Identification of the responsible proteins for increased selenium bioavailability in the brain of transgenic rats overexpressing selenoprotein M

  • Authors:
    • Yona Kim
    • Jun Seo Goo
    • Il Yong Kim
    • Ji Eun Kim
    • Moon Hwa Kwak
    • Jun Go
    • Sunbo Shim
    • Jin Tae Hong
    • Dae Youn Hwang
    • Je Kyung Seong
  • View Affiliations

  • Published online on: September 24, 2014     https://doi.org/10.3892/ijmm.2014.1945
  • Pages: 1688-1698
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study was conducted to investigate whether the high antioxidant activity induced by selenium (Sel) treatment and selenoprotein M (SelM) overexpression affected the protein profile of the brain cortex. To accomplish this, the changes in global protein expression were measured in transgenic (Tg) rats expressing human SelM (CMV/hSelM) and non‑Tg rats using two‑dimensional electrophoresis (2‑DE). The results revealed that: ⅰ) CMV/hSelM Tg rats showed a high level of enzyme activity for antioxidant protein in the brain cortex compared to non‑Tg rats; ⅱ) the high activity of these enzymes induced a decrease in total antioxidant concentration and γ‑secretase activity in CMV/hSelM Tg rats; ⅲ) five proteins were upregulated and three were downregulated by SelM overexpression; ⅳ) among the five upregulated proteins, two associated with creatine kinase B‑type (B‑CK) and E3 ubiquitin‑protein ligase RING1 (RING finger protein 1) were further increased in the two groups following Sel treatment, whereas synaptotagmin-15 (SytXV), eukaryotic translation initiation factor 4H (eIF-4H) and lactate dehydrogenase B (LDH-B) were increased or decreased under the same conditions; ⅴ) the three downregulated proteins did not induce a significant change in expression following Sel treatment; and ⅵ) the protein expression level alterations of the two selected spots (B‑CK and SytXV) identified by 2‑DE were extremely similar to the results from western blot analysis. Overall, the results of the present study provide primary novel biological evidence that new functional protein groups and individual proteins in the brain cortex of CMV/hSelM Tg rats are associated with Sel biology, including the response to Sel treatment and SelM overexpression.

Introduction

Selenium (Sel) is an ubiquitous trace element in nature that has been shown to be essential to various aspects of human health (1). This trace element has also been shown to be required for normal growth and reproduction during spermatogenesis (2). Furthermore, Sel deficiency induces multiple diseases associated with oxidative damage, such as fatal cardiomyopathy, which is endemic in Keshan (China) (3), and muscular dystrophy in patients subjected to long-term unsupplemented parenteral nutrition (4). It is also well known that vitamin E can partially replace Sel deficiency (5,6). Sel exists naturally in organic (such as selenomethionine and selenocysteine) and inorganic forms (such as selenite, selenate and selenide) (7). However, these compounds require catabolizing into inorganic precursors prior to insertion into proteins, and the rare amino acid selenocysteine (Sec) is essential for the catalytic function of selenoenzymes (8). Sec, which is the 21st proteinogenic amino acid, was not initially recognized in the classical genetic code as it is encoded by the UGA ‘STOP’ codon. For Sec insertion at UGA codons in the translation process, a specific RNA stem-loop structure is required. In eukaryotes, this loop resides in the 3′-untranslated region of the mRNA, known as the Sec-insertion-sequence (9). Sel has also been shown to be an essential part of mammalian enzymes, such as glutathione peroxidase (GPx), thyroid hormone deiodinase and thioredoxin reductase. Thus far, 25 genes encoding selenoproteins in the sequenced human genome have been identified (10). Selenoprotein M (SelM) was first reported as a 0.7-kb cDNA gene that encoded a new selenoprotein identified from the mammalian EST database. This gene has a 145-amino acid open reading frame beginning with an ATG codon in a favorable Kozak context and contains an in-frame TGA as the Sec codon. Furthermore, homologous proteins have been identified in the rat, zebra fish and other vertebrates, and Sec was conserved in these homologs (11). There have also been several functional studies of SelM. For example, the study by Müller et al (12) showed that this protein plays a major role in spicule formation in the demosponge Suberites domuncula. In addition, Hwang et al (13) indicated that SelM plays a suppressive or protective role in the pathology of patients with Alzheimer’s disease (AD). However, there have been no studies for whether SelM overexpression could affect the changes in global gene expression in CMV/hSelM transgenic (Tg) rats following Sel treatment.

Sel is maintained at high levels in the brain, even upon prolonged dietary Sel deficiency (14). Changes in Sel concentration in the brain and blood have been detected in AD, Parkinson’s disease (PD), multiple sclerosis and brain tumors. Several studies have shown that Sel treatment leads to reduced seizures, improved electroencephalogram recordings (15), protection against the depletion of striatal dopamine (16) and a reduction in the progression of neurodegeneration (17). Furthermore, several selenoproteins have been expressed in the brain. Among these proteins, GPx has been localized in glial cells, and its expression level was significantly upregulated in damaged areas in PD (18). High expression of selenoprotein P (SelP) was also observed in the olfactory bulb, hippocampus and frontal cortex (19). Genetically-engineered models, including Tg and knock-out models, can provide a platform to define the in vivo function of genes and to also study the molecular events responding to environmental changes (20). Previous studies using a knock-out model of SelP showed that it had the important function of Sel delivery into the brain tissue (21,22). Other selenoproteins, including selenoprotein W, thioredoxin reductase, 15-kDa selenoprotein and SelP, have also been detected in the brain. However, numerous questions regarding the roles of these proteins in neuronal function remain.

In the present study, the global change of gene expression affected by SelM overexpression and Sel treatment in the brain cortex was investigated. Two-dimensional electrophoresis (2-DE) analysis showed that eight proteins were significantly changed in CMV/hSelM Tg rats. These results indicated that the information isolated from SelM overexpression and Sel treatment may be useful for studying the association between antioxidant conditions and brain disease, which shows a higher level of oxidative stress condition in specific tissues.

Materials and methods

Maintenance and identification of CMV/hSelM Tg rats

The CMV/hSelM Tg rats used in the study, showing high antioxidant status in various tissues, were developed by microinjection of the CMV/hSelM recombinant gene into fertilized rat eggs (13). The animal protocol was reviewed and approved based on the ethical and scientific care procedures of the Korea Food and Drug Administration (KFDA)-Institutional Animal Care and Use Committee. All rats were maintained in an accredited KFDA animal facility in accordance with AAALAC International Animal Care policies (Accredited Unit-KFDA; unit No. 000996). The rats were provided a standard irradiated chow diet (Purina Mills Inc., St. Louis, MO, USA) ad libitum and maintained in a specified pathogen-free state under a strict light cycle (lights on at 06:00 h and off at 18:00 h). All the pedigrees were hemizygous for their transgenes.

Experimental design and Sel treatment

Sodium selenite (NaSeO3) purchased from Sigma-Aldrich Co., (St. Louis, MO, USA) was dissolved in distilled water to a final concentration of 0.2 μmol/μl (23,24). Ten-week-old rats were randomly divided into two subgroups (n=6 per group). The first subgroup of the CMV/hSelM Tg and non-Tg rat groups each received a comparable volume of distilled water daily via intraperitoneal injection (vehicle-treated CMV/hSelM Tg and non-Tg groups), whereas rats in the second subgroup each received 5 μmol/kg body weight/day of sodium selenite via intraperitoneal injection for three weeks (Sel-treated CMV/hSelM Tg and non-Tg groups). At three weeks after Sel-solution injection, the animals were immediately euthanized using CO2 gas, following which the cortex samples from their brains were prepared and stored in Eppendorf tubes at −70°C until assayed.

Analysis of GPx and superoxide dismutase (SOD) activities and total oxidized products concentration

The levels of GPx and SOD in the brain cortex of CMV/hSelM Tg and non-Tg rats were detected by following the colorimetric assay procedure using Bioxytech SOD-525 and Bioxytech GPx-340 kits (OxisResearch™, Portland, OR, USA). The level of total oxidized products in the sera of CMV/hSelM Tg and non-Tg rats was detected using a Total Antioxidant Status kit (Randox Laboratories Ltd., Antrim, UK) as previously described (13).

γ-secretase activity analysis

The γ-secretase activity was detected with a γ-Secretase Activity kit (R&D System Inc., Minneapolis, MN, USA) using the manufacturer’s instructions. Initially, cortex tissue was homogenized with a glass homogenizer in cold 1X extraction buffer to yield a final protein concentration of roughly 0.5–2.0 mg. These mixtures were incubated on ice for 10 min, after which they were centrifuged at 10,000 × g for 1 min to remove the unbroken fragments. The final supernatants collected from the centrifuged mixture were subsequently used for detection of secretase activity, and this process was carried out in the microplate provided by the manufacturer. To perform the enzyme reaction, 50 μl tissue lysate was added to each well, followed by 50 μl 2X reaction buffer and 5 μl substrate. The microplates were gently mixed and incubated in the dark at 37°C for 1–2 h to induce an enzyme-substrate reaction. Final fluorescence produced by the reaction was read on a fluorescent microplate reader using a filter that allowed EDANS excitation at between 335–355 nm.

Sample preparation for 2-DE

Analyses of global protein expression by 2-DE were performed according to the methods established by our laboratory in previous studies (2527). The cortex samples isolated from the brain were homogenized in liquid nitrogen, following which the homogenized tissues were lysed in buffer [7 M urea, 2 M thiourea, 4% w/v CHAPS, 40 mM Tris and 100 mM dithioerythritol (DTE)]. The sample mixtures were subsequently centrifuged at 45,000 × g at 4°C for 1 h, after which protein concentrations were determined by the Bradford protein assay (Bio-Rad, Hercules, CA, USA). In this process, a cortex sample was generated from a pool of the six animals in each group. The pooled samples were analyzed three times.

2-DE analysis

One-dimensional isoelectric focusing (IEF) was performed using 24 cm immobilized pH gradient (IPG) strips (GE Healthcare, Uppsala, Sweden) in a pH range of 3.0–10.0 (non-linear). Protein (1-mg) was loaded in a total volume of 450 μl, following which the samples were diluted with rehydration solution [7 M urea, 2 M thiourea, 4 % w/v CHAPS, 40 mM Tris, 100 mM DTE and 2% IPG buffer (pH 3.0–10.0)]. After rehydration for 13 h, the strips were focused at 30 V for 2 h, 100 V for 2 h, 200 V for 1 h, 500 V for 1 h, 1,000 V for 1 h and finally at 8,000 V for 22 h to obtain ~100,000 VHr (IPGphor; GE Healthcare). Once IEF was completed, the strips were equilibrated in 6 M urea containing 20% glycerol, 2% SDS and 0.01% bromophenol blue with 10 mM tributyl phosphine. Two-dimensional SDS-PAGE was performed using 8–18% linear gradient acrylamide gels on an EttanDalt system (GE Healthcare). Proteins were visualized by staining with Coomassie blue G-250 (Bio-Rad).

To analyze changes in protein expression between the types of rats according to SelM level, an average gel representing non-Tg rats was compared to an average gel representing the CMV/hSelM Tg rats. Only the filtered spots exceeding an intensity threshold of a 1.5 or 2-fold increase or decrease between non-Tg and CMV/hSleM Tg rats were studied further, whereas the threshold regulation factor for the significance level was set at P≤0.05. Furthermore, any spot showing a significant difference in expression between non-Tg and CMV/hSleM Tg rats was analyzed in all the rats to map changes in expression according to Sel-related factors. Finally, the spots showing significant changes in expression were subsequently identified by mass spectrometry.

Identification of protein spots

The stained gels were scanned with a GS800 densitometer (Bio-Rad) and analyzed using Image master™ (Swiss Institute of Bioinfomatics, Geneva, Switzerland). The spots were digested using trypsin, following which supernatant peptide mixtures were loaded onto a Poros R2 column (Applied Biosystems, Foster City, CA, USA) that had been washed with the following solutions: i) 70% acetonitrile in 5% formic acid; ii) 100% acetonitrile; and iii) 5% formic acid. Peptides were eluted using 5 μl of α-cyano-4-hydroxycinnamic acid and analyzed with a matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometer (Voyager DE-PRO; Applied Biosystems). For protein identification, masses of peptides determined by MALDI-TOF were matched with theoretical peptides in the NCBI (http://www.ncbi.nih.gov/) database using the MASCOT (http://www.matrixscience.com) and ProFound programs (http://prowl.rockefeller.edu).

Western blot analysis

Total proteins prepared from a cortex sample of CMV/hSelM Tg and non-Tg rats were separated by electrophoresis on a 4–20% SDS-PAGE gel for 3 h and subsequently transferred to nitrocellulose membranes for 2 h at 40 V. Each membrane was incubated separately with anti-creatine kinase antibody (Abcam, Cambridge, UK), anti-synaptotagmin antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), and anti-actin (Sigma-Aldrich) antibodies overnight at 4°C. The membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G (Zymed Laboratories, Inc., South San Francisco, CA, USA) at a 1:1,000 dilution at room temperature for 2 h. The membrane blots were developed using a Chemiluminescence Reagent Plus kit (ECL; Amersham Pharmacia Biotech, Inc., Piscataway, NJ, USA).

Statistical analysis

Tests for significance between vehicle- and Sel-treated rats were performed using a one-way analysis of variance test of variance (SPSS for Windows, Release 10.10, Standard Version; SPSS, Inc., Chicago, IL, USA). Tests for significance between CMV/hSelM Tg and non-Tg rats were performed using a post-hoc test (SPSS for Windows, Release 10.10, Standard Version) of variance, and significance levels are provided throughout. All the values are reported as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

Enhancement of SOD and GPX activity in the brain cortex of CMV/hSelM Tg rats

To confirm whether or not alteration of antioxidative conditions is induced by Sel treatment and SelM overexpression in the cortex region of rat brains, the activity of SOD and GPX was measured in the brain tissue of CMV/hSelM Tg and non-Tg rats using a detection kit containing a specific substrate. In the vehicle-treated group, SOD/GPX activity in the CMV/hSelM Tg rats showed a higher level of enzyme activity than that of the non-Tg rats, although they showed differing rates of increase (Fig. 1A and C). Following Sel treatment, the activity of these enzymes increased significantly in the CMV/hSelM Tg and non-Tg rats. However, the rate of increase in the CMV/hSelM Tg rats was greater than that of the non-Tg rats (Fig. 1A and C). These results indicate that SelM overexpression and Sel treatment induced an increase of antioxidant protection in the brain cortex from CMV/hSelM Tg rats.

Change in total oxidized products concentration in the brain of CMV/hSelM Tg rats

To determine whether or not enhancement of SOD and GPX activity was accompanied by a decreased level of oxidized products, the concentration of total oxidized products was determined in the serum of CMV/hSelM Tg and non-Tg rats following Sel treatment using an ELISA kit. For the vehicle-treated group, the concentration of total oxidized products at the basal level was significantly lower in the CMV/hSelM Tg rats compared to the non-Tg rats. Following Sel treatment, these levels decreased simultaneously in the CMV/hSelM Tg and non-Tg rats, although the concentration of total oxidized products in the CMV/hSelM Tg rats was consistently maintained at a low level compared to the non-Tg rats. In particular, the decreasing rate of total oxidized products concentration in the Sel-treated group was significantly greater compared to the vehicle-treated group (Fig. 1B). These results indicate that the overactivation of SOD and GPX induced by SelM overexpression and Sel treatment contributes to the increase in the functional activity for oxidized products protection in the brain cortex of CMV/hSelM rats.

Change of γ-secretase activity in the brain cortex of CMV/hSelM Tg rats

Furthermore, γ-secretase plays an important role in the production of Aβ-42 peptide in the pathogenesis of AD (28). To investigate the effects of Sel treatment and SelM overexpression on the physiological changes in neurodegenerative pathology, the activity of γ-secretase, a critical enzyme of Aβ-42 peptides production, was measured in the brains of CMV/hSelM Tg and non-Tg rats using a detection kit containing a specific substrate. For the vehicle-treated group, the activity of γ-secretase in the CMV/hSelM Tg rats was slightly higher compared to non-Tg rats, although this difference was not significant. However, following Sel treatment, these activities were significantly lower in the two groups of rats. Furthermore, the rate of decrease in γ-secretase activity in the CMV/hSelM Tg rats was higher compared to the non-Tg rats (Fig. 1D). These findings indicate that SelM overexpression and Sel treatment may lead to a decreased incidence of neurodegenerative disease in the brain cortex through the regulation of γ-secretase activity.

Proteome analysis of total proteins in brain cortex from of CMV/hSelM Tg rats

To characterize the changes in global protein expression in the brain cortex of CMV/hSelM Tg rats in response to the induced bioavailability of Sel treatment and SelM overexpression, whole proteins extracted from the brain cortex of 10-week old CMV/hSelM Tg and non-Tg rats were analyzed on analytic 2-DE gels. Computer analysis of gel images showed good matching in four analytical replicates, including vehicle-treated non-Tg, Sel-treated non-Tg, vehicle-treated CMV/hSelM Tg and Sel-treated CMV/hSelM Tg rats. The 2-DE protein maps of the samples from the four groups are shown in Fig. 2. Approximately 270 spots were detected in one gel from brain cortex. The spots that showed significantly different expression were selected for further analysis. Eight spots were identified as key proteins differentially expressed in four experimental groups. Furthermore, in the vehicle-treated group, eight spots were classified into two groups, including upregulated spots (five spots) and downregulated spots (three spots), according to their expression level in CMV/hSelM Tg rats. As shown in Table I, the five upregulated proteins included creatine kinase B-type (B-CK), synaptotagmin-15 (SytXV), E3 ubiquitin-protein ligase RING1 (RING1 finger protein 1), lactate dehydrogenase B (LDH-B) and eukaryotic translation initiation factor 4H (eIF-4H), whereas the three downregulated included centromere protein N (CENP-N), proteasome subunit K and dihydropyrimidinase-related protein 2 (DRP-2).

Table I

List of the differentially expressed proteins in the four experimental groups.

Table I

List of the differentially expressed proteins in the four experimental groups.

Spot no.Protein nameGene nameAccession no.Sequence coverage, %Mw, Da/pIMascot scoreVehicleSelenium


Non-TgTgaNon-TgaTga
1Creatine kinase B-type (EC 2.7.3.2) (Creatine kinase B chain) (B-CK)CkbP073355342698/5.3912413.07±0.312.19±0.183.87±0.25
2E3 ubiquitin-protein ligase RING1 (EC 6.3.2.-) (Polycomb complex protein RING1) (RING finger protein 1)Ring1Q6MGB62342634/5.543012.72±0.182.98±0.213.62±0.28
3Synaptotagmin-15 (Synaptotagmin XV) (SytXV)Syt15P599262147562/8.453211.66±0.122.37±0.181.25±0.11
4L-lactate dehydrogenase B chain (EC 1.1.1.27) (LDH-B) (LDH heart subunit) (LDH-H)LdhbP421231436589/5.707612.24±0.153.19±0.251.57±0.19
5Eukaryotic translation initiation factor 4H (eIF-4H) (Williams-Beuren syndrome chromosomal region 1 protein homolog)Eif4hQ5XI725127307/6.672611.78±0.221.10±0.291.09±0.13
6Proteasome subunit alpha type-3 (EC 3.4.25.1) (Proteasome component C8) (Proteasome subunit K) (Macropain subunit C8) (Multicatalytic endopeptidase complex subunit C8)Psma3P184225828401/5.292510.71±0.050.86±0.070.70±0.06
7Centromere protein N (CENP-N)CenpnQ5U2W43139434/8.093710.53±0.041.14±0.250.49±0.05
8 Dihydropyrimidinase-related protein 2 (DRP-2) (Turned on after division, 64 kDa protein) (TOAD-64) (Collapsin response mediator protein 2) (CRMP-2)Dpysl2P479423262239/5.9512610.34±0.031.17±0.230.36±0.03

a Values in the last three columns on the right hand side are expressed as the relative value of the spot volume for the non-Tg vehicle group, which was defined as 1.

Following Sel treatment, the eight spots showed different patterns. The five-upregulated spots were classified into three groups according to their expression pattern. Two neighboring spots in the first group showed a marked increase in response to Sel treatment and were identified as B-CK and RING1 finger protein 1 (Fig. 3). The volume ratio of these two spots was significantly higher in the CMV/hSelM Tg rats compared to non-Tg rats. Following Sel treatment, these volumes were markedly increased in the two groups relative to the vehicle-treated group. Even though the volume of these two spots increased in the Sel-treated group, they remained at a higher level in the CMV/hSelM Tg rats compared to the non-Tg rats (Fig. 3). The next group consisted of two spots that showed moderate changes in SytXV and LDH-B (Table I). The spot identified as SytXV was expressed at higher levels in the CMV/hSelM Tg rats than non-Tg rats that had received vehicle treatment. Sel treatment induced an increase in spot volume in non-Tg rats, whereas it was significantly decreased in CMV/hSelM Tg rats (Fig. 4). The spot isolated as LDH-B showed a change similar to the spot of SytXV. However, the basic level of these spots was higher in the CMV/hSelM Tg rats compared to the non-Tg rats (Fig. 4). The third group was not affected by Sel treatment and was only induced by SelM overexpression. Spot eIF-4H showed a significantly higher level in CMV/hSelM Tg rats, but was significantly downregulated following Sel treatment, whereas it remained unchanged in the non-Tg rats (Fig. 5).

The group that showed downregulated SelM was further classified into two subgroups. One large spot known as proteasome subunit K belonged to the first subgroup. The volume of this spot was lower in CMV/hSelM Tg rats than non-Tg rats under vehicle-treated conditions. However, Sel treatment induced a decrease in the proteasome subunit K level in non-Tg rats, whereas it remained at a constant level in the CMV/hSelM Tg rats (Fig. 6). In the second subgroup, two spots identified as CENP-N and DRP-2 showed similar patterns under the two conditions (Table I), with a lower level being observed for CMV/hSelM Tg rats than non-Tg rats. These levels were not changed following Sel treatment in either group (Fig. 7).

Confirmation of B-CK and SytXV expression

Western blot analysis was conducted to validate the changes in the protein expression levels of the two selected spots (B-CK and SytXV) identified by 2-DE. As shown in Fig. 8, the expression of B-CK was significantly higher in the CMV/hSelM Tg rats than non-Tg rats. Following Sel treatment, its level increased further in the two groups, although its total expression pattern was maintained. In the case of SytXV, its expression level was higher in CMV/hSelM Tg rats compared to non-Tg rats under vehicle treatment conditions. However, expression of this protein increased significantly in response to Sel treatment in non-Tg rats, whereas CMV/hSelM rats showed no significant difference in expression. The expression pattern of the above proteins observed upon western blot analysis was extremely similar to that in the 2-DE gel image, indicating that the alteration of protein spots detected by 2-DE reflects changes in protein expression in the brains of the CMV/hSelM Tg and non-Tg rats.

Discussion

Sel deficiency is associated with a variety of serious diseases, including infectious disease, cardiovascular disease, cancer and neurodegenerative disorders (7). Savaskan et al (29) provided direct evidence that Sel plays a pivotal role in neuronal susceptibility to excitotoxic lesions. The results of this study indicated that the neuroprotective effects of Sel are not directly mediated via the antioxidative effects of selenite, but require de novo protein synthesis. Therefore, Sel deficiency in the brain tissue leads to increased oxidative stress with subsequent NK-κB activation and neuronal cell death. Several studies reported that intracellular ETKs were activated by either sodium selenite, an inorganic salt known to activate ERK (30), or transfected with a constitutively active mutant of MEK1, an immediate upstream activator of ERKR (31,32). The present study investigated whether the high level of antioxidants induced by Sel treatment and SelM overexpression affected the change in the cortex proteins of CMV/hSelM Tg and non-Tg rats. The CMV/hSelM Tg rats used in the study showed a higher level of antioxidant enzymes, such as SOD and GPX, compared to non-Tg rats. Additionally, the concentration of total oxidized products regulated by these enzymes was significantly decreased in the brain cortex of the CMV/hSelM Tg rats compared to non-Tg rats. These results indicate that the antioxidant condition was induced by Sel treatment and SelM overexpression in CMV/hSelM Tg rats. Furthermore, these findings are concordant with those of our previous study (13).

Aβ-42 peptide in the cortex of the brain is closely correlated with deposition at neuritic plaques in the pathogenesis of AD (28,33). The production of these peptides is tightly regulated by γ-secretase, which is composed of presenilin and four additional cofactors, nicastrin, Aph-1, Pen-2 and TMP-21, to create a multimeric protease complex (34,35). Thus far, limited studies investigating the correlation of Sel and γ-secretase have been conducted. The present study investigated whether the level of γ-secretase activity could be changed by Sel treatment and SelM overexpression in the brain of CMV/hSelM Tg rats. The results showed that the γ-secretase activity significantly decreased in response to Sel treatment. Therefore, the protein spot detected in the study may be correlated with the production of Aβ-42 peptides in CMV/hSelM Tg rats.

The study identified eight spots that were differentially expressed among four groups, which were vehicle-treated non-Tg, vehicle-treated CMV/hSelM Tg, Sel-treated non-Tg and Sel-treated CMV/hSelM Tg rats. Among these spots, creatine kinase was significantly upregulated in response to Sel treatment and SelM overexpression. B-CK, one of the creatine kinases, catalyzed the conversion of creatine to phosphocreatine, which was expressed in various tissue types (36). Generally, this enzyme is known as a buffering system of cellular ATP concentration and used as a marker of several diseases, including myocardial infarction, rhabdomyolysis, muscular dystrophy and renal failure (37). Decreased creatine kinase activity was recently reported to be tightly associated with neurodegenerative pathways in neurodegenerative diseases (38), ischemia (39) and other diseases (40,41). Creatine kinase and pyruvate kinase are crucial for energy homeostasis and antioxidant defense through inhibition of two enzyme activities (42). In the present study, the expression of B-CK was higher in CMV/hSelM Tg rats overexpressing SelM protein than non-Tg rats under vehicle-treated conditions. In addition, Sel treatment induced an increase of this enzyme expression in the two groups. Furthermore, this change pattern was observed in the spot identified as RING1. This enzyme, which is known as a Parkin, appeared to be part of the cell’s defense against damage caused by environmental insults (43). This enzyme ubiquitinates itself and promotes its own degradation, and the impairment of ubiquitin ligase function may cause familial autosomal recessive PD. Furthermore, oxidative and nitrosative stress were closely associated with the ubiquitin ligase function of PD and the associated α-synucleinopathy (44).

SytXV and LDH-B showed similar protein change patterns on the 2-DE gels. Syt constitutes a family of membrane-trafficking proteins that are characterized by an N-terminal transmembrane region, a variable linker and two C-terminal domains (45,46). This protein is known as a Ca2+ sensor that regulates Ca2+-dependent membrane trafficking, including endocrine exocytosis (47,48), synaptic vesicle exocytosis (49,50), plasma membrane repair (51,52) and acrosomal reaction (53). Several studies have also documented the correlation between neurotransmitter release and Syt function (54,55). However, limited studies of whether selenoprotein and Sel treatment were affected by SytXV expression have been conducted thus far. In the present study, the expression of this protein was significantly increased in response to SelM overexpression compared to non-Tg rats. However, in the CMV/hSelM Tg rats, Sel treatment did not induce a significant change in SytXV expression in the brain cortex. These results indicate that SelM overexpression could be affected by Ca2+-dependent membrane trafficking through the regulation of Syt expression. Another spot was identified as LDH-B, which catalyzes the interconversion of pyruvate and lactate with concomitant interconversion of NADH and NAD+ (56). Nazam Ansari et al (57) showed that serum lactate dehydrogenase and GPX were significantly lower in rats with ischemia. However, in the present study, LDH-B was increased by SelM overexpression in CMV/hSelM Tg rats, but increased by Sel treatment in non-Tg rats, indicating that selenoprotein and Sel treatment are extremely important for normal brain function.

eIF-4H was the last member in the groups showing upregulated expression in response to SelM overexpression. This protein regulates protein synthesis through facilitating the binding of initiator tRNA to ribosomes (58). Several stress-responding genes induced by ultraviolet light, ER stress and reactive oxygen species were tightly regulated by eIF2. Furthermore, an abnormally high concentration of NO in the nervous system increases the eIF2α level of phosphorylated forms, thus suppressing protein synthesis in neurons (59). The eIF2α level of the phosphorylated form was also increased in hippocampal CA1 neurons following ischemia (60). In the present study, SelM overexpression induced increased eIF2α expression, but Sel treatment did not induce any change in either group.

The expression of CENP-N was extremely similar to that of DRP-2 upon 2-DE analysis. CENP-N was first reported as a member of three new human centromere proteins (CENP-M, CENP-N and CENP-T), which comprised a CENP-A nucleosome-associated complex (CENP-A NAC) (61). Additionally, disruption of CENP-A NAC by reduction of these three proteins induced a mitotic defect or significant mitotic delay in cells. However, there have been no studies of the correlation between CENP-N and antioxidant status. Therefore, to the best of our knowledge, the present study is the first to indicate that SelM overexpression led to a significant decrease in the expression of CENP-N.

DRP-2 is turned on following division of the 64-kDa protein or collapsing response mediator protein 2 (CRMP-2). This protein is highly expressed in the adult brain, is an important molecule in neurite outgrowth and axonal guidance, and plays a role in several neurological diseases, including AD, epilepsy and ischemia (62,63). Furthermore, a dramatic decrease of intact CRMP-2 was observed following maitotoxin treatment, N-methyl-D-asparatate treatment and ischemia induction, accompanied by the appearance of distinct breakdown products of CRMP-2 (63). The present study showed that SelM overexpression led to significant induction of the decrease of DRP-2 in CMV/hSelM Tg rats compared to non-Tg rats. These results indicate that SelM could be referred to as a regulator of DRP-2, although further study of the degradation process is necessary.

All the aforementioned results showed that SelM overexpression and Sel treatment play a crucial role in the regulation of brain function via modulation of eight associated proteins. In addition, our proteomic analysis allowed the verification of the change in protein patterns of brain antioxidant regulation. However, intensive study is required to define the role and detailed mechanism of SelM and Sel in the cortex region of the brain.

Acknowledgements

The authors wish to acknowledge the animal technicians, S.M. Choi (BS) and Ms. J.L. Song, for directing the Animal Facility and Care at the Division of Laboratory Animal Resources. The present study was supported by the grants from the KFDA. This study was supported by the Bio and Medical Technology Development Program of the National Research Foundation (NRF) funded by the Ministry of Science, ICT and Future Planning (2012M3A9B6055344), and also supported by Bio-industry Technology Development Program, Ministry of Agriculture, Food and Rural Affairs (311054-03-3-HD110) to Professor Je Kyung Seong.

References

1 

Wilber CG: Toxicology of selenium: a review. Clin Toxicol. 17:171–230. 1980. View Article : Google Scholar

2 

Smith AM and Picciano MF: Evidence for increased selenium requirement for the rat during pregnancy and lactation. J Nutr. 116:1068–1079. 1986.PubMed/NCBI

3 

Yang GQ, Chen JS, Wen ZM, Ge KY, Zhu LZ, Chen XC and Chen XS: The role of selenium in Keshan disease. Adv Nutr Res. 6:203–231. 1984. View Article : Google Scholar : PubMed/NCBI

4 

van Rij AM, Thomson CD, McKenzie JM and Robinson MF: Selenium deficiency in total parenteral nutrition. Am J Clin Nutr. 32:2076–2085. 1979.PubMed/NCBI

5 

Schwarz K and Foltz CM: Selenium as an integral part of factor 3 against dietary necrotic liver degeneration. J Am Chem Soc. 79:3292–3293. 1957. View Article : Google Scholar

6 

Reddy PG, Morill JL, Minocha HC and Stevenson JS: Vitamin E is immunostimulatory in calves. J Dairy Sci. 70:993–999. 1987. View Article : Google Scholar : PubMed/NCBI

7 

Birringer M, Pilawa S and Flohé L: Trends in selenium biochemistry. Nat Prod Rep. 19:693–718. 2002. View Article : Google Scholar

8 

Carlson BA, Novoselov SV, Kumaraswamy E, Lee BJ, Anver MR, Gladyshev VN and Hatfield DL: Specific excision of the selenocysteine tRNA[Ser]Sec (Trsp) gene in mouse liver demonstrates an essential role of selenoproteins in liver function. J Biol Chem. 279:8011–8017. 2004.

9 

Berry MJ, Banu L, Chen YY, Mandel SJ, Kieffer JD, Harney JW and Larsen PR: Recognition of UGA as a selenocysteine codon in type I deiodinase requires sequences in the 3′ untransalated region. Nature. 353:273–276. 1991.PubMed/NCBI

10 

Kryukov GV, Castellano S, Novoselov SV, Lobanov AV, Zehtab O, Guigó R and Gladyshev VN: Characterization of mammalian selenoproteomes. Science. 300:1439–1443. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Korotkov KV, Novoselov SV, Hatfield DL and Gladyshev VN: Mammalian selenoprotein in which selenocysteine (Sec) incorporation is supported by a new form of Sec insertion sequence element. Mol Cell Biol. 22:1402–1411. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Müller WE, Borejko A, Brandt D, et al: Selenium affects biosilica formation in the demosponge Suberites domuncula. Effect on gene expression and spicule formation. FEBS J. 272:3838–3852. 2005.PubMed/NCBI

13 

Hwang DY, Sin JS, Kim MS, et al: Overexpression of human selenoprotein M differentially regulates the concentrations of antioxidants and H2O2, the activity of antioxidant enzymes, and the composition of white blood cells in a transgenic rat. Int J Mol Med. 21:169–179. 2008.PubMed/NCBI

14 

Chen J and Berry MJ: Selenium and selenoproteins in the brain and brain diseases (Review). J Neurochem. 86:1–12. 2003. View Article : Google Scholar

15 

Ramaekers VT, Calomme M, Vanden Berghe D and Makropoulos W: Selenium deficiency triggering intractable seizures. Neuropediatrics. 25:217–223. 1994. View Article : Google Scholar : PubMed/NCBI

16 

Imam SZ, el-Yazal J, Newport GD, Itzhak Y, Cadet JL, Slikker W Jr and Ali SF: Methamphetamine-induced dopaminergic neurotoxicity: role of peroxynitrite and neuroprotective role of antioxidants and peroxynitrite decomposition catalysts. Ann NY Acad Sci. 939:366–380. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Zafar KS, Siddiqui A, Sayeed I, Ahmad M, Salim S and Islam F: Dose-dependent protective effect of selenium in rat model of Parkinson’s disease: neurobehavioral and neurochemical evidences. J Neurochem. 84:438–446. 2003.PubMed/NCBI

18 

Takizawa S, Matsushima K, Shinohara Y, Ogawa S, Komatsu N, Utsunomiya H and Watanabe K: Immunohistochemical localization of glutathione peroxidase in infarcted human brain. J Neurol Sci. 122:66–73. 1994. View Article : Google Scholar : PubMed/NCBI

19 

Saijoh K, Saito N, Lee MJ, Fujii M, Kobayashi T and Sumino K: Molecular cloning of cDNA encoding a bovine selenoprotein P-like protein containing 12 selenocysteines and a (His-Pro) rich domain insertion, and its regional expression. Brain Res Mol Brain Res. 30:301–311. 1995. View Article : Google Scholar : PubMed/NCBI

20 

Kim IY, Shin JH and Seong JK: Mouse phenogenomics, toolbox for functional annotation of human genome. BMB Rep. 43:79–90. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Schomburg L, Schweizer U, Holtmann B, Flohé L, Sendtner M and Köhrle J: Gene disruption discloses role of selenoprotein P in selenium delivery to target tissues. Biochem J. 370:397–402. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Hill KE, Zhou J, McMahan WJ, Motley AK, Atkins JF, Gesteland RF and Burk RF: Deletion of selenoprotein P alters distribution of selenium in the mouse. J Biol Chem. 278:13640–13646. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Hwang DY, Seo SJ, Kim YK, et al: Selenium acts as an insulin-like molecule for the downregulation of diabetic symptoms via endoplasmic reticulum stress and insulin signalling proteins in diabetes-induced non-obese diabetic mice. J Biosci. 32:723–735. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Yim SY, Chae KR, Shim SB, et al: ERK activation induced by selenium treatment significantly downregulates beta/gamma-secretase activity and Tau phosphorylation in the transgenic rat overexpressing human selenoprotein M. Int J Mol Med. 24:91–96. 2009.

25 

Goo JS, Kim YN, Choi KM, et al: Proteomic analysis of kidneys from selenoprotein M transgenic rats in response to increased bioability of selenium. Clin Proteomics. 10:102013. View Article : Google Scholar : PubMed/NCBI

26 

Park JY, Seong JK and Paik YK: Proteomic analysis of diet-induced hypercholesterolemic mice. Proteomics. 4:514–523. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Kim BH, Park EY, Yoo KH, Choi KM, Kim Y, Seong Jk and Park JH: N-myc downstream-regulated gene 1 is involved in the regulation of cystogenesis in transgenic mice overexpressing human PKD2 gene. Proteomics. 13:134–141. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Selkoe DJ: Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev. 81:741–766. 2001.

29 

Savaskan NE, Bräuer AU, Kühbacher M, et al: Selenium deficiency increases susceptibility to glutamate-induced excitotoxicity. FASEB J. 17:112–114. 2003.

30 

Hu H, Jiang C, Li G and Lü J: PKB/AKT and ERK regulation of caspase-mediated apoptosis by methylseleninic acid in LNCaP prostate cancer cells. Carcinogenesis. 26:1374–1381. 2005. View Article : Google Scholar

31 

Jaaro H, Rubinfeld H, Hanoch T and Seger R: Nuclear translocation of mitogen-activated protein kinase kinase (MEK1) in response to mitogenic stimulation. Proc Natl Acad Sci USA. 94:3742–3747. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Seger R, Seger D, Reszka AA, et al: Overexpression of mitogen-activated protein kinase kinase (MAPKK) and its mutants in NIH 3T3 cells. Evidence that MAPKK involvement in cellular proliferation is regulated by phosphorylation of serine residues in its kinase subdomains VII and VIII. J Biol Chem. 269:25699–25709. 1994.

33 

Kim SK, Park HJ, Hong HS, Baik EJ, Jung MW and Mook-Jung I: ERK1/2 is an endogenous negative regulator of the gamma-secretase activity. FASEB J. 20:157–159. 2006.PubMed/NCBI

34 

Iwatsubo T: The gamma-secretase complex: machinery for intramembrane proteolysis. Curr Opin Neurobiol. 14:379–383. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Chen F, Hasegawa H, Schmitt-Ulms G, et al: TMP21 is a presenilin complex component that modulates gamma-secretase but not epsilon-secretase activity. Nature. 440:1208–1212. 2006. View Article : Google Scholar

36 

Bessman SP and Carpenter CL: The creatine-creatine phosphate energy shuttle. Annu Rev Biochem. 54:831–862. 1985. View Article : Google Scholar : PubMed/NCBI

37 

Schnyder T, Winkler H, Gross H, Eppenberger HM and Wallimann T: Crystallization of mitochondrial creatine kinase. Growing of large protein crystals and electron microscopic investigation of microcrystals consisting of octamers. J Biol Chem. 266:5318–5322. 1991.

38 

David S, Shoemaker M and Haley BE: Abnormal properties of creatine kinase in Alzheimer’s disease brain: correlation of reduced enzyme activity and active site photolabeling with aberrant cytosol-membrane partitioning. Brain Res Mol Brain Res. 54:276–287. 1998.PubMed/NCBI

39 

Tomimoto H, Yamamoto K, Homburger HA and Yanagihara T: Immunoelectron microscopic investigation of creatine kinase BB-isoenzyme after cerebral ischemia in gerbils. Acta Neuropathol. 86:447–455. 1993.PubMed/NCBI

40 

Gross WL, Bak MI, Ingwall JS, Arstall MA, Smith TW, Balligand JL and Kelly RA: Nitric oxide inhibits creatine kinase and regulates rat heart contractile reserve. Proc Natl Acad Sci USA. 93:5604–5609. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Hamman BL, Bittl JA, Jacobus WE, Allen PD, Spencer RS, Tian R and Ingwall JS: Inhibition of the creatine kinase reaction decreases the contractile reserve of isolated rat hearts. Am J Physiol. 269:H1030–H1036. 1995.PubMed/NCBI

42 

Rech VC, Feksa LR, Fleck RM, Athaydes GA, Dornelles PK, Rodrigues-Junior V and Wannmacher CM: Cysteamine prevents inhibition of thiol-containing enzymes caused by cystine or cystine dimethylester loading in rat brain cortex. Metab Brain Dis. 23:133–145. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Steece-Collier K, Maries E and Kordower JH: Etiology of Parkinson’s disease: genetics and environment revisited. Proc Natl Acad Sci USA. 99:13972–13974. 2002.

44 

Dawson TM: Parkin and defective ubiquitination in Parkinson’s disease. J Neural Transm Suppl. 70:209–213. 2006.

45 

Fukuda M and Mikoshiba K: Synaptotagmin-like protein 1–3: a novel family of C-terminal-type tandem C2 proteins. Biochem Biophys Res Commun. 281:1226–1233. 2001.

46 

Fukuda M, Saegusa C and Mikoshiba K: Novel splicing isoforms of synaptotagmin-like proteins 2 and 3: identification of the Slp homology domain. Biochem Biophys Res Commun. 283:513–519. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Sugita S, Shin OH, Han W, Lao Y and Südhof TC: Synaptotagmins form a hierarchy of exocytotic Ca(2+) sensors with distinct Ca(2+) affinities. EMBO J. 21:270–280. 2002. View Article : Google Scholar : PubMed/NCBI

48 

Fukuda M, Kanno E, Ogata Y, Saegusa C, Kim T, Loh YP and Yamamoto A: Nerve growth factor-dependent sorting of synaptotagmin IV protein to mature dense-core vesicles that undergo calcium-dependent exocytosis in PC12 cells. J Biol Chem. 278:3220–3226. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Chapman ER: Synaptotagmin: a Ca(2+) sensor that triggers exocytosis? Nat Rev Mol Cell Biol. 3:498–508. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Mikoshiba K, Fukuda M, Moreira JE, Lewis FMT, Sugimori M, Niinobe M and Llinás R: Role of the C2A domain of synaptotagmin in transmitter release as determined by specific antibody injection into the squid giant synapse preterminal. Proc Natl Acad Sci USA. 92:10703–10707. 1995. View Article : Google Scholar

51 

Detrait ER, Yoo S, Eddleman CS, Fukuda M, Bittner GD and Fishman HM: Plasmalemmal repair of severed neurites of PC12 cells requires Ca(2+) and synaptotagmin. J Neurosci Res. 62:566–573. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Reddy A, Caler EV and Andrews NW: Plasma membrane repair is mediated by Ca(2+)-regulated exocytosis of lysosomes. Cell. 106:157–169. 2001. View Article : Google Scholar : PubMed/NCBI

53 

Michaut M, De Blas G, Tomes CN, Yunes R, Fukuda M and Mayorga LS: Synaptotagmin VI participates in the acrosome reaction of human spermatozoa. Dev Biol. 235:521–529. 2001. View Article : Google Scholar : PubMed/NCBI

54 

Schiavo G, Osborne SL and Sgouros JG: Synaptotagmins: more isoforms than functions? Biochem Biophys Res Commun. 248:1–8. 1998. View Article : Google Scholar : PubMed/NCBI

55 

Fukuda M and Mikoshiba K: The function of inositol high polyphosphate binding proteins. Bioessays. 19:593–603. 1997. View Article : Google Scholar : PubMed/NCBI

56 

Philibert RA, Nelson JJ, Sandhu HK, Crowe RR and Coryell WH: Association of an exonic LDHA polymorphism with altered respiratory response in probands at high risk for panic disorder. Am J Med Genet B Neuropsychiatr Genet. 117:11–17. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Nazam Ansari M, Bhandari U, Islam F and Tripathi CD: Evaluation of antioxidant and neuroprotective effect of ethanolic extract of Embelia ribes Burm in focal cerebral ischemia/reperfusion-induced oxidative stress in rats. Fundam Clin Pharmacol. 22:305–314. 2008.PubMed/NCBI

58 

Kim HS, Choi Y, Shin KY, et al: Swedish amyloid precursor protein mutation increases phosphorylation of eIF2alpha in vitro and in vivo. J Neurosci Res. 85:1528–1537. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Petrov T, Underwood BD, Braun B, Alousi SS and Rafols JA: Upregulation of iNOS expression and phosphorylation of eIF-2alpha are paralleled by suppression of protein synthesis in rat hypothalamus in a closed head trauma model. J Neurotrauma. 18:799–812. 2001. View Article : Google Scholar : PubMed/NCBI

60 

Hayashi T, Saito A, Okuno S, et al: Oxidative damage to the endoplasmic reticulum is implicated in ischemic neuronal cell death. J Cereb Blood Flow Metab. 23:1117–1128. 2003. View Article : Google Scholar : PubMed/NCBI

61 

Foltz DR, Jansen LE, Black BE, Bailey AO, Yates JR 3rd and Cleveland DW: The human CENP-A centromeric nucleosome-associated complex. Nat Cell Biol. 8:458–469. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Zhang Z, Ottens AK, Sadasivan S, Kobeissy FH, Fang T, Hayes RL and Wang KK: Calpain-mediated collapsin response mediator protein-1, -2, and -4 proteolysis after neurotoxic and traumatic brain injury. J Neurotrauma. 24:460–472. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Chung MA, Lee JE, Lee JY, Ko MJ, Lee ST and Kim HJ: Alteration of collapsin response mediator protein-2 expression in focal ischemic rat brain. Neuroreport. 16:1647–1653. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 34 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kim Y, Goo JS, Kim IY, Kim JE, Kwak MH, Go J, Shim S, Hong JT, Hwang DY, Seong JK, Seong JK, et al: Identification of the responsible proteins for increased selenium bioavailability in the brain of transgenic rats overexpressing selenoprotein M. Int J Mol Med 34: 1688-1698, 2014
APA
Kim, Y., Goo, J.S., Kim, I.Y., Kim, J.E., Kwak, M.H., Go, J. ... Seong, J.K. (2014). Identification of the responsible proteins for increased selenium bioavailability in the brain of transgenic rats overexpressing selenoprotein M. International Journal of Molecular Medicine, 34, 1688-1698. https://doi.org/10.3892/ijmm.2014.1945
MLA
Kim, Y., Goo, J. S., Kim, I. Y., Kim, J. E., Kwak, M. H., Go, J., Shim, S., Hong, J. T., Hwang, D. Y., Seong, J. K."Identification of the responsible proteins for increased selenium bioavailability in the brain of transgenic rats overexpressing selenoprotein M". International Journal of Molecular Medicine 34.6 (2014): 1688-1698.
Chicago
Kim, Y., Goo, J. S., Kim, I. Y., Kim, J. E., Kwak, M. H., Go, J., Shim, S., Hong, J. T., Hwang, D. Y., Seong, J. K."Identification of the responsible proteins for increased selenium bioavailability in the brain of transgenic rats overexpressing selenoprotein M". International Journal of Molecular Medicine 34, no. 6 (2014): 1688-1698. https://doi.org/10.3892/ijmm.2014.1945