JOURNAL OF NEUROCHEMISTRY

| 2014 | 130 | 797–804

doi: 10.1111/jnc.12783

*Department of Pediatrics, Medical University of Vienna, Vienna, Austria †Department of Medical Chemistry, Medical University of Vienna, Vienna, Austria ‡Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria §Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna, Himberg, Austria

Abstract Proteoglycans (PGs) are major constituents of the extracellular matrix and have recently been proposed to contribute to synaptic plasticity. Hippocampal PGs have not yet been studied or linked to memory. The aim of the study, therefore, was to isolate and characterize rat hippocampal PGs and determine their possible role in spatial memory. PGs were extracted from rat hippocampi by anion-exchange chromatography and analyzed by nano LC-MS/MS. Twenty male Sprague–Dawley rats were tested in the morris water maze. PGs agrin, amyloid beta A4 protein, brevican, glypican-1, neurocan, phosphacan, syndecan-4, and versican were identified in the hippocampi. Brevican and versican levels in the membrane fraction were higher in the trained group, correlat-

ing with the time spent in the target quadrant. a-amino-3hydroxy-5-methylisoxazole-4-propionate receptor GluR1 was co-precipitated with brevican and versican. Levels for a receptor complex containing GluR1 was higher in trained while GluR2 and GluR3-containing complex levels were higher in yoked rats. The findings provide information about the PGs present in the rat hippocampus, demonstrating that versican and brevican are linked to memory retrieval in the morris water maze and that PGs interact with a-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor GluR1, which is linked to memory retrieval. Keywords: AMPA receptor complex, brevican, morris water maze, proteoglycans, spatial memory, versican. J. Neurochem. (2014) 130, 797–804.

The brain’s structural plasticity is largely modified during its development and maturation, and also at the time of remodeling processes in adulthood. In adult hippocampus, synaptic plasticity in association with memory formation is considered a prerequisite for the normal function of this brain area. This process involves cellular signaling from extracellular environment to the cell nucleus. Brain synapses are in a wrapped form, a dense meshwork of extracellular matrix comprising lectican-bound hyaluronic acid and tenascin, known as perineuronal net (PNN) (Celio et al. 1998), maintaining excitatory/inhibitory homeostasis in the adult brain (Hensch 2005). PNNs are diminished in patients with schizophrenia (Pantazopoulos et al. 2010) but preserved in patients with Alzheimer0 s disease (Morawski et al. 2012). Loss of PNNs is associated with aberrant hippocampal

activity and abnormal dopaminergic function (Shah and Lodge 2013). More than 15 proteoglycans are reported in the central nervous system (Bandtlow and Zimmermann 2000), however, no systematic studies have been carried out to identify proteoglycans from hippocampus. Although the pivotal role of proteoglycans in synaptic plasticity has been proposed Received February 14, 2014; revised manuscript received May 22, 2014; accepted June 1, 2014. Address correspondence and reprint requests to Prof Gert Lubec, Department of Pediatrics, Medical University of Vienna, W€ahringer G€urtel 18, 1090 Vienna, Austria. E-mail: [email protected] Abbreviations used: AMPA, a-amino-3-hydroxy-5-methylisoxazole4-propionate; ECL, enhanced chemiluminescence; LTP, long-term potentiation; PGs, proteoglycans; PNN, perineuronal net.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

797

798

S. R. Saroja et al.

(Daniels 2012; Orlando et al. 2012; Senkov et al. 2012), their role in cognitive functions including memory formation and storage remain elusive. Knockout studies of the proteoglycan brevican revealed that it was not involved in memory formation (Brakebusch et al. 2002). Nevertheless, PNNs were shown to prevent fear memory from extinction (Gogolla et al. 2009) and affected lateral diffusion of a-amino-3-hydroxy-5methylisoxazole-4-propionate (AMPA) receptors (Frischknecht et al. 2009), key elements of signaling in memory processes (Krugers and Hoogenraad 2009; Conboy and Sandi 2010; Rust et al. 2010; Yuen et al. 2011). These few studies suggest that PNN-associated proteoglycans may play a role in learning and memory. AMPARs execute their function through assembly to complexes rather than subunits and two major populations of AMPA receptor complexes are reported from rat hippocampus: Glutamate receptor subunits (GluR)1GluR2 and GluR2-GluR3 and a limited population of GluR1GluR3 complexes (Wenthold et al. 1996). The GluR1-GluR2 complex is trafficked to the membrane upon synaptic activity, while GluR2-GluR3 complexes undergo constitutive cycling (Emond et al. 2010). Therefore, it is challenging to study the link between expression of memory-linked proteoglycans and membrane AMPA receptor complexes for better understanding of the molecular mechanisms involved in memory mechanisms. In this study, proteoglycans were isolated by anionexchange chromatography from rat hippocampus and identified by mass spectrometry. Selected proteoglycans were quantified from hippocampi of rats that were tested in the spatial paradigm morris water maze (MWM). Modification of AMPAR complexes were analyzed and shown to interact with proteoglycans brevican and versican.

Materials and methods Animals Rats were bred and maintained in Makrolon cages, supplied with autoclaved woodchips, standard rodent diet (Altromin, Lage, Germany), and water in the Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna. Room temperature was 22  1°C and relative humidity was 50  10%. The light/dark rhythm was 14 : 10 h. Ventilation with 100% fresh air resulted in an air change rate of 15 times per hour. The room was illuminated with artificial light at an intensity of about 200 lx in 2 m from 5 AM to 7 PM Behavioral tests were performed between 8 AM and 1 PM. All procedures were carried out according to the guidelines of the European Communities Council Directive of 24 November 1986 (86/609/EEC) and were approved by Federal Ministry of Education, Science and Culture, Austria. Morris water maze Ten male Sprague–Dawley rats, 12–14 weeks old were trained in four daily acquisition sessions (four trials per day with 20 min intertrial interval) to find a hidden platform in a circular pool (150 cm diameter, wall depth 60 cm) filled with water (21  1°C). Rats that failed to locate the platform within 120 s were manually placed on

the platform and allowed to remain on it for 30 s. Memory retrieval was assessed on day 5 (60 s, platform removed) (Sase et al. 2013). Ten rats spending the same time in the MWM without training were used as controls. All experiments were recorded by computerized tracking/image analyzer system (video camcorder: 1/3’ SSAM HR EX VIEW HAD coupled to the computational tracking system: TiBeSplit). The rats were bred and maintained at the Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna. Isolation and identification of proteoglycans Rat hippocampi were homogenized in a buffer containing 50 mM Tris (pH 8.0), 0.15 M NaCl, 1% {3-[(3-Cholamidopropyl)dimethylammonio]-1-propanesulfonate}, and one complete protease inhibitor tablet [(Roche Molecular Biochemicals, Mannheim, Germany) per 50 mL]. Following incubation at 4°C for 1 h, samples were centrifuged at 20 000 g for 45 min. The pellet was discarded, while the supernatant was retained for further analysis. The efficiency of proteoglycan extraction is shown in Figure S2a. Proteoglycans (PGs) were isolated by using HiTrap DEAE Sepharose FF according to the manufacturer0 s instructions (17-5055-01; GE Healthcare, Freiburg, Germany). Isolated PGs were treated with chondroitinase ABC (C3667-5UN; Sigma, St Louis, MO, USA) (Herndon and Lander 1990) in order to cleave glycosaminoglycans to allow electrophoretic separation. Resulting PG proteins were separated by 8% sodium dodecyl sulfate–polyacrylamide gel electrophoresis and in-gel digested peptides were identified by Nano-LC-ESI-CID/ETDMS/MS (Ghafari et al. 2012a,b; Wan et al. 2012). Hippocampal membrane protein preparation All procedures were carried out at 4°C. Hippocampi were homogenized in ice-cold homogenization buffer [10 mM HEPES, pH 7.5, 300 mM sucrose, one complete protease inhibitor tablet (Roche Molecular Biochemicals) per 50 mL] by Ultra-Turrax (IKA, Staufen, Germany). The homogenate was centrifuged for 10 min at 1000 g and the pellet was discarded. The supernatant was centrifuged at 50 000 g for 30 min in an ultracentrifuge (Optima- L-90K, Beckman Coulter Inc., Brea, CA, USA). The pellet was resuspended in washing buffer (homogenization buffer without sucrose), kept on ice for 30 min and centrifuged at 50 000 g for 30 min to obtain a membrane fraction. Extraction buffer (1.5 M 6-aminocaproic acid, 300 mM Bis–Tris, pH 7.0) with 1% n-Dodecyl b-D-maltoside was added to the membrane pellets, incubated for 1 h with vortexing every 10 min. Following solubilization, samples were cleared by centrifugation at 20 000 g for 1 h. The protein content was estimated using the bicinchoninic acid protein assay kit (Pierce, Rockford, IL, USA). Extracted proteins were then aliquoted and stored at 80°C till use (Sase et al. 2012). Gel electrophoresis and western blotting For the quantification of AMPA receptor complexes and PGs hippocampal membrane fractions were used. AMPA receptor complexes were separated on 5–13% of blue native polyacrylamide gel electrophoresis (BN-PAGE) as described previously (Sase et al. 2012) while PGs were separated on 8% sodium dodecyl sulfate– polyacrylamide gel electrophoresis. Separated proteins were transferred to polyvinylidene difluoride membranes and blocked with 5% of non-fat dry milk in Tris-Buffered Saline and Tween 20 for 1 h. Primary antibodies were added and incubated overnight at 4°C. After

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

Hippocampal proteoglycans linked to spatial memory

membranes were washed for six times with Tris-Buffered Saline and Tween 20, corresponding secondary species-specific horseradish peroxidase-conjugated antibodies against IgG were added and incubated for 1 h at 21°C (Antibody details are given in Table S1). Membranes were developed with the enhanced chemiluminescence (ECL) solution (Clarity Western ECL Substrate, Bio-Rad) and hyperfilms (AmershamTM Hyperfifilm ECL, GETM Healthcare, GE Healthcare Europe GmbH, Vienna, Austria) were used. Eight to ten animals per group were used for quantification analysis. A rectangular area of immunoreactive bands was selected and arbitrary units of optical density were measured using the software Image J (http:// imagej.nih.gov/ij/features.html or http://howtowesternblot.net/dataanalysis-3/quantification/). Proteins from bands at the apparent molecular weights in the individual lanes from 35 kDa to 150 kDa for proteoglycans and from 720 kDa to 1236 kDa for AMPA receptors were used for normalization from coomassie blue R-350 stained membranes (Figure S1) (Welinder and Ekblad 2011; Lee et al. 2013). The intensity of individual immunoreactive band was divided by the corresponding lane intensity of coomassie blue R-350 stained membrane to get the normalized values. Selection of these particular molecular ranges would give low variations between samples and groups. This does not rule out, however, that learning and memory may change total protein levels in the gels although this fact has never been addressed in neurochemistry so far. Immunoprecipitation and antibody-shift assay For immunoprecipitation (IP) experiments, 3 lg of GluR1 (AB31232; Abcam, Cambridge, MA, USA), 5 lg of GluR2 (AB1768; Millipore Corporation, Bedford, MA, USA), 10 lg of GluR3 (3437; Cell Signaling Technology, Beverly, MA, USA), 20 lg of brevican (610895; BD Biosciences, San Jose, CA, USA), and versican (sc-25831; Santa Cruz Biotechnology, Santa Cruz, CA, USA) antibodies were used per 500–1000 lg of hippocampal membrane proteins. Anti-cAMP response element-binding protein antibody (Cell Signaling) was used for control IP (Figure S2b). IP procedures were carried out according to the manufacturer0 s instructions (Thermo Scientific Pierce Direct IP Kit, Rockford, IL, USA). For antibody-shift assays, 0.5 lg of either GluR1 or brevican antibodies were added to 100 lg of membrane proteins, incubated for 1 h at 4°C and then separated by 5–13% of BN-PAGE. GluR1 and brevican antibody-treated samples were probed with brevican and GluR1 antibodies, respectively. Statistical analysis One-Way ANOVA combined with post hoc multiple comparison test (Bonferroni) was used to analyze latency, path length, and speed. Time spent in target quadrant and western blot data were analyzed by an unpaired t-test. Pearson correlation coefficients were used for correlation studies. A probability level of p < 0.05 was considered statistically significant. Calculations were performed using Graphpad Prism 6.

Results Identification of rat hippocampal proteoglycans by nano LCMS/MS From 12 mg of total hippocampal protein ~ 15 lg of PGs were isolated. In order to get sufficient amounts of PGs for

799

Table 1 Mass spectrometric-based identification of proteoglycans and tenascin-R from rat hippocampus Gene ID

Protein Name

Swiss Prot No.

Agrn App Bcan Gpc1 Ncan Ptprz1 Sdc4 Tnr Vcan

Agrin Amyloid beta A4 protein Brevican Glypican 1 Neurocan Phosphacan Syndecan-4 Tenascin-R Versican

P25304 P08592 P55068 P35053 P55067 Q62656 P34901 Q05546 Q9ERB4

mass spectrometric analysis, five hippocampi were pooled for PGs isolation as described above. Agrin, amyloid beta A4 protein, brevican, glypican-1, neurocan, phosphacan, syndecan-4, and versican were identified by mass spectrometry. Tenascin-R, a major component of PNN glycoproteins, was also identified (Table 1). Information on peptides for unambiguous identification of proteoglycans is given in Table S2. Changes of PG levels upon spatial memory retrieval Spatial memory was accessed by the MWM. During the training period of 4 days, latencies to reach the platform (Fig. 1a) and path lengths (Fig. 1b) were significantly reduced as animals learned the task. Speed remained comparable between training days (Fig. 1c). On day 5, the time spent in the target quadrant was significantly increased in the trained animals as compared to remaining quadrants during the probe trial (Fig. 1d). Times spent in the target quadrant were comparable to the other quadrants in the yoked group (Fig. 1e). Membrane fraction, which contains both PNN-associated proteoglycans (Shah and Lodge 2013) and membrane-bound proteoglycans, were subjected to analyze memory-related proteoglycans as PNN had been shown to play a critical role in fear memory extinction (Gogolla et al. 2009) and lateral diffusion of AMPA receptor (Frischknecht et al. 2009). Agrin, brevican, glypican-1, phosphacan, tenascin-R, and versican were selected for western blotting-based quantitative analysis between yoked and trained animals. Western blot patterns are given in Fig. 2. Brevican bands at 145 kDa and 50 kDa and versican levels were significantly increased in hippocampi of trained animals as compared to yoked animals while the > 145 kDa band of brevican was not increased significantly (Table 2). Pearson correlation studies showed significant positive correlation between brevican and versican and the time spent in the target quadrant (Fig. 3a–c). So far PG changes were only demonstrated in the membrane fraction which are not necessarily because of changed synthesis or secretion but may simply be because of changed solubility or anchoring at membranes.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

800

S. R. Saroja et al.

(a)

(b)

(d)

(e)

(c)

Fig. 1 Morris water maze. Rats were trained in morris water maze (MWM). Time taken to reach the platform (latency; a), distance traveled to reach platform (pathlength; b) and the average speed (c) during training days were calculated with One-way ANOVA analysis. (d) Unpaired t-test was performed between time spent in the target

quadrant and remaining quadrants of trained animals during probe trails. (e) Unpaired t-test was performed between time spent in the target quadrant and remaining quadrants of yoked group during probe trails. Values are means  SD. ****p < 0.0001.

Table 2 Quantification of proteoglycans and tenascin-R in yoked and trained rat hippocampus

PGs Agrin 200 kDa Agrin 90 kDa Brevican > 145 kDa Brevican 145 kDa Brevican 50 kDa Glypican 1 Phosphacan Tenascin-R Versican

Yoked Mean  SD, n

Trained Mean  SD, n

p value

0.2978  0.0917, 9

0.3169  0.1048, 8

0.6861

0.1934  0.157, 9 0.1424  0.064, 9

0.2394  0.088, 8 0.1951  0.073, 10

0.476 0.118

0.1255  0.045, 9

0.1928  0.046, 10

0.0071

0.5702  0.111, 9

0.7813  0.2496, 10

0.0251

1.4669 0.0596 0.1233 0.0468

   

0.7612, 0.0213, 0.0315, 0.0087,

8 9 10 10

1.4451 0.0793 0.1174 0.1211

   

0.4902, 8 0.036, 8 0.0179, 10 0.0776, 10

0.9432 0.2058 0.6204 0.0075

Brevican and versican form a complex with GluR1 Although PNNs were shown to affect AMPA receptor (AMPAR) mobility, to the best of our knowledge direct interaction between PNN-associated proteoglycans and AMPARs was not reported yet. IP was performed using brevican and versican antibodies as both PGs were significantly modified in spatial memory retrieval and subsequently probed with a GluR1 antibody. Results from IP showed that

Fig. 2 Western blot imaging of proteoglycans and tenascin-R. Hippocampal membrane proteins were prepared from morris water maze (MWM) trained and yoked groups. Hippocampal membrane proteins of yoked and trained animals were separated on 8% of sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE) and probed with indicated proteoglycan and tenascin-R antibodies.

brevican (145 kDa) and versican were co-immunoprecipitated with GluR1 suggesting that these proteins are in a complex (Fig. 4a). However, 50 kDa band of brevican was not detected with GluR1 co-immunoprecipitation. In addition, antibody-shift assays were performed to confirm that GluR1 and brevican are in the same complex. Membrane protein fractions were pre-incubated with an antibody against either GluR1 or an antibody against brevican before separation by BN-PAGE. Samples pre-incubated with GluR1 antibody were probed using an anti-brevican antibody and

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

Hippocampal proteoglycans linked to spatial memory

(a)

(b)

801

(c)

Fig. 3 Pearson correlations. Learning and memory altered proteoglycans were statistically correlated with the time spent by the rats in the target quadrant of MWM. Pearson’s correlations between the 145 kDa band of brevican and the time spent in the target quadrant (a), the 50 kDa band of brevican and the time spent in the target quadrant (b), and versican levels and time spent in the target quadrant (c).

(a)

(b)

(c)

Fig. 4 Complex analysis. (a) Immunoprecipitation was performed from hippocampal membrane fraction using brevican and versican antibodies and subsequently probed with indicated antibodies. (b) Antibodyshift assay; in the first panel hippocampal membrane proteins were incubated with GluR1 antibody, separated on 5–13% blue native polyacrylamide gel electrophoresis (BN-PAGE) and probed with brevican antibodies. In the second panel membrane, proteins were

incubated with brevican or GluR1 antibody, separated on 5–13% BNPAGE and probed with GluR1 antibody. Membrane proteins without antibody were used as controls (first lanes). (c) Hippocampal membrane proteins of yoked and trained animals were separated on 5–13% of BNPAGE and probed with antibodies against AMPA receptor subunits GluR1-3. Note: In the western blot image of GluR2 and GluR3, blank spaces indicate that no samples were loaded as wells were damaged.

extracts pre-incubated with brevican using an antibody against GluR1, respectively. The clear shifts in antibodyincubated lanes confirmed that GluR1 and brevican were in the same complex (Fig. 4b).

Fig. 4c. Levels of a GluR1-containing receptor complex were significantly increased in the trained group while levels of GluR2-and GluR3- containing receptor complexes were significantly increased in the yoked group (Table 3). Pearson correlation studies showed that levels of the GluR1-containing complex were positively correlating while levels of the GluR3-containing complex were negatively correlated with the time spent in the target quadrant (Fig. 5a and b) GluR1containing receptor complex levels correlated significantly

Modification of AMPAR complex levels in spatial memory formation AMPAR complex level changes between trained and yoked groups were analyzed. Western blot bands are shown in

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

802

S. R. Saroja et al.

Table 3 AMPAR complexes in yoked and trained rat hippocampus AMPAR complexes

Yoked Mean  SD, n

Trained Mean  SD, n

p value

GluR1complex GluR2complex GluR3complex

0.1499  0.0158, 10

0.1832  0.012, 10

< 0.0001

0.0168  0.0059, 10

0.0111  0.0054, 10

0.1153  0.0321, 10

0.0404  0.009, 10

0.0388 < 0.0001

with levels of the 145 kDa form of brevican (Fig. 5c) while complex levels containing GluR3 correlated significantly with versican (Fig. 5d).

Discussion The most salient findings of this study show that a series of PGs are identified in rat hippocampus, and that brevican and versican are linked to spatial memory retrieval. A complex formation of brevican and versican with the AMPAR GluR1 has been observed, along with the fact that AMPARs GluR1and GluR3-containing complexes correlate with the time spent in the target quadrant, the parameter used for evaluation of retrieval in spatial memory. As shown in the results, agrin, amyloid beta A4 protein, brevican, glypican-1, neurocan, phosphacan, syndecan-4, tenascin-R, and versican were observed in rat hippocampus by mass spectrometry. Agrin was suggested to be associated with synapse maturation (Ferreira 1999) and amyloid beta A4 protein was proposed to be associated with Alzheimer disease and Down syndrome (Beyreuther et al. 1993). Hypoxicischemic brain injury altered brevican and versican expression in rat hippocampus (Aya-ay et al. 2005; Leonardo et al. 2008) and loss of synapses was associated with loss of brevican (Morawski et al. 2012). Neurocan knockout mice showed normal early-phase long-term potentiation (LTP) but (a)

late-phase LTP was impaired (Zhou et al. 2001). Proteolytic degradation of phosphacan was linked to neuronal degeneration in the hippocampus (Kurazono et al. 2001) and this PG was shown to contribute to mossy fiber outgrowth and regeneration (Butler et al. 2004). Electrophysiological studies have shown that Tenascin-R is involved in synaptic plasticity (Saghatelyan et al. 2000; Nikonenko et al. 2003). In this study, increased levels of brevican and versican in the trained group may point to involvement of these PGs in spatial memory in the rat. Brakebusch et al. (2002) have shown that brevican-deficient mice present with impaired LTP but learning and memory as evaluated in a MWM were not affected. Their finding of unchanged learning and memory does not contradict our results. As shown in the Results section, brevican and versican coprecipitated with AMPAR GluR1 and an antibody-shift assay clearly indicated that brevican and GluR1 were in the same complex. This complex formation was not shown before, but glypican 4 and 6 led to clustering of GluR1 on the cell surface (Allen et al. 2012) and the PNN, which mainly consists of PGs, was shown to affect AMPAR trafficking (Frischknecht et al. 2009). It is, however, unclear whether those proteoglycans interacted with GluR1 through core protein or glycosaminoglycan side chains. Like brevican and versican, GluR1-containing complexes were correlating with the time spent in the target quadrant and indeed, GluR1containing complexes were linked to spatial memory in the mouse (Ghafari et al. 2012a,b) and the link between GluR1 and memory per se is evident and well-documented. The correlation between GluR1-containing complex levels and brevican again would point to a role of this PG in spatial memory retrieval. Versican was found to negatively correlate with GluR3-containing complex levels, which were decreased in trained rats, and positively correlate with the time spent in the target quadrant, probably suggesting a role also for versican during memory retrieval. A previous study demonstrated that GluR3-containing complexes were decreased in spatial memory training (Falsafi et al. 2012), and here it was shown that GluR3-containing complex levels (c)

(b)

Fig. 5 Pearson correlations. Pearson’s correlations were performed between time spent in the target quadrant and significantly altered AMPA receptor complexes. To link AMPA receptor complexes with proteoglycans, correlations were also performed between significantly altered proteoglycans and AMAP receptor complexes. Correlations

(d)

between the GluR1-containing complex levels and the time spent in the target quadrant (a), GluR3-containing complex levels and the time spent in the target quadrant (b), GluR1-containing complex levels and levels of the 145 kDa band of brevican (c), and GluR3-containing complex levels and versican levels (d).

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

Hippocampal proteoglycans linked to spatial memory

were decreased in memory retrieval. AMPARs exert function by assembling into homo-and hetero-oligomers, and therefore AMPARs complexes rather than subunits were investigated (Emond et al. 2010). The complexity of AMPAR complexes has been addressed by several groups (Wenthold et al. 1996; Reimers et al. 2011; Schwenk et al. 2012) and we, hereby, are adding two proteoglycans, brevican and versican, to the probable interactome of AMPARs. Taken together, we have shown the presence of several PGs in the rat hippocampus, revealed the association of brevican and versican with AMPARs-containing GluR1 or GluR3 and the time spent in the target quadrant representing memory retrieval performance in the MWM. Moreover, it has been demonstrated that brevican and versican are probable complex components of GluR1- or GluR3-containing complexes. Therefore, we propose a tentative role of brevican and versican in spatial memory either directly or by interaction with AMPARs in the MWM.

Acknowledgments and conflict of interest disclosure The study was funded by the Medical University of Vienna, Austria. All experiments were conducted in compliance with the ARRIVE guidelines. There are no conflicts of interest to declare.

Supporting information Additional supporting information may be found in the online version of this article at the publisher's web-site: Figure S1. Coomassie blue R-350 stained membranes were used for normalization. Figure S2. (a) Hippocampal proteins were extracted with {3-[(3Cholamidopropyl)dimethylammonio]-1-propanesulfonate} as described in materials and methods section. Table S1. Details of antibodies used for western blotting.

References Allen N. J., Bennett M. L., Foo L. C., Wang G. X., Chakraborty C., Smith S. J. and Barres B. A. (2012) Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors. Nature 486, 410–414. Aya-ay J., Mayer J., Eakin A. K., Muffly B. G., Anello M., Sandy J. D. and Gottschall P. E. (2005) The effect of hypoxic-ischemic brain injury in perinatal rats on the abundance and proteolysis of brevican and NG2. Exp. Neurol. 193, 149–162. Bandtlow C. E. and Zimmermann D. R. (2000) Proteoglycans in the developing brain: new conceptual insights for old proteins. Physiol. Rev. 80, 1267–1290. Beyreuther K., Pollwein P., Multhaup G., Monning U., Konig G., Dyrks T., Schubert W. and Masters C. L. (1993) Regulation and expression of the Alzheimer’s beta/A4 amyloid protein precursor in health, disease, and Down’s syndrome. Ann. N. Y. Acad. Sci. 695, 91–102. Brakebusch C., Seidenbecher C. I., Asztely F. et al. (2002) Brevicandeficient mice display impaired hippocampal CA1 long-term

803

potentiation but show no obvious deficits in learning and memory. Mol. Cell. Biol. 22, 7417–7427. Butler C. D., Schnetz S. A., Yu E. Y., Davis J. B., Temple K., Silver J. and Malouf A. T. (2004) Keratan sulfate proteoglycan phosphacan regulates mossy fiber outgrowth and regeneration. J. Neurosci. 24, 462–473. Celio M. R., Spreafico R., De Biasi S. and Vitellaro-Zuccarello L. (1998) Perineuronal nets: past and present. Trends Neurosci. 21, 510–515. Conboy L. and Sandi C. (2010) Stress at learning facilitates memory formation by regulating AMPA receptor trafficking through a glucocorticoid action. Neuropsychopharmacology 35, 674–685. Daniels M. P. (2012) The role of agrin in synaptic development, plasticity and signaling in the central nervous system. Neurochem. Int. 61, 848–853. Emond M. R., Montgomery J. M., Huggins M. L., Hanson J. E., Mao L., Huganir R. L. and Madison D. V. (2010) AMPA receptor subunits define properties of state-dependent synaptic plasticity. J. Physiol. 588, 1929–1946. Falsafi S. K., Ghafari M., Pollak A., Hoger H. and Lubec G. (2012) Hippocampal AMPA-type receptor complexes containing GluR3 and GluR4 are paralleling training in the Multiple T-Maze. Neurochem. Int. 60, 425–430. Ferreira A. (1999) Abnormal synapse formation in agrin-depleted hippocampal neurons. J. Cell Sci. 112, 4729–4738. Frischknecht R., Heine M., Perrais D., Seidenbecher C. I., Choquet D. and Gundelfinger E. D. (2009) Brain extracellular matrix affects AMPA receptor lateral mobility and short-term synaptic plasticity. Nat. Neurosci. 12, 897–904. Ghafari M., Falsafi S. K., Hoeger H. and Lubec G. (2012a) Hippocampal levels of GluR1 and GluR2 complexes are modulated by training in the Multiple T-maze in C57BL/6J mice. Brain Struct. Funct. 217, 353–362. Ghafari M., Hoger H., Keihan Falsafi S., Russo-Schlaff N., Pollak A. and Lubec G. (2012b) Mass spectrometrical identification of hippocampal NMDA receptor subunits NR1, NR2A-D and five novel phosphorylation sites on NR2A and NR2B. J. Proteome Res. 11, 1891–1896. Gogolla N., Caroni P., Luthi A. and Herry C. (2009) Perineuronal nets protect fear memories from erasure. Science 325, 1258–1261. Hensch T. K. (2005) Critical period plasticity in local cortical circuits. Nat. Rev. Neurosci. 6, 877–888. Herndon M. E. and Lander A. D. (1990) A diverse set of developmentally regulated proteoglycans is expressed in the rat central nervous system. Neuron 4, 949–961. Krugers H. J. and Hoogenraad C. C. (2009) Hormonal regulation of AMPA receptor trafficking and memory formation. Front. Synaptic Neurosci. 1, 2. Kurazono S., Okamoto M., Mori S. and Matsui H. (2001) Recombinant core protein fragment of phosphacan, a brain specific chondroitin sulfate proteoglycan, promote excitotoxic cell death of cultured rat hippocampal neurons. Neurosci. Lett. 304, 169–172. Lee A. M., Kanter B. R., Wang D., Lim J. P., Zou M. E., Qiu C., McMahon T., Dadgar J., Fischbach-Weiss S. C. and Messing R. O. (2013) Prkcz null mice show normal learning and memory. Nature 493, 416–419. Leonardo C. C., Eakin A. K., Ajmo J. M. and Gottschall P. E. (2008) Versican and brevican are expressed with distinct pathology in neonatal hypoxic-ischemic injury. J. Neurosci. Res. 86, 1106–1114. Morawski M., Bruckner G., Jager C., Seeger G., Matthews R. T. and Arendt T. (2012) Involvement of perineuronal and perisynaptic extracellular matrix in Alzheimer’s disease neuropathology. Brain Pathol. 22, 547–561. Nikonenko A., Schmidt S., Skibo G., Bruckner G. and Schachner M. (2003) Tenascin-R-deficient mice show structural alterations of

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

804

S. R. Saroja et al.

symmetric perisomatic synapses in the CA1 region of the hippocampus. J. Comp. Neurol. 456, 338–349. Orlando C., Ster J., Gerber U., Fawcett J. W. and Raineteau O. (2012) Perisynaptic chondroitin sulfate proteoglycans restrict structural plasticity in an integrin-dependent manner. J. Neurosci. 32, 18009– 18017. Pantazopoulos H., Woo T. U., Lim M. P., Lange N. and Berretta S. (2010) Extracellular matrix-glial abnormalities in the amygdala and entorhinal cortex of subjects diagnosed with schizophrenia. Arch. Gen. Psychiatry 67, 155–166. Reimers J. M., Milovanovic M. and Wolf M. E. (2011) Quantitative analysis of AMPA receptor subunit composition in addictionrelated brain regions. Brain Res. 1367, 223–233. Rust M. B., Gurniak C. B., Renner M. et al. (2010) Learning, AMPA receptor mobility and synaptic plasticity depend on n-cofilinmediated actin dynamics. EMBO J. 29, 1889–1902. Saghatelyan A. K., Gorissen S., Albert M., Hertlein B., Schachner M. and Dityatev A. (2000) The extracellular matrix molecule tenascinR and its HNK-1 carbohydrate modulate perisomatic inhibition and long-term potentiation in the CA1 region of the hippocampus. Eur. J. Neurosci. 12, 3331–3342. Sase S., Khan D., Sialana F., Hoger H., Russo-Schlaff N. and Lubec G. (2012) Modafinil improves performance in the multiple T-Maze and modifies GluR1, GluR2, D2 and NR1 receptor complex levels in the C57BL/6J mouse. Amino Acids 43, 2285–2292. Sase A., Dahanayaka S., Hoger H., Wu G. and Lubec G. (2013) Changes of hippocampal beta-alanine and citrulline levels are paralleling

early and late phase of retrieval in the Morris Water Maze. Behav. Brain Res. 249, 104–108. Schwenk J., Harmel N., Brechet A. et al. (2012) High-resolution proteomics unravel architecture and molecular diversity of native AMPA receptor complexes. Neuron 74, 621–633. Senkov O., Tikhobrazova O. and Dityatev A. (2012) PSA-NCAM: synaptic functions mediated by its interactions with proteoglycans and glutamate receptors. Int. J. Biochem. Cell Biol. 44, 591–595. Shah A. and Lodge D. J. (2013) A loss of hippocampal perineuronal nets produces deficits in dopamine system function: relevance to the positive symptoms of schizophrenia. Transl. Psychiatry 3, e215. Wan J., Csaszar E., Chen W. Q., Li K. and Lubec G. (2012) Proteins from Avastinâ (bevacizumab) show tyrosine nitrations for which the consequences are completely unclear. PLoS ONE 7, e34511. Welinder C. and Ekblad L. (2011) Coomassie staining as loading control in Western blot analysis. J. Proteome Res. 10, 1416–1419. Wenthold R. J., Petralia R. S., Blahos J., II and Niedzielski A. S. (1996) Evidence for multiple AMPA receptor complexes in hippocampal CA1/CA2 neurons. J. Neurosci. 16, 1982–1989. Yuen E. Y., Liu W., Karatsoreos I. N., Ren Y., Feng J., McEwen B. S. and Yan Z. (2011) Mechanisms for acute stress-induced enhancement of glutamatergic transmission and working memory. Mol. Psychiatry 16, 156–170. Zhou X. H., Brakebusch C., Matthies H. et al. (2001) Neurocan is dispensable for brain development. Mol. Cell. Biol. 21, 5970– 5978.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 130, 797--804

Hippocampal proteoglycans brevican and versican are linked to spatial memory of Sprague-Dawley rats in the morris water maze.

Proteoglycans (PGs) are major constituents of the extracellular matrix and have recently been proposed to contribute to synaptic plasticity. Hippocamp...
530KB Sizes 0 Downloads 4 Views