fukaya M, Lorenzini I, Cleveland DW, Ostrow LW, Rothstein JD and Bergles DE: Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nat Neurosci. 16:571–579. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Castro-Caldas M, Carvalho AN, Rodrigues E, Henderson C, Wolf CR and Gama MJ: Glutathione S-transferase pi mediates MPTP-induced c-Jun N-terminal kinase activation in the nigrostriatal pathway. Mol Neurobiol. 45:466–477. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Korff A, Pfeiffer B, Smeyne M, Kocak M, Pfeiffer RF and Smeyne RJ: Alterations in glutathione S-transferase pi expression following exposure to MPP+ -induced oxidative stress in the blood of Parkinson’s disease patients. Parkinsonism Relat Disord. 17:765–768. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Sánchez-Font MF, Sebastià J, Sanfeliu C, Cristòfol R, Marfany G and Gonzàlez-Duarte R: Peroxiredoxin 2 (PRDX2), an antioxidant enzyme, is under-expressed in Down syndrome fetal brains. Cell Mol Life Sci. 60:1513–1523. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Tang WH, Martin KA and Hwa J: Aldose reductase, oxidative stress and diabetic mellitus. Front Pharmacol. 3:872012. View Article : Google Scholar

48 

Takase C, Nakano K, Ohta S, Nakagawa S and Matuda SY: Different distribution of dihydrolipoamide succinyltransferase, dihydrolipoamide acetyltransferase and ATP synthase beta-subunit in monkey brain. In Vivo. 10:495–501. 1996.PubMed/NCBI

49 

Ko YH, Pan W, Inoue C and Pedersen PL: Signal transduction to mitochondrial ATP synthase: evidence that PDGF-dependent phosphorylation of the delta-subunit occurs in several cell lines, involves tyrosine and is modulated by lysophosphatidic acid. Mitochondrion. 1:339–348. 2002. View Article : Google Scholar

50 

Adam-Vizi V and Tretter L: The role of mitochondrial dehydrogenases in the generation of oxidative stress. Neurochem Int. 62:757–763. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Batke J, Nazaryan KB and Karapetian NH: Complex of brain D-phosphoglycerate mutase and gamma enolase and its reactivation by D-glycerate 2,3-bisphosphate. Arch Biochem Biophys. 264:510–518. 1988. View Article : Google Scholar : PubMed/NCBI

52 

Zhao Y, Pan X, Zhao J, Wang Y, Peng Y and Zhong C: Decreased transketolase activity contributes to impaired hippocampal neurogenesis induced by thiamine deficiency. J Neurochem. 111:537–546. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Tolle SW, Dyson RD, Newburgh RW and Cardenas JM: Pyruvate kinase isozymes in neurons, glia, neuroblastoma and glioblastoma. J Neurochem. 27:1355–1360. 1976. View Article : Google Scholar : PubMed/NCBI

54 

Ahmed Z, Shaw G, Sharma VP, Yang C, McGowan E and Dickson DW: Actin-binding proteins coronin-1a and IBA-1 are effective microglial markers for immunohistochemistry. J Histochem Cytochem. 55:687–700. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Zaheer A, Zaheer S, Sahu SK, Knight S, Khosravi H, Mathur SN and Lim R: A novel role of glia maturation factor: induction of granulocyte-macrophage colony-stimulating factor and pro-inflammatory cytokines. J Neurochem. 101:364–376. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Shao W, Zhang SZ, Tang M, Zhang XH, Zhou Z, Yin YQ, Zhou QB, Huang YY, Liu YJ, Wawrousek E, Chen T, Li SB, Xu M, Zhou JN, Hu G and Zhou JW: Suppression of neuroinflammation by astrocytic dopamine D2 receptors via αB-crystallin. Nature. 494:90–94. 2013. View Article : Google Scholar

57 

An LN, Yue Y, Guo WZ, Miao YL, Mi WD, Zhang H, Lei ZL, Han SJ and Dong L: Surgical trauma induces iron accumulation and oxidative stress in a rodent model of postoperative cognitive dysfunction. Biol Trace Elem Res. 151:277–283. 2013. View Article : Google Scholar

58 

Cooper AJ: Possible treatment of end-stage hyperammonemic encephalopathy by inhibition of glutamine synthetase. Metab Brain Dis. 28:119–125. 2013. View Article : Google Scholar

59 

Plaitakis A, Zaganas I and Spanaki C: Deregulation of glutamate dehydrogenase in human neurologic disorders. J Neurosci Res. 91:1007–1017. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Frigerio F, Karaca M, De Roo M, Mlynárik V, Skytt DM, Carobbio S, Pajęcka K, Waagepetersen HS, Gruetter R, Muller D and Maechler P: Deletion of glutamate dehydrogenase 1 (Glud1) in the central nervous system affects glutamate handling without altering synaptic transmission. J Neurochem. 123:342–348. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Skytt DM, Klawonn AM, Stridh MH, Pajęcka K, Patruss Y, Quintana-Cabrera R, Bolaños JP, Schousboe A and Waagepetersen HS: siRNA knock down of glutamate dehydrogenase in astrocytes affects glutamate metabolism leading to extensive accumulation of the neuroactive amino acids glutamate and aspartate. Neurochem Int. 61:490–497. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Hovens IB, Schoemaker RG, van der Zee EA, Heineman E, Nyakas C and van Leeuwen BL: Surgery-induced behavioral changes in aged rats. Exp Gerontol. 48:1204–1211. 2012. View Article : Google Scholar

Related Articles

Journal Cover

August-2015
Volume 12 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
Li Y, Wang S, Ran K, Hu Z, Liu Z and Duan K: Differential hippocampal protein expression between normal aged rats and aged rats with postoperative cognitive dysfunction: A proteomic analysis. Mol Med Rep 12: 2953-2960, 2015
APA
Li, Y., Wang, S., Ran, K., Hu, Z., Liu, Z., & Duan, K. (2015). Differential hippocampal protein expression between normal aged rats and aged rats with postoperative cognitive dysfunction: A proteomic analysis. Molecular Medicine Reports, 12, 2953-2960. https://doi.org/10.3892/mmr.2015.3697
MLA
Li, Y., Wang, S., Ran, K., Hu, Z., Liu, Z., Duan, K."Differential hippocampal protein expression between normal aged rats and aged rats with postoperative cognitive dysfunction: A proteomic analysis". Molecular Medicine Reports 12.2 (2015): 2953-2960.
Chicago
Li, Y., Wang, S., Ran, K., Hu, Z., Liu, Z., Duan, K."Differential hippocampal protein expression between normal aged rats and aged rats with postoperative cognitive dysfunction: A proteomic analysis". Molecular Medicine Reports 12, no. 2 (2015): 2953-2960. https://doi.org/10.3892/mmr.2015.3697