Accepted Manuscript iTRAQ-based proteomics analysis of hippocampus in spatial memory deficiency rats induced by simulated microgravity
Tingmei Wang, Hailong Chen, Ke Lv, Guohua Ji, Yongliang Zhang, Yanli Wang, Yinghui Li, Lina Qu PII: DOI: Reference:
S1874-3919(17)30097-0 doi: 10.1016/j.jprot.2017.03.013 JPROT 2801
To appear in:
Journal of Proteomics
Received date: Revised date: Accepted date:
10 November 2016 2 March 2017 17 March 2017
Please cite this article as: Tingmei Wang, Hailong Chen, Ke Lv, Guohua Ji, Yongliang Zhang, Yanli Wang, Yinghui Li, Lina Qu , iTRAQ-based proteomics analysis of hippocampus in spatial memory deficiency rats induced by simulated microgravity. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Jprot(2017), doi: 10.1016/j.jprot.2017.03.013
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT 1
iTRAQ-based proteomics analysis of hippocampus in spatial memory deficiency rats induced by simulated microgravity Tingmei Wang1, 2, Hailong Chen1, 3, Ke Lv1, Guohua Ji1, Yongliang Zhang1, 2, Yanli Wang1, 2, Yinghui Li1, 2, and Lina Qu1,* 1
State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China 2
Space Institute of Southern China (Shenzhen), Shenzhen, 518117, China
RI
3
PT
School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072, China
SC
*Corresponding author: Lina Qu:
NU
State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, No.26 Beiqing Road, Haidian District, Beijing, 100094, P. R.
MA
China.
AC
CE
PT E
D
E-mail:
[email protected]
ACCEPTED MANUSCRIPT 2
iTRAQ-based proteomics analysis of hippocampus in spatial memory deficiency rats induced by simulated microgravity Tingmei Wang1, 2, Hailong Chen1, 3, Ke Lv1, Guohua Ji1, Yongliang Zhang1, 2, Yanli Wang1, 2, Yinghui Li1, 2 , Lina Qu1,* 1
State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut
PT
Research and Training Center, Beijing, 100094, China 2
School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072, China Space Institute of Southern China (Shenzhen), Shenzhen, 518117, China
RI
3
SC
Abstract:
It has been demonstrated that simulated microgravity (SM) may lead to cognitive dysfunction.
NU
However, the underlying mechanism remains unclear. In present study, tail-suspension (30°) rat was employed to explore the effects of 28 days of SM on hippocampus-dependent learning and
MA
memory capability and the underlying mechanisms. We found that 28-day tail-suspension rats displayed decline of learning and memory ability in Morris water maze (MWM) test. Using
D
iTRAQ-based proteomics analysis, a total of 4774 proteins were quantified in hippocampus. Of
PT E
these identified proteins, 147 proteins were differentially expressed between SM and control group. Further analysis showed these differentially expressed proteins (DEPs) involved in different molecular function categories, and participated in many biological processes. Based on
CE
the results of PANTHER pathway analysis and further western blot verification, we observed the expression of glutamate receptor 1 (GluR1) and glutamate receptor 4 (GluR4) which involved in
AC
metabotropic glutamate receptor group III pathway and ionotropic glutamate receptor pathway were significantly induced by SM. Moreover, an increased concentration of glutamic acid (Glu) was also found in hippocampus while the concentrations of 5-hydroxytryptamine (5-HT), dopamine (DA), γ-amino acid butyric acid (GABA) and epinephrine (E) were decreased. Our finding confirms that 28-day SM exposure can cause degrading of the spatial learning and memory capability and the possible mechanisms might be related with glutamate excitotoxicity and imbalances in specific neurotransmitters. Key
words:
Excitotoxicity
Simulated
microgravity;
iTRAQ;
Cognitive
dysfunction;
Hippocampus;
ACCEPTED MANUSCRIPT 3
Significance statement: The goal of sending astronauts farther into space and extending the duration of spaceflight missions from months to years will challenge the current capabilities of bioastronautics. The investigation of the physiological and pathological changes induced by spaceflight will be critical in developing countermeasures to ensure astronauts to complete spaceflight mission accurately
PT
and effectively and return to earth safely. It has been demonstrated that spaceflight may lead to impairments in cognitive function which is crucial for mission success. Here we show that
RI
long-term simulated microgravity, the most potent environment risk factor during spaceflight, impairs the spatial learning and memory of rats and the underlying mechanism may be involved in
SC
glutamate excitotoxicity and imbalances in specific neurotransmitters release in hippocampus, which may provide new insight for the countermeasures of cognitive impairment during
AC
CE
PT E
D
MA
NU
spaceflight.
ACCEPTED MANUSCRIPT 4
Introduction The plan of future space exploration mission requires astronauts to spend months or even years in space. It was reported that spaceflight can have many detrimental effects on physiological function of astronauts, due to various harmful stressors such as microgravity, radiation, physical isolation and high workload[1]. Among all these aeronautic environmental stressors, microgravity is
PT
considered to be one of the main hazards threatening human health [1-3]. Actual (spaceflight) or simulated microgravity can directly induce various physiological adaptations, including those in
RI
the central nervous system [4]. Previous studies have suggested that microgravity affects several aspects of brain functions, such as cognitive performance, posture control, locomotion, manual
SC
control [5-10]. Since the success of the spaceflight is highly dependent on astronauts’ mental health, the investigation of the underlying molecular mechanisms involved in the initiation and
NU
progression of psychopathologies is one of the main challenges of the space neurobiology. Hippocampus is part of the limbic system and plays important roles in learning and memory
MA
formation, and specifically spatial learning and episodic memory. Any malfunction and/or structural changes of the hippocampus could have substantial consequences on learning and
D
memory consolidation as well as general cognitive performance. According to the previous studies,
PT E
7 days of SM exposure significantly changed the expression of cytoskeleton, metabolism, mitochondrial function related proteins in hippocampus [11, 12]. 21 days of SM induced changes in neurotransmitters (GABA and Glu), and metabolism related proteins [2, 12]. Moreover,
CE
different period (7/14/21/28 days) SM-induced oxidative stress was found in many areas of brain, including hippocampus [13-15]. It has been suggested that behavioral alterations induced by 7 or
AC
14 days of SM are likely to be associated with cytomorphological changes and loss of neurons in hippocampus of rats [10, 16]. So far, studies of cognitive impairment induced by SM are still relatively small and existing evidences are far from enough to clarify its inherent mechanisms. On the other hand, the effect of microgravity on hippocampus (brain) might be correlated with exposing time although this hasn’t been explored. As the duration of flights has increased, the effect of microgravity, especially long-term microgravity, on the cognitive function and the underlying mechanisms need to be explored imperatively. Therefore, the current study was performed to investigate the effects of long-term exposure to SM environment on the spatial
ACCEPTED MANUSCRIPT 5
learning and memory ability. Specifically, comprehensive analyses of protein expression were
AC
CE
PT E
D
MA
NU
SC
RI
PT
performed to explore the underlying mechanisms of spatial memory deficiency.
ACCEPTED MANUSCRIPT 6
Experimental Procedures Animals. Adult male Sprague-Dawley (SD) rats of SPF grade (8 weeks old, Beijing Vital River Laboratory Animal Technology Co. Ltd. China, Certification No: SCXK 2012-0001) were used in the study. All rats were housed individually in the cages, kept on a 23–25°C room temperature, 12-h light/12-h dark cycle and free access to food and water. All experimental procedures were
PT
approved by China Astronaut Research and Training Center Animal Care and Use Committee, and performed in accordance with the ARRIVE (Animal Research: Reporting of In Vivo Experiments)
RI
guidelines (http://www.nc3rs.org.uk/sites/default/files/documents/Guidelines/ARRIVE%20in%20 mandarin%20v3. pdf).
SC
Tail-suspension model (30°). Rats were randomly divided into 2 groups, including the control group and SM group (8 rats for each group). Tail-suspension rats have been widely used as a
NU
model to simulate microgravity effects [14, 17]. Briefly, the rats were individually housed in a special tail-suspension cage with their tail suspended in the center of the cage and allowed only
MA
the forelimbs to touch the bottom of the cage; and the rat’s body was inclined at approximately 30° from the ground. For the control group, rats were kept exactly at the same condition with the
D
tail-suspensions except their tails do not need to hang up. All animals had free access to water and
PT E
food during entire process. On the 28th day, the rats were sacrificed, and the hippocampus were removed immediately. The samples were placed in liquid nitrogen for later determination. Morris water maze (MWM) test. The MWM was adapted for rats from the prototype originally
CE
described by Morris with some modifications [18, 19]. Briefly, The MWM test was performed after 28 days tail-suspension. The maze consisted of a circular pool filled with water (~24°C) that
AC
was made opaque with non-toxic black pigment. An escape platform was placed in the third quadrant of the maze and 5 cm below the water surface. Colorful, distinguishing, geometric clues with magnetic were affixed at particular visible locations to the rats while in the maze. Rats were trained on a total of 12 trials for 4 days, with 3 trials per session and 1 session per day. Prior to the first training trial, rats were given an ordinary habituation trial without the platform. Trials were 120s and rats that did not find the platform within that time were guided to the destination by the experimenter. Once on the top of the platform, rats were left for an extra 10s before being removed. Start locations (the first, second and fourth quadrant) were pseudo-randomized so that each outset was used fair-minded. On the fifth day, cognitive function was assessed in probe trials
ACCEPTED MANUSCRIPT 7
wherein the platform was removed and rats searched the empty pool for 90s. Behavioral tests were performed during the daytime and recorded by a video camera. iTRAQ(isobaric tags for relative and absolute quantitation)-based proteomics analysis. Protein extraction. Hippocampus sample of each animal was ground into powder in liquid nitrogen and extracted with lysis buffer (8 M urea, 1% DTT, PMSF) and sonicated at 400 W for 30
PT
times (1s for each time). After centrifugation at 4℃, 40000g for 30min, the supernatant was collected and mixed well with 6×volume of chilled acetone and incubated at -20℃ for at least 1h.
RI
After centrifugation at 4℃, 40000g for 5min, the supernatant was discarded, and the precipitate was dissolved with 20l dissolution buffer (iTRAQ Reagent Multi-Plex Kit, Applied Biosystems).
SC
Protein concentrations were quantified by Bradford method (Pierce™ Coomassie (Bradford) Protein Assay Kit, ThermoFisher). Protein processing and iTRAQ labeling. Total protein (100g)
NU
was taken out of each sample and mixed well with 1% SDS (1l), and 2l reducing reagent was added and incubated at 60℃ for 1h. Subsequently, cysteine blocking reagent was added and
MA
incubated for 10min under room temperature, and then digested overnight at 37℃ by trypsin with an enzyme/protein ratio of 1:50. The sample was then labeled with iTRAQ reagent as follows: the
D
control group, labeled with iTRAQ reagent 114 and 115; the tail-suspension group, labeled with
PT E
iTRAQ reagent 116 and 117. First dimensional high-PH RPLC separation. The iTRAQ labelled peptide mixture was fractionated using a high-pH RPLC column from Agela (250×4.6 mm i.d, C18, 5μm). The samples were loaded onto the column in buffer A1 (2% ACN+98% H2O, pH = 10)
CE
and eluted by buffer B1 (98% ACN+2% H2O, pH = 10, flow rate = 0.7 mL/min) with a gradient of 5%-35% B1 for 30min, 35%-95% B1 for 2min, 95% B1 for 5min, 95%-5% B1 for 2min, 5% B1
AC
for 6min. The absorbance at 214 nm was monitored, and a total of 24 fractions were collected and dried by a rotary vacuum concentrator. LC-MS/MS analysis. Each sample was resuspended in buffer A2 (1.9% ACN+98% H2O+0.1% FA) and centrifuged at 12000g for 3min. 10l supernatant was loaded onto an Eksigent Nano LC 2D plus HPLC by the autosampler onto a 5μm C18 trap column (ID100μm, 20mm length). The peptides were then eluted onto a 3μm analytical C18 column (ID75μm, 120mm length) packed in-house. Separation was run at 330 nl/min starting from 5% B2 (98% ACN+1.9% H 2O+0.1% FA), followed by stepwise gradient (8% B2 for 5min, 22% B2 for 34min, 32% B2 for 41min, 90% B2 for 42min), maintenance at 90% B2 for another 46min, and finally a return to 5% B2 for 1min. Data acquisition was performed
ACCEPTED MANUSCRIPT 8
with a TripleTOF™ 5600 mass spectrometer (AB Sciex). Data were acquired using an ion spray voltage of 2.0kV, mass range 300-1400Da, and capillary temperature of 320°C. Raw data files acquired from the TripleTOF 5600 System were converted into MGF files using Proteome Discoverer 1.3 (Thermo Electron, San Jose, CA, USA). Proteins were identified using Mascot server engine (Matrix Science, London, UK; version 2.3.02) uniprot-rat database.
PT
Bioinformatics analysis. To determine possible biological functions of DEPs, Gene Ontology (GO) analysis was performed in biological process, cellular component, and molecular function
RI
using DAVID Bioinformatics Resources. The protein-protein network analysis of the DEPs was performed by the “Analyze Network Algorithm” provided by MetacoreTM sofware. Pathway
identify significantly enriched pathways in DEPs.
SC
maps of MetaCore and PANTHER classification system (http://www.pantherdb.org/) are used to
NU
Western blot. Hippocampus were collected and lysed in RIPA Lysis Buffer (Cat. #C1053). The lysates were incubated for 10min on ice, centrifuged for 10min at 12000g at 4℃, and supernatant
MA
was collected. The total protein concentration was determined using the Pierce○BCA Protein R
Assay Kit (Cat. #OC185139.). The resolved proteins were transferred to a pvdf membrane
D
(Millipore). After blocking in 5% skimmed milk at room temperature for 1h, the membranes were
PT E
incubated in 5% BSA with primary antibodies against GluR1 (CST, #13185, 1:1000), GluR4 (CST, #3824, 1:1000), β-Actin (CST, #3700, 1:1000) at the lysates were subjected to 10% SDS-PAGE
CE
followed by immunoblotting.
Biochemical analysis of neurotransmitters. The level of Glu, GABA, 5-HT, DA, acetylcholine
AC
(Ach), noradrenalin (NE) and E were determined by using LC-MS/MS system (Shimadzu Prominence UFLC connected with an Applied Biosystem 5500 Q-Trap mass detector). Briefly, hippocampus samples were homogenized in 0.1ml precooled normal saline (NS) containing 0.2% formic acid (FA), followed by mixed with 0.2ml ice-cold acetonitrile and incubated at 4℃ for 30min. After centrifuged at 4℃, 12000g for 15min, the supernatant was collected and injected into LC-MS/MS system for analysis. The neurotransmitters and internal standard were detected in multiple reactions monitoring (MRM) mode. The MRM transitions and the related optimized declustering potential (DP), entrance potential (EP), collision energy (CE) and collision cell exit potential (CXP) for the analytes are shown in Table1. Ratios of the peak areas of the analyte vs.
ACCEPTED MANUSCRIPT 9
the internal standard were used to quantify the neurotransmitter concentrations. Statistical analysis. All data were presented as mean ± SEM per experimental condition, and P<0.05 was considered statistically significant. The statistical analyses were performed using SigmaPlot 12.0. The MWM data were evaluated with two-way analysis of the variance (ANOVA) followed by Bonferroni post-test. For all other experiments, statistical significances were tested by
AC
CE
PT E
D
MA
NU
SC
RI
PT
the one-way ANOVA or the Student’s t-test.
ACCEPTED MANUSCRIPT 10
Results Long-term SM impairs spatial learning and memory To evaluate the effects of long-term SM on hippocampus-dependent learning and memory function, we measured 28-day tail-suspension rats using the MWM, a test of spatial learning and memory. The results indicated that both SM and control rats learned the task over this 4 days
PT
training period, manifested by spending less time and swimming shorter distances to find the hidden platform (Figure1.A-B). Interestingly, analysis of latency to platform on day 3 and day 4 of
RI
training showed that SM rats needed more time than control rats to find the hidden platform (p<0.05; Figure1.A). SM rats swam significantly longer distances than control rats to find the
SC
hidden platform on day 3 and day 4 (p<0.05; Figure1.B). In addition, we also found there was no significant difference in the swimming speeds between tail-suspension rats and the control rats
NU
(Figure1.C). In the probe test, 1day after training (day5), SM rats exhibited significantly poorer memory to locate the original position of the removed platform, because they spent less time in
MA
the target Q3 quadrant searching the hidden platform compared with the controls (p<0.05; Figure1.D-E). These data suggest that long-term simulated microgravity may impair spatial
D
learning and memory.
PT E
Overview of proteome changes in hippocampus of SM rats We next investigated the effect of SM on the expression of proteins in hippocampus of rats using iTRAQ-based proteomics analysis. In both control and SM condition, a total of 4774 proteins
CE
were quantified (with FDR<0.01). To determine DEPs between SM rats and control rats, the correlation coefficient between two biological replicates was evaluated firstly. The two controls
AC
(i.e. tags 114 and 115) were used as denominators, respectively. Only the proteins that were quantified with iTRAQ ratios were used to calculate correlation coefficient, and the ratios (SM vs. control) were then log-transformed and plotted against each other. As shown in Figure2, the correlation coefficients between two biological replicates were 0.84 (P<2.2E-16) and 0.87 (P<2.2E-16), thereby indicating the biological reproducibility of SM-regulated protein expression. Further, the biological variations between two replicates were estimated to set an optimal cut-off value of fold change for determining significantly altered differential protein expression levels. A fold change cutoff of ≥1.5 or ≤0.67 was defined as indicating up- or down-regulation, and only proteins, which were identified by three or more peptides and had >1.5-fold change in at least two
ACCEPTED MANUSCRIPT 11
of four comparisons (i.e. tag 116/114, 117/114, 116/115 and 117/115 for SM samples vs. the controls), were considered to be DEPs. The up-regulated and down-regulated DEPs were summarized in Table 2 and Table 3, respectively. For further understanding the mechanisms of spatial learning and memory impairment induced by SM, Gene Ontology (GO) classification and pathway enrichment of DEPs were performed. The
PT
result indicated that 28 days of SM might affect the metabolism function, cytoskeleton system, synaptic transmission, apoptosis, etc. According to GO classification, the DEPs involved in
RI
oxidative phosphorylation (GO: 0006119), generation of precursor metabolites and energy (GO: 0006091), negative regulation of cytoskeleton organization (GO: 0051494), regulation of action
SC
potential in neuron (GO: 0019228), regulation of cytoskeleton organization (GO: 0051493), negative regulation of actin filament polymerization (GO: 0030837), regulation of action potential
NU
(GO: 0001508), transmission of nerve impulse (GO: 0019226), regulation of membrane potential (GO: 0042391), regulation of synaptic transmission (GO: 0050804) and other biological processed
MA
(Table 4, more details see supplemental material). DEPs were mainly located in proton-transporting two-sector ATPase complex (GO: 0016469), proton-transporting V-type
D
ATPase complex (GO: 0033176), respiratory chain (GO: 0070469), mitochondrion (GO:
PT E
0005739), cytoskeleton (GO: 0005856), cytoskeletal part (GO: 0044430), microtubule (GO: 0005874), intermediate filament cytoskeleton (GO: 0045111), microtubule associated complex (GO: 0005875), voltage-gated sodium channel complex (GO: 0001518) (Table 5, more details see
CE
supplemental material). DEPs in molecular function are embodied in ATPase binding (GO: 0051117), ATPase activity (GO: 0016887), voltage-gated sodium channel activity (GO: 0005248)
AC
(Table 6, more details see supplemental material). The MetaCore pathway map analysis of DEPs was used to identify the 10 most statistically significant pathways, based on calculated P values (Table 7). As shown in Table 7, the DEPs involved in oxidative phosphorylation, ATP metabolism, and ATP/ITP metabolism. Moreover, two of ten MetaCore pathways were found to be directly associated with neurophysiological process, which is the neurophysiological process GABA-A receptor life cycle and neurophysiological process_dynein-dynactin motor complex in axonal transport in neurons (Figure 3A&B.). Additionally, protein-protein interactions were noted among all the DEPs (Figure 3C). SM exposure increases GluR1 and GluR4 expression in hippocampus
ACCEPTED MANUSCRIPT 12
Glutamate receptors (GluRs) play an important role in synaptic plasticity (long-term potentiation & long-term depression), which is directly involved in the process of learning and memory [20, 21]. The memory impairment observed in tail-suspension rats might be related with abnormal expression of GluRs, at least in part. Firstly, based on the preliminary PANTHER pathway analysis, DEPs were significantly enriched in metabotropic glutamate receptor group III pathway
PT
(P00039) and ionotropic glutamate receptor pathway (P00037) (Table 8). Secondly, the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid selective glutamate receptor complex
RI
(GO: 0032281) were found to be enriched in cellular component of DEPs (Table 5.). We therefore validated the expression of P19490 (Gria1 or GluR1) and P19493 (Gria4 or GluR4) which were
SC
functionally involved in metabotropic glutamate receptor group III pathway and ionotropic glutamate receptor pathway (Table 8) by western blot. Considered with the result of proteome, the
NU
higher levels of GluR1 and GluR4 were observed in hippocampus after 28 days of tail-suspension (Figure 4).
MA
SM exposure disturbs neurotransmitter release
Next, the concentrations of Glu, 5-HT, DA, GABA, Ach, E and NE in SM group and control
D
group were analyzed (Figure 5). This metabolites analysis was carried out using student-t tests. A
PT E
significant difference was observed after exposure to SM for 28 days, suggesting that exposure to SM may lead to neurotransmitters variations in rat hippocampus. As shown in Figure 5A, the concentration of Glu was significantly up-regulated after SM exposure (p<0.05) (Figure 5A). On
CE
the contrary, concentrations of 5-HT, DA, GABA, and E were significantly down-regulated by SM (p<0.05) (Figure 5B-E). In addition, there is non-significant change in concentrations of Ach and
AC
NE (Figure 5F-G).
ACCEPTED MANUSCRIPT 13
Discussion The investigation of the physiological and pathological changes induced by spaceflight or microgravity will be critical in developing countermeasures to ensure astronauts to complete spaceflight mission effectively and return to earth safely. It has been demonstrated the spaceflight may lead to impairments in cognitive function [3, 22, 23]. Human cognitive performance is an
PT
important factor for the success of the spaceflight mission. How microgravity affects cognitive function need to be investigated since highly effective countermeasures might be required.
RI
In present study, 30° tail-suspension rat model was typically employed to simulate microgravity, and we observed 28-day tail-suspension induced spatial learning and memory deficit in rats during
SC
MWM test. Spatial memory refers to the part of the memory system that encodes stores, recognizes and recalls spatial information about one's environment and spatial orientation. The
NU
MWM was adapted for rats from the prototype formerly described by Morris[18]. It was also reported that 14 days of SM significantly decreased the escape latency and swimming distance of
MA
rats compared with the controls[24], suggesting that SM might lead to impairment in spatial learning and memory. This finding is consistent with our present study as shown in Figure 1A&B.
D
Swimming speed is related to escape latency and swimming distance. It was established that SM
PT E
model (rat hindlimb uploading) causes rat hindlimb muscle atrophy[25]. In order to eliminate the interference of muscle atrophy on swimming in tail-suspension rats, we therefore compared the swimming speed of the two groups, so as to make more accurate evaluation of spatial learning and
CE
memory ability. Our results showed there was no significant change in swimming speed after 28 days of SM exposure, indicating the muscle atrophy of rat hindlimb could not disturb the results of
AC
the escape latency and swimming distance during MWM test . Hippocampus is closely related to spatial learning and memory function in the brain. We therefore investigate the effect of 28 days SM on the protein expression in hippocampus. Based on iTRAQ-based proteomics analysis, 75 proteins were found overexpressed and 72 proteins were found lower expressed in hippocampus of rats after 28 days of SM exposure in comparison to the controls (Table 2&3). Learning and memory are encoded within the mammalian brain as biochemical and physical changes at synapses that alter synaptic transmission, a process known as synaptic plasticity. Synaptic plasticity could be regulated by altering the efficacy of neurotransmitter release or by changing the numbers and properties of neurotransmitter
ACCEPTED MANUSCRIPT 14
receptors[26]. Therefore, neurotransmitters and its specific receptors mediated pathways play an essential role in the processes of learning and memory [27-29]. Disturbance of neurotransmitter metabolism plays an important role in cognitive deficits induced by variety of neurological diseases or noxious stimuli [28-33]. Further bioinformatics analysis of proteomics data showed many DEPs were specifically associated with synaptic transmission, and the results of LC-MS/MS displayed the concentration of Glu was significantly increased after SM exposure while the
PT
concentrations of 5-HT, DA, GABA, and E were significantly decreased. These results suggested
RI
that spatial learning and memory impairment induced by 28 days of SM might be related to specific neurotransmitters level.
SC
Glu acting on Amino-acid-3-hydroxy-5-methyl-isoxazol-4-propionic acid (AMPA) receptors mediates most fast excitatory synaptic transmission in the brain [26, 34, 35]. Regulating the
NU
numbers or properties of AMPA receptors is crucial to excitatory synaptic transmission and the consequent formation of appropriated neural connections during learning and memory[36].
MA
Normally, Glu released into the synapse binds to its GluRs, which results in induction of long-term potentiation (LTP) and synaptic strengthening, and then contributes to learning and
D
memory formation. However, excessive production and release of Glu was reported to cause
PT E
excitotoxic cell death due to overstimulation GluRs [37-40]. AMPA receptor is a hetero-oligomeric protein complex consisting of combinations of four different kinds of subunits GluR1- GluR4, also known as GluRA-GluRD, each encoded by a separate gene: gria1-gria4[41,
CE
42]. The important role of AMPA on learning and memory has been studied in numerous studies [21, 43-45]. As well, the excessive activation of AMPA receptors has been demonstrated to be
AC
potently toxic for the brain [46, 47], including GluR1 and GluR4 [42, 48, 49]. In present study, we found the expression of GluR1 and GluR4 were significantly up-regulated by SM exposure (Figure 4). Excitotoxicity is a major cause of neuronal death in numerous neurological disorders including ischemia, traumatic brain injury and neurodegenerative disease [39, 50, 51]. Studies have reported that increased apoptosis can be found in hippocampus after 7 or 14 days of SM [10, 16]. We previously investigated the effects of 28-day SM on neuronal morphology, expression of apoptosis-related molecules in rat hippocampus. The results indicated that the numbers of hippocampal neurons were decreased after 28 days of simulated microgravity compared with the controls, accompanied by elevated expression of apoptosis-related proteins[52]. Furthermore, the
ACCEPTED MANUSCRIPT 15
pathways analysis in the present study showed the DEPs exerted functions in apoptosis signaling pathway (P00006) (Table 8). All these results implied that 28 days of SM exposure might result in excitotoxicity in hippocampus, which might be one of mechanisms of cognitive dysfunction induced by SM. DEPs were involved in a wide range of biological processes, suggesting the impact of SM on
PT
learning and memory was complex and multifaceted. The malfunction of other biological process may also be involved in learning and memory processes during SM exposure. The cytoskeleton is
RI
the main structure giving shape and mechanical strength to cells. It is now widely accepted that cytoskeleton plays a role in sensing changes in gravity[53]. Cytoskeleton changes induced by
SC
microgravity were found in various cell types, including hippocampus neuron cell [11, 53]. Cytoskeleton is associated with the changes of cell shape, function and signaling. Emerging
NU
evidence suggested that various high level brain functions depend on the generation of neuronal networks involved the establishment, maintenance, and repairing of synaptic contacts, such as
MA
cognitive function [54-56]. Neuronal cytoskeleton is not only required for establishment and maintenance of synaptic contacts but also provides the structural basis for synaptic transmission
D
[54-56]. In addition, disruption of synaptic function has been implicated in a wide range of
PT E
neurodevelopmental and neurodegenerative diseases, such as AD [54-56]. In present study, based on GO cellular component analysis, most DEPs were located in cytoskeleton, such as cytoskeleton (GO: 0005856), cytoskeletal part (GO: 0044430), and microtubule (GO: 0005874), et al. More
CE
details see Figure 5 and supplemental material. These results imply that hippocampus dysfunction induced by SM may be related to the SM-associated cytoskeleton changes.
AC
In summary, our data demonstrated that long-term SM exposure impaired the spatial learning and memory function. Based on proteomics analysis, a total of 147 proteins were found to be differentially expressed in 28-day SM rats’ hippocampus. GO function ontology classification indicated that DEPs were enriched in 41 GO biological processes, 11 GO molecular functions, 25 GO cellular component and 16 PANTHER pathways. It was also shown the expression of GluR1 and GluR4 were up-regulated, and the level of Glu was increased, while the concentration of 5-HT, DA, GABA, and E were declined after28 days of SM exposure. All of these results suggest that SM exposure may lead to comprehensive biological effects on hippocampus. The impairment of learning and memory function induced by SM is a complicated process, and one of the possible
ACCEPTED MANUSCRIPT 16
mechanisms might be related with glutamate excitotoxicity and imbalances in specific
AC
CE
PT E
D
MA
NU
SC
RI
PT
neurotransmitters.
ACCEPTED MANUSCRIPT 17
Acknowledgements: This work was supported by the National Major Scientific Instrument and Equipment Development Project (2013YQ19046706, 2012YQ0401400901); Foundation of China Space Medicine Engineering Advanced Research (2015SY54A0501); National Natural Science Fund (31401010); Shenzhen Science & Technology Institute (20150629164441050); and Foundation of
PT
State Key Laboratory of Space Medicine Fundamentals and Application (SMFA15B01). Conflict of Interest:
AC
CE
PT E
D
MA
NU
SC
RI
We declare that we have no conflict of interest.
ACCEPTED MANUSCRIPT 18
References [1] Eddy DR, Schiflett SG, Schlegel RE, Shehab RL. Cognitive performance aboard the life and microgravity spacelab. Acta astronautica. 1998;43:193-210. [2] Wang Y, Iqbal J, Liu Y, Su R, Lu S, Peng G, et al. Effects of simulated microgravity on the expression of presynaptic proteins distorting the GABA/glutamate equilibrium--A proteomics approach. Proteomics. 2015;15:3883-91. [3] Manzey D, Lorenz B. Mental performance during short-term and long-term spaceflight. Brain research Brain research reviews. 1998;28:215-21.
PT
[4] Santucci D, Kawano F, Ohira T, Terada M, Nakai N, Francia N, et al. Evaluation of gene, protein and neurotrophin expression in the brain of mice exposed to space environment for 91 days. PloS one. 2012;7:e40112.
RI
[5] Reschke MF, Bloomberg JJ, Harm DL, Paloski WH, Layne C, McDonald V. Posture, locomotion, spatial orientation, and motion sickness as a function of space flight. Brain research Brain research
SC
reviews. 1998;28:102-17.
[6] Lackner JR, DiZio P. Motor function in microgravity: movement in weightlessness. Current opinion in neurobiology. 1996;6:744-50.
NU
[7] Kornilova LN. Vestibular function and sensory interaction in altered gravity. Advances in space biology and medicine. 1997;6:275-313.
[8] Bock O, Abeele S, Eversheim U. Sensorimotor performance and computational demand during
MA
short-term exposure to microgravity. Aviat Space Environ Med. 2003;74:1256-62. [9] Liu Q, Zhou R, Zhao X, Oei TP. Effects of prolonged head-down bed rest on working memory. Neuropsychiatric disease and treatment. 2015;11:835-42. [10] Sun XQ, Xu ZP, Zhang S, Cao XS, Liu TS. Simulated weightlessness aggravates
D
hypergravity-induced impairment of learning and memory and neuronal apoptosis in rats.
PT E
Behavioural brain research. 2009;199:197-202. [11] Sarkar P, Sarkar S, Ramesh V, Hayes BE, Thomas RL, Wilson BL, et al. Proteomic analysis of mice hippocampus in simulated microgravity environment. Journal of proteome research. 2006;5:548-53. [12] Wang Y, Javed I, Liu Y, Lu S, Peng G, Zhang Y, et al. Effect of Prolonged Simulated Microgravity on
CE
Metabolic Proteins in Rat Hippocampus: Steps toward Safe Space Travel. Journal of proteome research. 2016;15:29-37.
[13] Chen HL, Qu LN, Li QD, Bi L, Huang ZM, Li YH. Simulated microgravity-induced oxidative stress in
AC
different areas of rat brain. Sheng li xue bao : [Acta physiologica Sinica]. 2009;61:108-14. [14] Sarkar S, Wise KC, Manna SK, Ramesh V, Yamauchi K, Thomas RL, et al. Activation of activator protein-1 in mouse brain regions exposed to simulated microgravity. In vitro cellular & developmental biology Animal. 2006;42:96-9. [15] Wise KC, Manna SK, Yamauchi K, Ramesh V, Wilson BL, Thomas RL, et al. Activation of nuclear transcription factor-kappaB in mouse brain induced by a simulated microgravity environment. In vitro cellular & developmental biology Animal. 2005;41:118-23. [16] Ranjan A, Behari J, Mallick BN. Cytomorphometric Changes in Hippocampal CA1 Neurons Exposed to Simulated Microgravity Using Rats as Model. Frontiers in neurology. 2014;5:77. [17] Wang J, Liu C, Li T, Wang Y, Wang D. Proteomic analysis of pulmonary tissue in tail-suspended rats under simulated weightlessness. Journal of Proteomics. 2012;75:5244-53. [18] Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. Journal of neuroscience methods. 1984;11:47-60.
ACCEPTED MANUSCRIPT 19
[19] Qiong W, Yong-Liang Z, Ying-Hui L, Shan-Guang C, Jiang-Hui G, Yi-Xi C, et al. The memory enhancement effect of Kai Xin San on cognitive deficit induced by simulated weightlessness in rats. Journal of ethnopharmacology. 2016;187:9-16. [20] Riedel G, Reymann KG. Metabotropic glutamate receptors in hippocampal long ‐ term potentiation and learning and memory. Acta Physiologica Scandinavica. 1996;157:1-19. [21] Riedel G, Platt B, Micheau J. Glutamate receptor function in learning and memory. Behavioural brain research. 2003;140:1-47. [22] Bock O, Weigelt C, Bloomberg JJ. Cognitive demand of human sensorimotor performance during an extended space mission: a dual-task study. Aviat Space Environ Med. 2010;81:819-24.
PT
[23] Demertzi A, Van Ombergen A, Tomilovskaya E, Jeurissen B, Pechenkova E, Di Perri C, et al. Cortical reorganization in an astronaut's brain after long-duration spaceflight. Brain structure &
RI
function. 2015.
[24] Qiong W, Yong-liang Z, Ying-hui L, Shan-guang C, Jiang-hui G, Yi-Xi C, et al. The memory
SC
enhancement effect of Kai Xin San on cognitive deficit induced by simulated weightlessness in rats. Journal of ethnopharmacology. 2016;187:9-16.
[25] Il'ina-Kakueva EI, Kaplanskii AS. Muscle atrophy in microgravity and during its simulation.
NU
Aviakosmicheskaia i ekologicheskaia meditsina = Aerospace and environmental medicine. 2005;39:43-9.
[26] Anggono V, Huganir RL. Regulation of AMPA receptor trafficking and synaptic plasticity. Current
MA
opinion in neurobiology. 2012;22:461-9.
[27] Peng S, Zhang Y, Zhang J, Wang H, Ren B. Glutamate receptors and signal transduction in learning and memory. Molecular biology reports. 2011;38:453-60. [28] Nieoullon A. Dopamine and the regulation of cognition and attention. Progress in neurobiology.
D
2002;67:53-83.
[29] Rossetti ZL, Carboni S. Noradrenaline and dopamine elevations in the rat prefrontal cortex in
PT E
spatial working memory. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2005;25:2322-9.
[30] Meeter M, Talamini L, Schmitt JA, Riedel WJ. Effects of 5-HT on memory and the hippocampus: model and data. Neuropsychopharmacology : official publication of the American College of
CE
Neuropsychopharmacology. 2006;31:712-20. [31] Haider S, Khaliq S, Haleem DJ. Enhanced serotonergic neurotransmission in the hippocampus following tryptophan administration improves learning acquisition and memory consolidation in rats.
AC
Pharmacological reports : PR. 2007;59:53-7. [32] Haider S, Naqvi F, Batool Z, Tabassum S, Perveen T, Saleem S, et al. Decreased Hippocampal 5-HT and DA Levels Following Sub-Chronic Exposure to Noise Stress: Impairment in both Spatial and Recognition Memory in Male Rats. Scientia pharmaceutica. 2012;80:1001-11. [33] Ray RS, Rai S, Katyal A. Cholinergic receptor blockade by scopolamine and mecamylamine exacerbates global cerebral ischemia induced memory dysfunction in C57BL/6J mice. Nitric Oxide. 2014;43:62-73. [34] Rossner B, Klingler M, Bulat T, Sase A, Zeilinger A, Spitzwieser M, et al. Hippocampal GluA2 and GluA4 protein but not corresponding mRNA and promoter methylation levels are modulated at retrieval in spatial learning of the rat. Amino acids. 2017;49:117-27. [35] Han J, Wu P, Wang F, Chen J. S-palmitoylation regulates AMPA receptors trafficking and function: a novel insight into synaptic regulation and therapeutics. Acta pharmaceutica Sinica B. 2015;5:1-7.
ACCEPTED MANUSCRIPT 20
[36] Hanley JG. Subunit-specific trafficking mechanisms regulating the synaptic expression of Ca2+-permeable AMPA receptors. Seminars in Cell & Developmental Biology. 2014;27:14-22. [37] Fachim HA, Pereira AC, Iyomasa-Pilon MM, Rosa ML. Differential Expression of AMPA Subunits Induced by NMDA Intrahippocampal Injection in Rats. Frontiers in neuroscience. 2016;10:32. [38] Lowry ER, Kruyer A, Norris EH, Cederroth CR, Strickland S. The GluK4 kainate receptor subunit regulates memory, mood, and excitotoxic neurodegeneration. Neuroscience. 2013;235:215-25. [39] Hoque A, Hossain MI, Ameen SS, Ang C-S, Williamson N, Ng DCH, et al. A beacon of hope in stroke therapy—Blockade of pathologically activated cellular events in excitotoxic neuronal death as potential neuroprotective strategies. Pharmacology & therapeutics. 2016;160:159-79.
PT
[40] Hota SK, Barhwal K, Ray K, Singh SB, Ilavazhagan G. Ceftriaxone rescues hippocampal neurons from excitotoxicity and enhances memory retrieval in chronic hypobaric hypoxia. Neurobiol Learn
RI
Mem. 2008;89:522-32.
[41] Sanderson DJ, Good MA, Seeburg PH, Sprengel R, Rawlins JN, Bannerman DM. The role of the
SC
GluR-A (GluR1) AMPA receptor subunit in learning and memory. Progress in brain research. 2008;169:159-78.
[42] Santos AE, Duarte CB, Iizuka M, Barsoumian EL, Ham J, Lopes MC, et al. Excitotoxicity mediated death and differentiation. 2006;13:652-60.
NU
by Ca2+-permeable GluR4-containing AMPA receptors involves the AP-1 transcription factor. Cell [43] Sanderson DJ, Bannerman DM. The role of habituation in hippocampus-dependent spatial
MA
working memory tasks: evidence from GluA1 AMPA receptor subunit knockout mice. Hippocampus. 2012;22:981-94.
[44] Timofeeva OA, Levin ED. Glutamate and nicotinic receptor interactions in working memory: importance for the cognitive impairment of schizophrenia. Neuroscience. 2011;195:21-36.
D
[45] Genoux D, Montgomery JM. Glutamate receptor plasticity at excitatory synapses in the brain. Clinical and experimental pharmacology & physiology. 2007;34:1058-63.
PT E
[46] Simões PF, Silva AP, Pereira FC, Marques E, Grade S, Milhazes N, et al. Methamphetamine induces alterations on hippocampal NMDA and AMPA receptor subunit levels and impairs spatial working memory. Neuroscience. 2007;150:433-41. [47] Trudeau F, Gagnon S, Massicotte G. Hippocampal synaptic plasticity and glutamate receptor
CE
regulation: influences of diabetes mellitus. European journal of pharmacology. 2004;490:177-86. [48] Yu Z, Cheng G, Wen X, Wu GD, Lee WT, Pleasure D. Tumor necrosis factor alpha increases neuronal vulnerability to excitotoxic necrosis by inducing expression of the AMPA-glutamate receptor
AC
subunit GluR1 via an acid sphingomyelinase- and NF-kappaB-dependent mechanism. Neurobiology of disease. 2002;11:199-213. [49] Lu C, Fu W, Salvesen GS, Mattson MP. Direct cleavage of AMPA receptor subunit GluR1 and suppression of AMPA currents by caspase-3: implications for synaptic plasticity and excitotoxic neuronal death. Neuromolecular medicine. 2002;1:69-79. [50] Connolly NM, D'Orsi B, Monsefi N, Huber HJ, Prehn JH. Computational Analysis of AMPK-Mediated Neuroprotection Suggests Acute Excitotoxic Bioenergetics and Glucose Dynamics Are Regulated by a Minimal Set of Critical Reactions. PloS one. 2016;11:e0148326. [51] Wang Y, Qin ZH. Molecular and cellular mechanisms of excitotoxic neuronal death. Apoptosis : an international journal on programmed cell death. 2010;15:1382-402. [52] Ting-mei W, Yong-liang Z, Yan-li W, Hai-long C, Gou-hua J, Ke L, et al. Effects of simulated microgravity on the memory and apoptosis related protein expression in rat hippocampus. . Space
ACCEPTED MANUSCRIPT 21
Medicine& Medical Engineering. 2016;29:235-9. [53] Vorselen D, Roos WH, MacKintosh FC, Wuite GJ, van Loon JJ. The role of the cytoskeleton in sensing changes in gravity by nonspecialized cells. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2014;28:536-47. [54] Stefen H, Chaichim C, Power J. Regulation of the Postsynaptic Compartment of Excitatory Synapses by the Actin Cytoskeleton in Health and Its Disruption in Disease. 2016;2016:2371970. [55] Kevenaar JT, Hoogenraad CC. The axonal cytoskeleton: from organization to function. Neural plasticity. 2015;8:44. [56] Bodaleo FJ, Gonzalez-Billault C. The Presynaptic Microtubule Cytoskeleton in Physiological and
PT
Pathological Conditions: Lessons from Drosophila Fragile X Syndrome and Hereditary Spastic
AC
CE
PT E
D
MA
NU
SC
RI
Paraplegias. Frontiers in molecular neuroscience. 2016;9:60.
ACCEPTED MANUSCRIPT 22
Figure legends: Figure1. Tail-suspension rats show spatial learning and memory deficits in the Morris water maze test. A, Escape latencies of tail-suspension and control rats. B, Swimming distance of tail-suspension and control rats. C, swimming speed did not differ among the SM and control group during the four sessions. D, Path length in target quadrant (%) of tail-suspension and control
AC
CE
PT E
D
MA
NU
SC
RI
tail-suspension and control rats in the probe tests. (n=6, *p<0.05).
PT
rats. E, Representative Morris water maze program generated-swimming tracing pattern of
ACCEPTED MANUSCRIPT 23
Figure2. Correlation coefficients of biological replicates. The ratios of quantified proteins were
AC
CE
PT E
D
MA
NU
SC
RI
PT
log-transformed and plotted between two biological replicates.
ACCEPTED MANUSCRIPT 24
Figure3. Overview of proteomics in hippocampus of SM rats. A, Neurophysiological process: dynein-dynactin motor complex in axonal transport in neurons. B, Neurophysiological process:
AC
CE
PT E
D
MA
NU
SC
RI
PT
GABA-A receptor life cycle. C, Protein-protein interaction network of the DEPs.
ACCEPTED MANUSCRIPT 25
Figure4. GluR1 and GluR4 expression in rat hippocampus was induced by SM. A, GluR1 and GluR4 level in hippocampus of control and SM rats was quantified by western blot normalized to β-actin level (GluR1 180KD, GluR4 200 KD, β-actin 40KD). B&C. Quantitative analysis of
AC
CE
PT E
D
MA
NU
SC
RI
PT
western blot (n=4, *p<0.05, **p<0.01).
ACCEPTED MANUSCRIPT 26
Figure5. Contents of neurotransmitters in SM and control groups. A. Increases in the content of Glu under SM (g/g). B. Decreases in the content of GABA under SM (g/g). C. Decreases in the content of 5-HT under SM (ng/g). D. Decreases in the content of DA under SM (ng/g). E. Decreases in the content of epinephrine under SM (ng/g). F. non-significant change in the content of Ach under SM (ng/g). G. non-significant change in the content of noradrenaline under SM
AC
CE
PT E
D
MA
NU
SC
RI
PT
(ng/g). (n=5,*p<0.05, **p<0.01).
ACCEPTED MANUSCRIPT 27
Table 1. Multiple reaction monitoring (MRM) transitions, DP (declustering potential), EP (entrance potential), CE (collision energy) and CXP (collision cell exit potential) of the analytes. Q1 → Q3 (m/z)
DP(V)
EP(V)
CE(eV)
CXP(V)
DA E NE 5-HT Ach GABA Glu
154 → 137 184 → 166 170 → 152 177 → 160 146 → 87 104 → 87 148 → 84
30 40 95 65 160 20 20
12 10 12 12 10 10 10
15 12 15 10 20 25 40
40 15 10 8 30 8 9
AC
CE
PT E
D
MA
NU
SC
RI
PT
Compounds
ACCEPTED MANUSCRIPT 28
Table 2. Up-regulated proteins identified by iTRAQ technology after 28 days of SM exposure in hippocampus. Accession
Protein names Mannose-P-dolichol utilization defect 1
D3Z8Z7
Protein Ankdd1a (Fragment)
D3ZM97
Olfactory receptor
D3ZZQ8
Protein RGD1561381
E9PSV8
Neuronal membrane glycoprotein M6-b
F1LZG6
Protein Lrba (Fragment)
M0R451
Glyceraldehyde-3-phosphate dehydrogenase
M0R8M1
Uncharacterized protein (Fragment)
M0R907
Protein Snrpd3
P19493
Glutamate receptor 4
P35467
Protein S100-A1
P54900
Sodium channel subunit beta-2
P63081
V-type proton ATPase 16 kDa proteolipid subunit
P63155
Crooked neck-like protein 1
P80431
Cytochrome c oxidase subunit 7B, mitochondrial
Q1HL14
Longevity assurance-like protein 1
Q5FVN2
Transmembrane protein 41B
Q66H18
Protein Sypl1
Q6IRG7
Claudin
Q7M730
Sodium channel subunit beta-4
Q7TNK0
Serine incorporator 1
Q80W89
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 11
D4AC70
Procollagen, type VIII, alpha 1 (Predicted)
F1LNN1
Proteasome subunit beta type
F1MAM6
Protein Dnah8
M0R5G5
Protein Pkd1l2
P00159
RI SC
NU
MA
D
PT E
CE
O54715
PT
D3Z865
V-type proton ATPase subunit S1 Cytochrome b Solute carrier family 2, facilitated glucose transporter member 1
AC
P11167 Q05140
Clathrin coat assembly protein AP180
Q09426
2-hydroxyacylsphingosine 1-beta-galactosyltransferase
Q64244
ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1
A0A0A0MY37
Leukocyte surface antigen CD47
D3ZPU3
Estradiol 17-beta-dehydrogenase 12
D3ZXG2
Protein Armc4
D4ABE8
Uncharacterized protein
D4ABI7
Protein LOC100365676
D4ABK1
Protein Syngr3
D4ADV4
Sodium-driven chloride bicarbonate exchanger
F1LMZ4
Ribosome-releasing factor 2, mitochondrial
F1LPA7
Tetraspanin (Fragment)
ACCEPTED MANUSCRIPT 29
Continued Table 2 Accession
Protein names Choline transporter-like protein 1 (Fragment)
F1LRV4
Heat shock 70 kDa protein 4
F1M9V3
Enolase (Fragment)
G3V879
Ubiquinone biosynthesis protein COQ7 homolog
G3V8Y3
Kinesin-like protein
G3V912
Protein Tmx4
G3V9W6
Aldehyde dehydrogenase
M0R8J4
Protein Wdr87 (Fragment)
M0R9I6
Aminomethyltransferase
M0RBE5
Neuritin (Fragment)
O35795
Ectonucleoside triphosphate diphosphohydrolase 2
P01048
T-kininogen 1
P05508
NADH-ubiquinone oxidoreductase chain 4
P09006
Serine protease inhibitor A3N
P13852
Major prion protein
P19490
Glutamate receptor 1
P34926
Microtubule-associated protein 1A
P38650
Cytoplasmic dynein 1 heavy chain 1
P47863
Aquaporin-4
P61805
Dolichyl-diphosphooligosaccharide--protein glycosyltransferase subunit DAD1
Q3SWT7
Nuclear receptor binding protein
Q498C8
Protein RER1
Q4QQW8
Putative phospholipase B-like 2
Q568Z4
Signal peptidase complex subunit 3
Q5EBD0
SEC14-like 2 (S. cerevisiae)
Q5U2M9
Protein Rfx3
Q63910
Alpha globin
Q68FW4
RI
SC
NU
MA
D
PT E
Protein Samd8 Syntaxin-18 Biliverdin reductase A
AC
Q6AZ33
CE
Q641X0
PT
F1LPF3
Q6EIX2
Mitochondrial import inner membrane translocase subunit TIM16
Q8CFD0
Sideroflexin-5
Q8SEZ0
NADH-ubiquinone oxidoreductase chain 5
Q99MS0
SEC14-like protein 2
ACCEPTED MANUSCRIPT 30
Table 3. Down regulated proteins identified by iTRAQ technology after 28 days of SM exposure in hippocampus. Accession
Protein names Protein Fsip2
A0JN27
General transcription factor IIH subunit 2
B0K010
Protein Txndc17
B2GUV5
ATPase, H transporting, lysosomal V1 subunit G1
D3ZPI7
Glyceraldehyde-3-phosphate dehydrogenase
D3ZS15
Uncharacterized protein
D3ZWL9
Protein Dido1
D4A6P0
Protein LOC100359747
D4A783
Uncharacterized protein
D4A7I6
Protein RGD1309995
D4A7Q3
Protein LOC685888
E9PTN4
Protein Srpk1 (Fragment)
F1LMQ2
Farnesyl pyrophosphate synthase
F1LPU4
Choline O-acetyltransferase
F1LWR6
Protein Mlip (Fragment)
RI SC
NU
Positive cofactor 2, multiprotein complex, glutamine/Q-rich-associated protein (Predicted), isoform CRA_a
MA
G3V684
PT
A0A096MJX4
Uncharacterized protein (Fragment)
O35314
Secretogranin-1
P02625
Parvalbumin alpha
P19132
Ferritin heavy chain
P60841
Alpha-endosulfine
P62329
Thymosin beta-4
Q03344
ATPase inhibitor, mitochondrial
Q1KQ07
Signal transducer and activator of transcription
Q505I4
UPF0235 protein C15orf40 homolog
Q5RKI9
Protein FRA10AC1 homolog Ribosome-recycling factor, mitochondrial 28 kDa heat- and acid-stable phosphoprotein
AC
Q62785
CE
Q5FVF1
PT E
D
M0RCM1
Q64350
Translation initiation factor eIF-2B subunit epsilon
Q66H40
High mobility group nucleosome-binding domain-containing protein 3
Q6AZ25
Tropomyosin 1, alpha
Q6P756
Adaptin ear-binding coat-associated protein 2
Q925G0
RNA-binding protein 3
Q9EPJ0
Nuclear ubiquitous casein and cyclin-dependent kinase substrate 1
Q9WV63
Kinesin-like protein KIF2A
B1WBQ7
DNA mismatch repair protein Msh2
D3ZC13
Olfactory receptor
D3ZFJ6
Lactamase, beta (Predicted)
D3ZUZ5
Ferritin
D4AB12
Uncharacterized protein
ACCEPTED MANUSCRIPT 31
Continued Table 3 Accession
Protein names Angel homolog 2 (Drosophila) (Predicted), isoform CRA_a
F1M9F6
Protein Gbp5
F1M9F9
Jouberin
G3V8R0
Protein RGD1311703
M0RBL8
Protein Tceal6
P62634
Cellular nucleic acid-binding protein
Q63638
Striated muscle-specific serine/threonine-protein kinase
Q6IG03
Keratin, type II cytoskeletal 73
Q6J2U6
E3 ubiquitin-protein ligase RNF114
Q8K3F3
Protein phosphatase 1 regulatory subunit 14B
B1WC50
Ewsr1 protein
D4ADP2
Protein Ube2q2l
F1LSK5
Protein Heatr5a
F1LUC1
Protein Fam13c
F1M208
Protein Piezo2
F1M2K7
Enolase (Fragment)
F1MAQ5
Microtubule-associated protein
F7FC39
Protein Spta1
M0RCH6
Protein Chmp4b
O35820
2'-deoxynucleoside 5'-phosphate N-hydrolase 1
P15865
Histone H1.4
P35745
Acylphosphatase-2
P46844
Biliverdin reductase A
P50617
Dendrin
Q4V898
RNA-binding motif protein, X chromosome
Q6ED65
Echinoderm microtubule-associated protein-like 5
Q6IFW6
Keratin, type I cytoskeletal 10
Q6IG04
Keratin, type II cytoskeletal 2 epidermal Keratin, type II cytoskeletal 72 FK506-binding protein-like
AC
Q6MG81
CE
Q6IG02
PT E
D
MA
NU
SC
RI
PT
F1LT13
Q76MV3
Cytochrome C oxidase assembly protein COX17
Q9Z339
Glutathione S-transferase omega-1
ACCEPTED MANUSCRIPT 32
%
PValue
GO:0045785~positive regulation of cell adhesion
4.31
6.96E-04
GO:0030155~regulation of cell adhesion
5.17
1.18E-03
GO:0051494~negative regulation of cytoskeleton organization
3.45
2.84E-03
GO:0010639~negative regulation of organelle organization
3.45
8.33E-03
GO:0019228~regulation of action potential in neuron
3.45
9.86E-03
GO:0006119~oxidative phosphorylation
3.45
1.15E-02
GO:0006811~ion transport
9.48
1.24E-02
7.76
1.58E-02
6.90
1.69E-02
3.45
1.70E-02
2.59
1.78E-02
2.59
2.35E-02
6.90
3.00E-02
3.45
3.55E-02
6.03
3.56E-02
GO:0043933~macromolecular complex subunit organization
7.76
3.62E-02
GO:0055082~cellular chemical homeostasis
6.03
3.76E-02
5.17
3.93E-02
2.59
4.01E-02
2.59
4.01E-02
GO:0015672~monovalent inorganic cation transport
5.17
4.30E-02
GO:0048878~chemical homeostasis
6.90
4.39E-02
4.31
4.41E-02
GO:0051129~negative regulation of cellular component organization
3.45
5.09E-02
GO:0000041~transition metal ion transport
2.59
5.46E-02
GO:0044271~nitrogen compound biosynthetic process
5.17
6.67E-02
GO:0065003~macromolecular complex assembly
6.90
6.80E-02
GO:0051249~regulation of lymphocyte activation
3.45
7.01E-02
GO:0009150~purine ribonucleotide metabolic process
3.45
7.12E-02
GO:0006091~generation of precursor metabolites and energy
4.31
7.28E-02
GO:0015992~proton transport
2.59
7.63E-02
GO:0009259~ribonucleotide metabolic process
3.45
7.71E-02
GO:0006818~hydrogen transport
2.59
7.82E-02
GO:0042391~regulation of membrane potential
3.45
8.08E-02
GO:0006164~purine nucleotide biosynthetic process
3.45
8.70E-02
GO:0030837~negative regulation of actin filament polymerization
1.72
8.88E-02
GO:0002694~regulation of leukocyte activation
3.45
9.08E-02
GO:0044270~nitrogen compound catabolic process
2.59
9.21E-02
GO:0050804~regulation of synaptic transmission
3.45
9.60E-02
GO:0034621~cellular macromolecular complex subunit organization
4.31
9.85E-02
GO:0042592~homeostatic process
7.76
9.90E-02
PT
Term
CE
Table 4. GO biological process of DEPs (DAVID).
GO:0006812~cation transport GO:0050801~ion homeostasis GO:0043242~negative regulation of protein complex disassembly GO:0019725~cellular homeostasis GO:0051493~regulation of cytoskeleton organization
GO:0008366~axon ensheathment
D
GO:0007272~ensheathment of neurons
MA
GO:0019226~transmission of nerve impulse
NU
GO:0006873~cellular ion homeostasis
SC
GO:0043244~regulation of protein complex disassembly
RI
GO:0001508~regulation of action potential
AC
PT E
GO:0006163~purine nucleotide metabolic process
ACCEPTED MANUSCRIPT 33
Table 5. GO cellular component of DEPs (DAVID). %
PValue
GO:0016469~proton-transporting two-sector ATPase complex
3.45
6.94E-03
GO:0005856~cytoskeleton
12.93
7.00E-03
GO:0033176~proton-transporting V-type ATPase complex
2.59
9.03E-03
GO:0045095~keratin filament
3.45
9.29E-03
GO:0044430~cytoskeletal part
10.34
1.11E-02
GO:0031090~organelle membrane
12.07
1.69E-02
GO:0032281~alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
1.72
2.86E-02
4.31
3.09E-02
5.17
3.62E-02
3.45
3.95E-02
3.45
4.24E-02
2.59
4.53E-02
9.48
5.13E-02
5.17
5.40E-02
4.31
5.43E-02
6.03
5.80E-02
6.03
6.10E-02
12.93
6.32E-02
7.76
6.52E-02
5.17
6.68E-02
GO:0005875~microtubule associated complex
2.59
7.82E-02
GO:0016023~cytoplasmic membrane-bounded vesicle
6.90
8.17E-02
GO:0001518~voltage-gated sodium channel complex
1.72
8.35E-02
GO:0031982~vesicle
7.76
8.52E-02
6.90
9.74E-02
PT
Term
selective glutamate receptor complex GO:0005874~microtubule
RI
GO:0043025~cell soma GO:0005882~intermediate filament
SC
GO:0045111~intermediate filament cytoskeleton GO:0070469~respiratory chain GO:0005743~mitochondrial inner membrane GO:0005773~vacuole GO:0005740~mitochondrial envelope GO:0005739~mitochondrion GO:0031410~cytoplasmic vesicle
PT E
D
GO:0019866~organelle inner membrane
MA
GO:0031226~intrinsic to plasma membrane
NU
GO:0005783~endoplasmic reticulum
AC
CE
GO:0031988~membrane-bounded vesicle
ACCEPTED MANUSCRIPT 34
Table 6. GO molecular function of DEPs (DAVID). %
PValue
GO:0051117~ATPase binding
2.59
3.72E-03
GO:0016887~ATPase activity
6.03
8.63E-03
GO:0015078~hydrogen ion transmembrane transporter activity
3.45
2.29E-02
GO:0015077~monovalent inorganic cation transmembrane transporter activity
3.45
2.62E-02
GO:0003697~single-stranded DNA binding
2.59
3.84E-02
GO:0042803~protein homodimerization activity
5.17
5.90E-02
GO:0022890~inorganic cation transmembrane transporter activity
3.45
6.64E-02
2.59
7.42E-02
GO:0042625~ATPase activity, coupled to transmembrane movement of ions
2.59
7.61E-02
GO:0005248~voltage-gated sodium channel activity
1.72
8.26E-02
6.90
9.29E-02
PT
Term
RI
GO:0003777~microtubule motor activity
AC
CE
PT E
D
MA
NU
SC
GO:0042802~identical protein binding
ACCEPTED MANUSCRIPT 35
Table 7. Top ten maps given by MetaCore. Maps
Proteins
pValue
Oxidative phosphorylation
NDUFA11, COX VIIb-1, MT-ND5
1.033E-03
Neurophysiological process_Dynein-dynactin
PRNP, Dynein 1
4.847E-03
Ubiquinone metabolism
NDUFA11, MT-ND5
8.940E-03
Heme metabolism
BVRA, FTH1
1.682E-02
ATP metabolism
CD38, ENTPD2-alpha
1.775E-02
ATP/ITP metabolism
ACYP2, ENTPD2-alpha
2.383E-02
wtCFTR and deltaF508-CFTR traffic / Clathrin
AP180
3.835E-02
coated vesicles formation (normal and CF) SERPINA3 (ACT)
JAK-STAT pathway GluR1
SC
Translation_Role of Retinoic acid signaling in the initiation of translation Neurophysiological process_GABA-A receptor
Dynein 1
AC
CE
PT E
D
MA
NU
life cycle
RI
Development_Thrombopoetin signaling via
PT
motor complex in axonal transport in neurons
4.211E-02 4.211E-02 5.144E-02
ACCEPTED MANUSCRIPT 36
Table 8. Pathway analysis by PANTHER (http://www.pantherdb.org) Pathway
Proteins
Huntington disease (P00029)
F1MAM6,
% D3ZPI7,
21.40
D4AB12, 3ZPI7, F1M2K7, M0R451,
21.40
D4AB12,
M0R451, P38650,M0RCM1
Glycolysis (P00024)
M0RCM1, F1M9V3 P19490, P19493
7.10
Ionotropic glutamate receptor pathway (P00037)
P19490, P19493
7.10
Gonadotropin-releasing hormone receptor pathway (P06664)
P11167
3.60
JAK/STAT signaling pathway (P00038)
Q1KQ07
Blood coagulation (P00011)
P01048
Interleukin signaling pathway (P00036)
Q1KQ07
Apoptosis signaling pathway (P00006)
D3ZS15
3.60
Integrin signalling pathway (P00034)
D4AC70
3.60
RI
SC
Inflammation mediated by chemokine and cytokine signaling pathway (P00031)
NU
Vasopressin synthesis (P04395) ATP synthesis (P02721) CCKR signaling map (P06959)
MA
PDGF signaling pathway (P00047)
CE
PT E
D
EGF receptor signaling pathway (P00018)
AC
PT
Metabotropic glutamate receptor group III pathway (P00039)
3.60 3.60 3.60
Q1KQ07
3.60
Q568Z4
3.60
D3ZS15
3.60
Q64244
3.60
Q1KQ07
3.60
Q1KQ07
3.60
ACCEPTED MANUSCRIPT
NU
SC
RI
PT
37
AC
CE
PT E
D
MA
Fig. 1
ACCEPTED MANUSCRIPT
PT
38
AC
CE
PT E
D
MA
NU
SC
RI
Fig. 2
ACCEPTED MANUSCRIPT
PT
39
AC
CE
PT E
D
MA
NU
SC
RI
Fig. 3
ACCEPTED MANUSCRIPT
PT
40
AC
CE
PT E
D
MA
NU
SC
RI
Fig. 4
ACCEPTED MANUSCRIPT
NU
SC
RI
PT
41
AC
CE
PT E
D
MA
Fig. 5
ACCEPTED MANUSCRIPT
SC
RI
PT
42
AC
CE
PT E
D
MA
NU
Graphical abstract
ACCEPTED MANUSCRIPT 43
Significance statement: The goal of sending astronauts farther into space and extending the duration of spaceflight missions from months to years will challenge the current capabilities of bioastronautics. The investigation of the physiological and pathological changes induced by spaceflight will be critical in developing countermeasures to ensure
PT
astronauts complete spaceflight mission accurately and effectively and return to earth safely. It has been demonstrated the spaceflight may lead to impairments in cognitive
RI
function which is crucial for mission success. Here we show that long-term simulated microgravity, the most potent environment risk factor during spaceflight, impairs the
SC
spatial learning and memory of rats and the underlying mechanism may be involved in glutamate excitotoxicity and imbalances in specific neurotransmitters release in
NU
hippocampus, providing new insight for the countermeasures of cognitive impairment
AC
CE
PT E
D
MA
during spaceflight.
ACCEPTED MANUSCRIPT 44
Highlights Long-term simulated microgravity(SM) impairs spatial learning and memory function of rats. 149 proteins were differentially expressed between SM group and the controls. SM up-regulated GluR1 and GluR4 expression and Glu release.
AC
CE
PT E
D
MA
NU
SC
RI
PT
The concentration of 5-HT, DA, GABA and E were down-regulated by SM.