217

Journal of Alzheimer’s Disease 45 (2015) 217–233 DOI 10.3233/JAD-142469 IOS Press

Neuroprotective Effects of Hydrated Fullerene C60: Cortical and Hippocampal EEG Interplay in an Amyloid-Infused Rat Model of Alzheimer’s Disease Vasily Vorobyova,∗ , Vladimir Kaptsova , Rita Gordona , Ekaterina Makarovab , Igor Podolskib and Frank Sengpielc a Institute b Institute

of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region,

Russia c School of Biosciences and Neuroscience & Mental Health Research Institute, Cardiff University, Museum Avenue,

Cardiff, UK Handling Associate Editor: Vladimir Buchman

Accepted 17 November 2014

Abstract. We studied the effects of fullerene C60 nanoparticles, namely hydrated fullerene C60 (C60 HyFn), on interrelations between EEG frequency spectra from the frontal cortex and the dorsal hippocampus (CA1) on an amyloid-␤ (A␤) rat model of Alzheimer’s disease (AD). Infusion of A␤1-42 protein (1.5 ␮l) into the CA1 region two weeks before EEG testing diminished hippocampal theta (3.8–8.4 Hz) predominance and eliminated cortical beta (12.9–26.2 Hz) predominance observed in baseline EEG of rats infused with saline (control) or with C60 HyFn alone. In contrast, these A␤1-42 effects were abolished in rats pretreated with C60 HyFn, 30 min apart. Dopaminergic mediation in AD has been shown to be involved in neuronal plasticity and A␤ transformation in different ways. To clarify its role in the cortex-hippocampus interplay in the A␤ model of AD, we used peripheral injection of a dopamine agonist, apomorphine (APO), at a low dose (0.1 mg/kg). In rats infused with C60 HyFn or A␤1-42 alone, APO attenuated the cortical beta predominance, with immediate and delayed phases evident in the A␤1-42 rats. Pretreatment with C60 HyFn diminished the APO effect in the A␤1-42 -treated rats. Thus, we show that intrahippocampal injection of A␤1-42 dramatically disrupts cortical versus hippocampal EEG interrelations and that pretreatment with the fullerene eliminates this abnormality. We suggest that some effects of C60 HyFn may be mediated through presynaptic dopamine receptors and that water-soluble C60 fullerenes have a neuroprotective potential. Keywords: Amyloid-␤, brain oscillation, dopamine agonist, neurodegenerative disorder, neuroprotection

INTRODUCTION Amyloid-␤ peptides (A␤) are hypothesized to play a central role in the pathogenesis of Alzheimer’s disease (AD) [1–3]. A␤ are well known to initiate a series of ∗ Correspondence

to: Dr. Vasily Vorobyov, PhD, Institute of Cell Biophysics, Pushchino, 142290, Russia. Tel.: +7 4967 739 468; Fax: +7 4967 330 509; E-mail: [email protected].

pathological processes in the hippocampus and in the cortex (for review, see [4, 5]). Although recent studies demonstrate that neurodegeneration and memory impairment are observed before development of amyloidosis [6], A␤ involvement in the pathogenesis of AD is still widely accepted (for review, see [7]). Transgenic animal models of AD may facilitate a breakthrough in the understanding of disease mechanisms [8–11]. On the other hand, microinjections of

ISSN 1387-2877/15/$35.00 © 2015 – IOS Press and the authors. All rights reserved

218

V. Vorobyov et al. / Fullerene and EEG in AD Model

A␤ into the hippocampus allow investigation of direct effects of A␤ on the brain. In fact, intrahippocampal infusions of A␤1-42 revealed symptomatic and pathophysiological similarities of this model to AD and underline its usefulness for the development and evaluation of potential new drugs [12, 13]. A␤42/43 has been shown to initiate disruptions of hippocampal and cortical circuitries and their electrical oscillatory activities that are associated with cognitive dysfunctions and dementia typical of AD [6, 14]. In the electroencephalogram (EEG) frequency spectra of AD patients, an increase of power in the deltaand theta-bands (0.5–4.0 Hz and 4.0–8.0 Hz, respectively) and a reduction in the alpha- and beta-bands (8.0–12.0 Hz and 25–60 Hz, respectively) have been shown [15, 16]. In a few studies of the oscillatory processes in animal models of AD, reduced cortical theta versus enhanced beta-gamma activities in A␤PP/PS1mice [17] and increased delta versus reduced theta activities in the hippocampus and the cortex have been revealed [18]. In rats, a decrease in low-frequency theta-band oscillations and the weakening of binding between the dorsal hippocampus and the frontal cortex after microinjection of A␤25-35 into the lateral ventricle have been shown [19] that may underlie the spatial memory breakdown observed in AD. Fullerene C60 , a condensed ring aromatic compound with extended pi systems, is a novel carbon allotrope, whose water-soluble derivatives have potent and selective pharmacological effects on organs, cells, enzymes, and nucleic acids (for review, [20]). The main perspectives of water-soluble C60 are associated with its free radical-scavenging and direct nitric oxidequenching activities (for review, [21]), and with its ability to pass through lipid membranes into cells [22]. In the current study, we are using hydrated fullerene C60 (C60 HyFn) nanoparticles with combined colloidalhydrophilicpropertiesandhighantioxidantactivity[23] to inactivate the free radicals induced by A␤1-42 and linked to neurodegeneration in AD (for review, [24]). C60 HyFn has recently been shown to inhibit A␤ protein fibrillation in vitro and to improve spatial learning, disrupted by injection of A␤25-35 into the lateral ventricles in rats [25, 26]. These results are in line with those of subsequent studies (for review, [27]). The protective effect of C60 HyFn has been shown in ribosomal protein synthesis and in neurodegeneration [28]. However, how fullerenes affect oscillatory processes in the cortex and in the hippocampus remains unclear while an imbalance in network activities between brain areas has been shown to be important in cognitive processes [29] and in both AD [30] and Parkinson’s disease [31].

We therefore focus our efforts on estimating relative differences in frequency spectra of cortical and hippocampal EEGs in rats, intrahippocampally infused either with A␤1-42 alone or pretreated with hydrated fullerene C60 . Given a powerful A␤ regulation of synaptic transmission (for review, see [5]) and close association between cognitive dysfunction and receptor modification in AD and Parkinson’s disease (for review, see [21]), we are also interested in the EEG balance between these brain structures before and after peripheral injection of apomorphine (APO), an agonist of dopamine D1 /D2 receptors [32, 33]. Dysfunction of dopaminergic transmission has been shown to alter long-term depression-like plasticity in AD patients [34],whereasdopaminereceptoractivationbyAPOwas accompanied by promotion of A␤ degradation [35, 36]. We show for the first time that intrahippocampal injection of A␤1-42 dramatically disrupts cortical versus hippocampal EEG interrelations and that pretreatment with the fullerene eliminates this abnormality. We propose that effects of C60 HyFn may, at least in part, be mediated through presynaptic dopamine receptors and that water-soluble fullerenes C60 have a neuroprotective potential. MATERIALS AND METHODS Animals Twenty two Wistar male rats (290–330 g) obtained from the vivarium of the Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences were used in the EEG study. An additional thirty rats were involved in morphological and histochemical evaluation of neuronal viability. During one-month adaptation to the experimental conditions and consequent one-month experimentation, the animals were kept in a standard regime, with food and water ad libitum and a 12-h/12-h day/night cycle. The procedures were carried out in accord with the principles enunciated in the Guide for Care and Use of Laboratory Animals, NIH publication No 85-23, and approved by the Institutional Ethical Review Committee. Intrahippocampal infusions Anesthesia was induced with subcutaneous (s.c.) injection of tiletamine/zolazepam (Zoletil® , Virbac, France) in combination with xylazine (Rometar® , Bioveta, Czech Republic) at doses of 25 mg/kg and 1 mg/kg, respectively. Saline, C60 HyFn (at 0.5 nmol/␮l), and A␤1-42 protein (Sigma, USA, at 2 nmol/␮l), in

V. Vorobyov et al. / Fullerene and EEG in AD Model

volumes of 1.5–2.0 ␮l, were slowly infused (for ∼2 min) symmetrically into the dorsal hippocampus (CA1, AP −3.8, L 2.5, H 3.5) [37]. C60 HyFn was obtained courtesy of Dr. G. Andrievsky who developed this highly stable water-soluble form of the fullerene C60 (MER Corp, USA) [23]. A temporarily inserted guide cannula (blunt stainless steel needle, 0.8 mm inner diameter) with flexible silastic tubing attached to a 5-␮l Hamilton syringe was used for the infusion. The needle was kept in place for 3 min before withdrawal. Before injection of A␤1-42 , its aliquots were defrosted and incubated at 37◦ C for two days to aggregate the protein into fibrils [38] that were assessed by electron microscopy. Four groups of rats with two intrahippocampal injections (30 min apart) were studied. They were arbitrarily allocated to “saline + saline”, “C60 HyFn + saline”, “saline + A␤1-42 ”, and “C60 HyFn + A␤1-42 ” (4 rats in each group). In control (“na¨ive”) rats (n = 6), no cannulation was performed. Electrode implantation Immediately after the intrahippocampal injections, monopolar recording electrodes (nichrom wires, 0.1 mm in diameter, with tips free from insulation for 0.1 mm) were implanted symmetrically over the frontal cortex and underlying the dorsal hippocampus, CA1, of both hemispheres (AP −2.8, L 2.0, H 0 and 3.0, respectively) [37]. A reference electrode (stainless steel wire 0.4 mm in diameter) was placed in the nasal bone close to the midline and at a distance of 8-9 mm in front of bregma to minimize an influence of oscillations from the olfactory bulbs. Its tip was positioned at the optimal depth to minimize respiration artefacts. The implanted electrodes were linked to a female connector (Sullins Connector Solutions, CA) and fixed to the skull with dental cement. The electrode positions were verified postmortem in brain slices prepared with a freezing microtome.

219

was temporarily interrupted when they woke up. This ensured that, before commencing the main part of the experiment, the initial state of each animal was similar to enable correct comparison of the results obtained in different experimental groups. After the baseline interval, EEG recordings were continued either for 60 min on Day 11 or for 120 min after injection of APO (Sigma, Milan, Italy) at a dose of 0.1 mg/kg (s.c.) on Day 14. To minimize the effect of oxidation only freshly dissolved APO was used. Computation of EEG spectra Thefrequencyspectraofsuccessive12-sEEGepochs were analyzed on-line in the range of 0.5–25 Hz (bandpass filtered at 0.1–50 Hz) via a computerized system. Each epoch was digitized with a multichannel A/D DT2814 converter (Data Translation, Inc., Marlboro, MA, USA) using a sampling rate of 2 kHz. Taken into account the non-stationarity of EEG signals [39], we have used a modified version of period-amplitude analysis [40] which, contrary to the Fourier transform, is not affected by the non-stationary nature of the signals.Themainmodificationsofthealgorithmdeveloped by Stigsby et al. [41] were several types of frequency spectra normalization. The integrated power in twenty selected narrow EEG frequency bands and power ratios for each band over the integrated power in the 0.25–30.5 Hz range were calculated. The bands are marked in the text and figures by their center (mean) frequency values. The EEG spectra were averaged for every successive 10-min interval. The terms “lower” and “higher” in “classical”EEGbandsdelta(0.5–3.6Hz),theta(3.8–8.4 Hz), alpha (8.6–12.4 Hz), beta1 (12.9–16.8), and beta2 (18.4–26.2 Hz), are used below to differentiate corresponding frequency subranges of each band relative to its center frequency. For more details, see [42]. Morphological and histochemical evaluation of neuronal viability

EEG recording and APO treatment Between Day 4 and Day 11 after surgery, the rats were adapted, for 1 h/day, both to an experimental cage (Perspex, 25 × 25 × 30 cm) placed in an electrically shielded chamber and to handling (connecting/disconnecting the animal to/from the recording cable). EEG experiments were performed on Day 11 and on Day 14. Baseline EEGs were recorded for 10 min, starting about 20 min after placing the animal in the cage. During the baseline period, animals were in a relaxed posture, with closed eyes, and EEG recording

In an additional five groups of six rats in each group (na¨ive and infused into CA1 either with saline, C60 HyFn, A␤25-35 , or C60 HyFn+A␤25-35 ), we evaluated the distribution of surviving pyramidal neurons (which are most vulnerable to A␤ [43]), in the hippocampal slices on Day 14 versus Day 2 (three rats for the day from each group). A␤25-35 was chosen since this peptide, usually used in a synthetic form, on the one hand replicates A␤1-42 toxicity [44, 45] while on the other hand, it is not involved directly in recovery mechanisms associated with A␤1-42 [46].

220

V. Vorobyov et al. / Fullerene and EEG in AD Model

After decapitation under diethyl ether, one sample of brain was fixed in Carnow’s mixture (ethanol, chloroform, and acetic acid, at ratios of 6:3:1, respectively), whereas another one was put in 4% paraformaldehyde with PBS and then embedded in paraffin. Prepared blocks were 7-␮m sectioned on Leica’s microtome (Germany), and 25–30 sections from each animal were scanned and transferred through a digital camera to Axio Imager M1 optical microscope (Zeiss, Germany) for further analyses with use of Image J 1.44 (USA) software. Neuronal degeneration was evaluated after Nissl staining of the sections with cresyl violet (SigmaAldrich, St. Louis, USA). The immunohistochemical study was performed according to a standard technique [43]. To enhance sensitivity to A␤, the sections were treated with 70% formic acid for 4 min. To block the endogenous peroxidase activity, they were incubated in 0.5% H2 O2 /ethanol mixture, and 5% normal goat serum was used to block unspecific binding. The sections were further incubated with primary antibodies: rabbit polyclonal anti-A␤1-40 (1:400, Sigma) at a temperature of +4◦ C for 15 h. The antibodies were recognized by anti-rabbit IgG (1:800, Sigma) which were identified with StreptABComplex/HRP (1:800, Sigma). The reaction was visualized with 3,3’-diaminobenzidine. For control sections, the primary antibodies were replaced with PBS. For more details, see [28]. Statistics Differences in the averaged EEG spectra were evaluated by either a two-tailed non-parametric Wilcoxon test (p < 0.05 was considered statistically significant) or by one- or two-way ANOVA for repeated measures, when appropriate. Morphological data were analyzed by a Student’s t-test (p < 0.05 was deemed statistically significant).

after averaging in 10-min intervals (Fig. 2) and were relatively stable for 60 min of continuous recording (Table 1). Relative stability of the EEG spectral differences in other groups of rats was observed on Day 11, although their profiles were dependent on the treatment protocols (Table 2). On Day 14, individual baseline EEGs and their spectra in both the “saline + saline” (Fig. 3a and 3A, respectively) and “C60 HyFn + saline” (Fig. 3b and 3B, respectively) groups resembled those observed in the “na¨ıve” rats (Fig. 1). Intrahippocampal injection of A␤1-42 two weeks before EEG recordings (“saline + A␤1-42 ” group) dramatically slowed hippocampal and cortical baseline EEGs and made them similar both in individual 12-s fragments (Fig. 3c) and in their spectra (Fig. 3C). In contrast, pretreatment with the fullerene (“C60 HyFn + A␤1-42 ” group) practically eliminated this effect of A␤1-42 (Fig. 3D). The hippocampal-cortical differences observed in the individual baseline EEG spectra (Fig. 3) were more evident in those averaged for 10-min intervals. Briefly, in the groups of “saline + saline” (Figs. 4A, 5A) and “C60 HyFn + saline” (Figs. 4B, 5B), the main differences in cortical and hippocampal baseline EEG were associated with significant predominance of theta activity in the hippocampus and beta activity in the cortex (2-way ANOVA (brain areas X frequency subranges): F(1,36) = 18.2 and F(1,24) = 17.9, respectively, p < 0.001, in both). In the “saline + A␤1-42 ” group (Figs. 4C, 5C), the cortex-hippocampus EEG differences were negligible (2-way ANOVA: F(1,36) and F(1,24) 0.2, in both), moreover, they tended to be inverted in the beta range. In the “C60 Hy Fn + A␤1-42 ” group (Figs. 4D, 5D), the EEG differences between the cortex and the hippocampus were fully recovered in the beta band (2-way ANOVA: F(1,24) = 7.4, p = 0.012) and partly in the theta range. Apomorphine EEG effects

RESULTS Baseline EEG In “na¨ıve” rats, the baseline EEGs recorded on Day 14 from the cortex and the hippocampus were characterized by patterns of high amplitude slow waves, spindle activity and theta oscillations (Fig. 1A). The patterns were represented in EEG frequency spectra by peaks in the delta, beta, and theta bands, respectively, with theta activity dominating in the hippocampus and beta in the cortex (Fig. 1B). The cortical-hippocampal differences in EEG spectra reached significant values

To evaluate the APO specific effects on cortical versus hippocampal interrelations, relative differences between cortical and hippocampal EEG spectra after APO injection were normalized to those revealed in the baseline EEGs (Table 3). This made the enhanced sensitivity of EEG beta activity to APO in the “saline + A␤1-42 ” group more evident. Furthermore, the evolution of APO effects over time examined in the classical frequency bands showed the enhanced sensitivity to APO of the fullerene- versus saline-treated rats in both thethetaandthebetaband(Fig.6Aand6C,respectively; 2-way ANOVA: F(1,552) = 20 and F(1,360) = 60.4,

V. Vorobyov et al. / Fullerene and EEG in AD Model

221

Fig. 1. Typical patterns in baseline 12-s EEG fragments (A) and their frequency spectra (B) from the cortex and the hippocampus of the left hemisphere in a na¨ive rat. Ordinate on B is mean EEG amplitude in arbitrary units (AU), in 20 frequency subranges marked with their central (mean) value (in Hertz) on the abscissa. White and gray vertical bars on B mark the “classical” frequency bands in the EEG spectra (from left to right): delta, theta, alpha, and beta. Time calibration in A is 1 s; amplitude calibration (vertical bars left to the EEG recordings) is 100 ␮V. Table 1 Cortical versus hippocampal EEG interplay in na¨ive rats (n = 6) two weeks after electrode implantation

Values are calculated as a ratio of averaged baseline EEG amplitude differences per frequency subrange for (Cortex - Hippocampus)/Hippocampus (in %) in 10-min intervals (−10–0 – baseline interval, see text for details). Filled cells indicate significant prevalence (Wilcoxon test, p < 0.05) of the EEG rhythms in the cortex (dark colour) or in the hippocampus (grey colour). Twenty subranges in EEG frequency spectra are marked with their mean values, while Greek symbols (see the bottom line) denote so-called “classical” EEG frequency bands.

p  0.001, in both). A␤1-42 produced robust effect in the beta band (c.f., Fig. 6D and 6C, black and grey lines, respectively; 2-way ANOVA: F(1,360) = 137,

p  0.001, for rows, and F(11,360) = 4.6, p < 0.001, for interaction). The C60 HyFn pretreatment eliminated this effect of A␤1-42 (Fig. 6D, grey line; 2-way

222

V. Vorobyov et al. / Fullerene and EEG in AD Model Table 2 Cortical versus hippocampal baseline EEG interplay in rats eleven days after intrahippocampal microinjections of saline, C60 HyFn, and A␤1-42 at different combinations (n = 4, in each group)

Values are calculated as a ratio of averaged baseline EEG amplitude differences per frequency subrange, (Cortex - Hippocampus) / Hippocampus (in %) in 10-min intervals (−10–0 – baseline interval, see text for details). Filled cells indicate significant prevalence (Wilcoxon test, p < 0.05) of the EEG rhythms in the cortex (dark color) or in the hippocampus (grey color). Twenty subranges in EEG frequency spectra are marked with their mean values, while Greek symbols (see the bottom line) denote so-called “classical” EEG frequency bands.

ANOVA: F(1,360) = 47.8, p < 0.001, for rows, and F(11,360) = 3.8, p < 0.001, for interaction). Interestingly, the fullerene effect in the theta band was diminished by A␤1-42 (c.f. Fig. 6A and 6B, black and grey lines, respectively; 2-way ANOVA: F(1,552) = 11, p < 0.001). Although the fullerene effect (versus saline) in the beta band (Fig. 6C, black line; 2-way ANOVA: F(1,360) = 46.7, p  0.001) tended to be attenuated by A␤1-42 (Fig. 6D, grey line), the difference did not reach significance (2-way ANOVA: F(1,360) = 3.4, p = 0.07). Finally, the APO effect in the beta band was more powerfully expressed (2-way ANOVA: F(1,360) = 25, p  0.001, for rows, and F(11,360) = 5.3, p  0.001, for interaction) in rats treated with A␤1-42 alone (Fig. 6D, black line) than in

those treated with the fullerene alone (Fig. 6C, black line). Behavior During the baseline period, animals were in a relaxed posture, with closed eyes, that was temporarily interrupted when they woke up. APO (0.1 mg/kg, s.c.) injection provoked, with a short delay (1-2 min), similar changes in behavior and in particular yawning, in rats from different groups. The time-course of these patterns of behavior in individual rats varied in phase and duration, indicating therefore that the change in behavior is unlikely to be the main cause of the EEG changes revealed in our study.

V. Vorobyov et al. / Fullerene and EEG in AD Model

223

Fig. 2. Differences in the frequency spectra of 12-s baseline EEG fragments averaged for 10-min intervals in the cortex (gray lines) and hippocampus (black lines) in six na¨ive rats. Ordinate on A is mean EEG amplitude in arbitrary units (AU) in 20 frequency subranges marked with their central (mean) value (in Hertz) on the abscissa. Ordinate on B is relative difference of the EEG amplitudes between cortex and hippocampus calculated as ratio (Cortex - Hippocampus) / Hippocampus, in %. White and gray vertical bars on A–D mark the “classical” frequency bands in the EEG spectra (from left to right): delta, theta, alpha, and beta. Diamonds on A denote significant difference between the cortex and hippocampus (p < 0.05, Wilcoxon test). Error bars show 1 SEM.

Morphology and histochemistry On Day 14, pyramidal neurons from CA1 in na¨ive rats had uniformly stained cytoplasm and nucleoplasm, distinct plasmatic and nuclear membranes, and well-

defined nuclei (Fig. 7A, B). Similar “healthy” images were typical for the hippocampi of rats infused with saline (Fig. 7C, D) or C60 HyFn (Fig. 7E, F), whereas A␤25-35 produced neuronal necrosis expressed in swelling of the cytoplasm and its vacuolization,

224

V. Vorobyov et al. / Fullerene and EEG in AD Model

Fig. 3. Typical patterns in baseline 12-s EEG fragments (a–d) and their frequency spectra (A–D) from the cortex and the hippocampus in individual rats from different groups: “saline + saline” (a & A), “C60 HyFn + saline” (b & B), “saline + A␤1-42 ” (c & C), and “C60 HyFn + A␤1-42 ” (d & D). Ordinate on A – B is mean EEG amplitude in arbitrary units (AU), in 20 frequency subranges marked with their central (mean) value (in Hertz) on the horizontal axis (abscissa). White and gray vertical bars on A–D mark the “classical” frequency bands in the EEG spectra (from left to right): delta, theta, alpha, and beta. Time calibration on a-d is 1 s; amplitude calibration (vertical bars left to the EEG recordings) is 100 ␮V.

V. Vorobyov et al. / Fullerene and EEG in AD Model

225

Fig. 4. Differences in the frequency spectra of baseline 12-s EEG fragments averaged for 10-min intervals from the cortex (gray lines) and the hippocampus (black lines) in rats from different groups (n = 4, in each group): “saline + saline” (A), “C60 HyFn + saline” (B), “saline + A␤1-42 ” (C), and “C60 HyFn + A␤1-42 ” (D). Ordinate is mean EEG amplitude in arbitrary units (AU) in 20 frequency subranges marked with their central (mean) value (in Hertz) on the abscissa. White and gray vertical bars on A–D mark the “classical” frequency bands in the EEG spectra (from left to right): delta, theta, alpha, and beta. Diamonds denote significant difference between the cortex and hippocampus (p < 0.05, Wilcoxon test). Error bars show 1 SEM.

226

V. Vorobyov et al. / Fullerene and EEG in AD Model

Fig. 5. Cortical versus hippocampal EEG relative frequency spectra profiles averaged for 10-min baseline intervals in rats from different groups (n = 4, in each group): “saline + saline” versus “C60 HyFn + saline” (A, C), “saline + A␤1-42 ” versus “C60 HyFn + A␤1-42 ” (B, D). Ordinate is ratio of averaged EEG frequency band amplitudes from the cortex and the hippocampus calculated as (Cortex-Hippocampus)/Hippocampus, in %. Abscissa is frequency subrange marked with its central (mean) value, in Hertz. White and gray vertical bars mark the “classical” frequency bands in the EEG spectra (from left to right): delta, theta, alpha, and beta. Error bars show 1 SEM.

V. Vorobyov et al. / Fullerene and EEG in AD Model

forming huge clusters of indistinct and dark pyramidal neurons (Fig. 7G, H). In rats pretreated with C60 HyFn, A␤25-35 was obviously ineffective (Fig. 7I, J). These differences were confirmed by quantitative analyses of surviving cells distribution in the hippocampi of rats from different groups (Fig. 8A, Day 14). Interestingly, the distributions on Day 2 were practically identical in all groups (Fig. 8A, Day 2) that emphasized specificity of the A␤25-35 effect on Day 14. The immunohistochemical analyses of the brain sections on Day 14 after A␤25-35 infusion revealed significantly enlarged deposits of diffuse A␤1-42 in the cytoplasm of CA1 pyramidal neurons (Fig. 8B), whereas in salinepretreated rats, no immunostained cells were observed. In rats pretreated with C60 HyFn, the A␤25-35 effect was practically eliminated (Fig. 8B).

DISCUSSION In our study, A␤1-42 , infused in the hippocampus two weeks before EEG testing, eliminated both hippocampal theta and cortical beta wave predominance in baseline EEG spectra observed in both na¨ive and salinetreated rats. However, intrahippocampal pretreatment with hydrated fullerene C60 (C60 HyFn), which when infused alone tended to increase the baseline differences, diminished the effect of A␤1-42 . Following apomorphine-injection, the fullerene practically eliminated A␤1-42 -evoked changes in the beta range. Baseline EEG in our study was recorded during a relatively stable sleep-like state (see details in the Materials and Methods). The differences in frequency spectra of cortical and hippocampal baseline EEG in control rats (Fig. 2) are in line with the idea that various cortical and subcortical brain structures have specific sleep characteristics [47]. The effects of C60 HyFn (Figs. 4B, 5B) and A␤1-42 (Figs. 4C, 5C) on the baseline EEG indicate a rearrangement of neuronal networks in different brain areas involved in this physiological state. Sleep has been shown to play a role in memory consolidation that has been supposed to be based on an information transfer between the hippocampus and the neocortex through neurophysiological mechanisms that generate sleep-specific slow waves, spindles, and ripples (for review, see [48, 49]). Thus, the elimination/inversion of the beta predominance in the cortex after A␤1-42 treatment (Figs. 4C, 5C) is in line both with the memory/learning impairment shown in the A␤25-35 model of AD in rats [25, 26] and with the reduction of EEG sleep spindles observed in AD patients suffering disrupted performance [50].

227

The preservation of the sleep-dependent cortical beta predominance from the A␤1-42 -produced toxicity by C60 HyFn (Figs. 4D, 5D) may additionally highlight its protective and therapeutic potential for neurodegenerative diseases [28, 51, 52] and open the gate for the understanding of its neurophysiological mechanisms. In particular, our data indicate that C60 HyFn is able to enhance some specific activities in the cortex and in the hippocampus (c.f. Figs. 4B, 5B and Figs. 4A, 5A, in the beta and the theta range, respectively). This is in line with in vitro experiments on hippocampal slices where C60 HyFn-enhanced excitability of pyramidal neurons has been demonstrated [53]. The hippocampal theta predominance in baseline EEG spectra from control (na¨ive) rats (Fig. 2, Table 1) seems to reflect an optimal level of interrelation between the cortex and the hippocampus tuned to play a role, after awaking, in the sensory-motor integration [54] and in the neural coding of place, in particular (for review, see [55, 56]). The elimination of the hippocampal theta predominance in the A␤1-42 -treated rats (Figs. 4C, 5C) and its partial preservation from the A␤1-42 -produced toxicity by C60 HyFn (Figs. 4D, 5D) are in line with the results on spatial learning obtained in the A␤25-35 model of AD in rats [25, 26, 57]. Apomorphine, an agonist of DA receptors, has been shown to stimulate postsynaptic receptors at moderate to high doses (0.5–1.0 mg/kg) whereas it activates presynaptic receptors at low doses, tending to cause sleep [32] (for review, see [58]). In our study, a low dose of APO (0.1 mg/kg), produced significant suppression of the cortical beta dominance in rats treated with A␤1-42 alone, with two phases in the time-course of the effect (Table 3, Fig. 6D, black line). This might provide additional support for the involvement of presynaptic DA receptors in A␤1-42 -treated rats since immediate and delayed effects of APO at low doses have been shown in previous studies [59, 60]. Suppression of the cortical beta dominance in baseline EEG from A␤1-42 -treated rats (Fig. 5C) and its potentiation by APO (Fig. 6D, black line, Table 3) are thought to have common mediatory mechanism(s) and might be associated, in particular, with an inflammatory ability of A␤1-42 [12]. Indeed, bilateral intrahippocampal injection of aggregated A␤1-42 has been shown to produce protracted (above 30 days) inflammatory-associated impairments in rat’s operant behavior [13]. Inflammatory reactions are well known to be important factors in AD pathogenesis, however, microglia activated through receptors for GABA, acetylcholine, and adrenaline has been revealed to suppress inflammatory responses whereas its activation

228

V. Vorobyov et al. / Fullerene and EEG in AD Model Table 3 Cortical versus hippocampal EEG interplay after subcutaneous injections of apomorphine (0.1 mg/kg) in rats from different groups (n = 4, in each group)

Values are calculated as a ratio of averaged EEG amplitude differences per frequency subrange, (Cortex - Hippocampus) / Hippocampus (in %) in 10-min intervals after injection of apomorphine (normalized versus baseline values). Filled cells indicate significant prevalence (Wilcoxon test, p < 0.05) of the EEG rhythms in the cortex (dark color) or in the hippocampus (grey color). Twenty subranges in EEG frequency spectra are marked with their mean values, while Greek symbols (bottom line) denote so-called “classical” EEG frequency bands.

V. Vorobyov et al. / Fullerene and EEG in AD Model

229

Fig. 6. Evolution of cortical-hippocampal EEG interrelations in the theta and the beta frequency bands (A, B and C, D, respectively) after APO injection (0.1 mg/kg, s.c.) in rats from different groups (n = 4, in each group): “saline + saline” versus “C60 HyFn + saline” (A, C), “saline + A␤1-42 ” versus “C60 HyFn + A␤1-42 ” (B, D). Ordinate is the ratio of averaged EEG frequency band amplitudes in the cortex and the hippocampus calculated as (Cortex-Hippocampus)/ Hippocampus, in %. Normalized baseline values for each of the groups are denoted by “0” at ‘BL’ point on abscissa and extended as a horizontal dashed line. Abscissa shows time after apomorphine injection, marked in 10-min intervals. Diamonds denote significant difference between the groups (p < 0.05, Wilcoxon test). Error bars show 1 SEM.

through receptors for ATP and adenosine stimulates release of pro-inflammatory factors (for review, see [61]). Interestingly, glutamate and dopamine can be pro- or anti-inflammatory depending on the receptor subtype expressed in microglia. Pharmacological blockade of dopamine D2 receptors has been shown to suppress both tau aggregation and neurotoxicity [62] while their activation by rotigotine, an agonist with high affinity for the D2 /D3 receptors, results in both increased cortical excitability and restored central cholinergic transmission in AD patients [63]. The brain inflammation has been shown to be accompanied by increased concentration of serum phenylalanine, i.e., by its impaired conversion into the neurotransmitters, in particular, dopamine [64]. This dopamine

pool shrinkage reduces, in turn, the probability of spontaneous binding of dopamine to its receptors, thus opening the gate for different effects of exogenetic APO molecules depending on their agonist affinities for various types of dopamine receptors [65]. Significantly diminished beta predominance in the cortical EEG after injection of APO in A␤1-42 -treated rats (Fig. 6D, gray line) is in line with this suggestion and with supposedly potentiating effect of APO on A␤associated toxicity [62]. Finally, the enlarged deposits of diffuse A␤1-42 in the cytoplasm of CA1 pyramidal neurons revealed after pretreatment with A␤25-35 (Fig. 8B) seems to be a result of its inflammatory ability that is typical for A␤ (for review, see [66]), and for A␤1-42 in particular [12, 13].

230

V. Vorobyov et al. / Fullerene and EEG in AD Model

Fig. 7. Microphotographs of the hippocampal (CA1) sections stained with cresyl violet in rats from different groups on Day 14 (n = 3, in each group): na¨ive (A, B) and after intrahippocampal infusion of either saline (C, D), C60 HyFn (E, F), A␤25-35 , (G, H), or C60 HyFn + A␤25-35 (I, J).

Thus, the neuroprotective effect of C60 HyFn (Fig. 6D, grey line) against the A␤ toxicity, seemingly associated with the induction of diffuse A␤1-42 deposits in the neuronal cytoplasm (Fig. 8B), is thought to be produced by the well-known scavenging activity of fullerenes against free radicals (for review, see [67]) and, presumably, against the pro-inflammatory cytokines released by activated astrocytes. The latter

might allow both the disruption of a vicious inflammatory cycle of A␤ generation between astrocytes and the redirection of cytokines from their neurodegenerative to neuroprotective roles (for review, see [66]). Regardless of possible mechanisms underlying the effects of C60 HyFn, this approach could be one of potentially protective interventions forwarding future investigation in AD (for review, see [68]).

V. Vorobyov et al. / Fullerene and EEG in AD Model

231

Author’s disclosures available online (http://j-alz. com/manuscript-disclosures/14-2469r1). REFERENCES [1]

[2] [3] [4] [5]

[6]

[7]

[8]

Fig. 8. Intact cell density (A) in the hippocampal (CA1) sections from rats on Day 2 and Day 14 after intrahippocampal infusion of saline (I), C60 HyFn (II), A␤25-35 (III), or C60 HyFn + A␤25-35 (IV) (n = 3, in each group/day) and distribution of diffuse A␤1-42 deposits in the cytoplasm of CA1 pyramidal neurons on Day 14 in rats from different groups (n = 3, in each group). * marks significant difference (p < 0.001, Student’s t-test).

[9]

[10]

CONCLUSIONS [11]

Two weeks after intrahippocampal amyloid-␤1-42 infusion, cortical versus hippocampal EEG interplay was dramatically disrupted. Pretreatment with hydrated fullerene C60 significantly attenuated the amyloid-␤1-42 effect. Dopaminergic D1 /D2 receptors are involved in the A␤1-42 effects. The water-soluble fullerenes C60 have a neuroprotective potential.

[12]

[13]

ACKNOWLEDGMENTS [14]

The authors are extremely grateful to Dr. G. Andrievsky of the Institute of Scintillating Materials, National Academy of Sciences (Ukraine), for a generous supply of the hydrated fullerene C60 . This work was supported by a grant of “Basic sciences for medicine” from the Russian Academy of Sciences (IP) and by the State Contract of RF No P1052 (IP).

[15]

[16]

Hardy J, Allsop D (1991) Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol Sci 12, 383-388. Hardy J (2006) A hundred years of Alzheimer’s disease research. Neuron 52, 3-13. Selkoe DJ (2001) Clearing the brain’s amyloid cobwebs. Neuron 32, 177-180. German DC, Eisch AJ (2004) Mouse models of Alzheimer’s disease: Insight into treatment. Rev Neurosci 15, 353-369. Turner PR, rsquo O, Connor K, Tate WP, Abraham WC (2003) Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory. Prog Neurobiol 70, 1-32. Goutagny R, Gu N, Cavanagh C, Jackson J, Chabot JG, Quirion JG, Krantic S, Williams S (2013) Alterations in hippocampal network oscillations and theta-gamma coupling arise before A␤ overproduction in a mouse model of Alzheimer’s disease. Eur J Neurosci 37, 1896-1902. Dong S, Duan Y, Hu Y, Zhao Z (2012) Advances in the pathogenesis of Alzheimer’s disease: A re-evaluation of amyloid cascade hypothesis. Transl Neurodegener 1, 18. Bobkova NV, Garbuz DG, Nesterova I, Medvinskaya N, Samokhin A, Alexandrova I, Yashin V, Karpov V, Kukharsky MS, Ninkina NN, Smirnov AA, Nudler E, Evgen’ev M (2014) Therapeutic effect of exogenous hsp70 in mouse models of Alzheimer’s disease. J Alzheimers Dis 38, 425-435. Devi L, Ohno M (2010) Genetic reductions of beta-site amyloid precursor protein-cleaving enzyme 1 and amyloid-beta ameliorate impairment of conditioned taste aversion memory in 5XFAD Alzheimer’s disease model mice. Eur J Neurosci 31, 110-118. Peters OM, Connor-Robson N, Sokolov VB, Aksinenko AY, Kukharsky MS, Bachurin SO, Ninkina N, Buchman VL (2013) Chronic administration of dimebon ameliorates pathology in TauP301S transgenic mice. J Alzheimers Dis 33, 1041-1049. Peters OM, Connor-Robson N, Sokolov VB, Aksinenko AY, Kukharsky MS, Bachurin SO, Ninkina N, Buchman VL (2013) Chronic administration of Dimebon does not ameliorate amyloid-␤ pathology in 5xFAD transgenic mice. J Alzheimers Dis 36, 589-596. McLarnon JG, Ryu JK (2008) Relevance of Abeta1-42 intrahippocampal injection as an animal model of inflamed Alzheimer’s disease brain. Curr Alzheimer Res 5, 475-480. O’Hare E, Scopes DI, Treherne JM, Monaghan J, Palmer PM, Amijee H, Kim EM (2011) Novel anti-inflammatory compound SEN1176 alleviates behavioral deficits induced following bilateral intrahippocampal injection of aggregated amyloid-␤1-42. J Alzheimers Dis 25, 219-229. Babiloni C, Vecchio F, Lizio R, Ferri R, Rodriguez G, Marzano N, Frisoni GB, Rossini PM (2011) Resting state cortical rhythms in mild cognitive impairment and Alzheimer’s disease: Electroencephalographic evidence. J Alzheimers Dis 26(Suppl 3), 201-214. Delbeuck X, Van der Linden M, Collette F (2003) Alzheimer’s disease as a disconnection syndrome? Neuropsychol Rev 13, 79-92. Jeong J (2004) EEG dynamics in patients with Alzheimer’s disease. Clin Neurophysiol 115, 1490-1505.

232 [17]

[18]

[19]

[20]

[21]

[22]

[23]

[24]

[25]

[26]

[27]

[28]

[29] [30]

[31]

[32]

[33]

[34]

V. Vorobyov et al. / Fullerene and EEG in AD Model Wang J, Ikonen S, Gurevicius K, Groen T, Tanila H (2002) Alteration of cortical EEG in mice carrying mutated human APP transgene. Brain Res 943, 181-190. S´anchez-Alavez M, Chan SL, Mattson MP, Criado JR (2007) Electrophysiological and cerebrovascular effects of the alphasecretase-derived form of amyloid precursor protein in young and middleaged rats. Brain Res 1131, 112-117. Mugantseva EA, Podolski IY (2009) Animal model of Alzheimer’s disease: Characteristics of EEG and memory. Cent Eur J Biol 4, 507-514. Satoh M, Takayanagi I (2006) Pharmacological studies on fullerene (C60), a novel carbon allotrope, and its derivatives. J Pharmacol Sci 100, 513-518. Xu Y, Yan J, Zhou P, Li J, Gao H, Xia Y, Wang Q (2012) Neurotransmitter receptors and cognitive dysfunction in Alzheimer’s disease and Parkinson’s disease. Prog Neurobiol 97, 1-13. Cheng B, Gong H, Xiao H, Petersen RB, Zheng L, Huang K (2013) Inhibiting toxic aggregation of amyloidogenic proteins: A therapeutic strategy for protein misfolding diseases. Biochim Biophys Acta 1830, 4860-4871. Andrievsky GV, Bruskov VI, Tykhomyrov AA, Gudkov SV (2009) Peculiarities of the antioxidant and radioprotective effects of hydrated C60 fullerene nanostuctures in vitro and in vivo. Free Radic Biol Med 47, 786-793. Butterfield DA (2002) Amyloid beta-peptide (1-42)-induced oxidative stress and neurotoxicity: Implications for neurodegeneration in Alzheimer’s disease brain. A review. Free Radic Res 36, 1307-1313. Podolski IY, Podlubnaya ZA, Kosenko EA, Mugantseva EA, Makarova EG, Marsagishvili LG, Shpagina MD, Kaminsky YG, Andrievsky GV, Klochkov VK (2007) Effects of hydrated forms of C60 fullerene on amyloid 1-peptide fibrillization in vitro and performance of the cognitive task. J Nanosci Nanotechnol 7, 1479-1485. Podolski IY, Podlubnaya ZA, Godukhin OV (2010) Fullerenes C60, antiamyloid action, the brain, and cognitive processes. Biophysics 55, 71-76 (in Russian). Li C, Mezzenga R (2013) The interplay between carbon nanomaterials and amyloid fibrils in bio-nanotechnology. Nanoscale 5, 6207-6218. Makarova EG, Gordon RY, Podolski IY (2012) Fullerene C60 prevents neurotoxicity induced by intrahippocampal microinjection of amyloid-beta peptide. J Nanosci Nanotechnol 12, 119-126. Ward LM (2003) Synchronous neural oscillations and cognitive processes. Trends Cogn Sci 7, 553-559. Bobkova N, Vorobyov V, Medvinskaya N, Aleksandrova I, Nesterova I (2008) Interhemispheric EEG differences in olfactory bulbectomized rats with different cognitive abilities and brain beta-amyloid levels. Brain Res 1232, 185-194. Vorobyov V, Sengpiel F (2008) Apomorphine-induced differences in cortical and striatal EEG and their glutamatergic mediation in 6-hydroxydopamine-treated rats. Exp Brain Res 191, 277-287. Carlsson A, Kehr W, Lindqvist M (1977) Agonist–antagonist interaction on dopamine receptors in brain, as reflected in the rates of tyrosine and tryptophan hydroxylation. J Neural Transm 40, 99-113. Poewe W, Wenning GK (2000) Apomorphine: An underutilized therapy for Parkinson’s disease. Mov Disord 15, 789-794. Koch G, Esposito Z, Codec, agrave C, Mori F, Kusayanagi H, Monteleone F, Di Lorenzo F, Bernardi G, Martorana A (2011) Altered dopamine modulation of LTD-like plastic-

[35]

[36] [37] [38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48] [49] [50]

[51]

[52]

[53]

ity in Alzheimer’s disease patients. Clin Neurophysiol 122, 703-707. Himeno E, Ohyagi Y, Ma L, Nakamura N, Miyoshi K, Sakae N, Motomura K, Soejima N, Yamasaki R, Hashimoto T, Tabira T, LaFerla FM, Kira J (2011) Apomorphine treatment in Alzheimer mice promoting amyloid-␤ degradation. Ann Neurol 69, 248-256. Ribariˇc S (2012) The pharmacological properties and therapeutic use of apomorphine. Molecules 17, 5289-5309. Pellegrino LJ, Pellegrino AS, Cushman AJ (1979) A Stereotaxic Atlas of the Rat Brain, Plenum Press, New York. Garai K, Frieden C (2013) Quantitative analysis of the time course of A␤ oligomerization and subsequent growth steps using tetramethylrhodamine-labeled A␤. Proc Natl Acad Sci U S A 110, 3321-3326. Gray CM, Engel AK, Koenig P, Singer W (1992) Synchronization of oscillatory neuronal responses in cat striate cortex: Temporal properties. Vis Neurosci 8, 337-347. Gal’chenko AA, Vorobyov VV (1999) Analysis of electroencephalograms using a modified amplitude-interval algorithm. Neurosci Behav Physiology 29, 157-160. Stigsby B, Obrist WD, Sulg IA (1973) Automatic data acquisition and period-amplitude analysis of the electroencephalogram. Comput Programs Biomed 3, 93-104. Vorobyov V, Kaptsov V, Kovalev G, Sengpiel F (2011) Effects of nootropics on the EEG in conscious rats and their modification by glutamatergic inhibitors. Brain Res Bull 85, 123-132. Nie J, Luo Y, Huang XN, Gong QH, Wu Q, Shi JS (2010) Icariin inhibits beta-amyloid peptide segment 25-35 induced expression of beta-secretase in rat hippocampus. Eur J Pharmacol 626, 213-218. Iversen LL, Mortishire-Smith RJ, Pollack SJ, Shearman MS (2003) The toxicity in vitro of beta-amyloid protein. Biochem J 311(Pt 1), 1-16. Frozza RL, Horn AP, Hoppe JB, Sim, atilde, o F, Gerhardt D, Comiran RA, Salbego CG (2009) A comparative study of beta-amyloid peptides Abeta1-42 and Abeta25-35 toxicity in organotypic hippocampal slice cultures. Neurochem Res 34, 295-303. Plant LD, Boyle JP, Smith IF, Peers C, Pearson HA (2003) The production of amyloid beta peptide is a critical requirement for the viability of central neurons. J Neurosci 23, 5531-5535. Nobili L, De Gennaro L, Proserpio P, Moroni F, Sarasso S, Pigorini A, De Carli F, Ferrara M (2012) Local aspects of sleep: Observations from intracerebral recordings in humans. Prog Brain Res 199, 219-232. Fell J, Axmacher N (2011) The role of phase synchronization in memory processes. Nat Rev Neurosci 12, 105-118. Inostroza M, Born J (2013) Sleep for preserving and transforming episodic memory. Ann Rev Neurosci 36, 79-102. Rauchs G, Schabus M, Parapatics S, Bertran F, Clochon P, Hot P, Denise P, Desgranges B, Eustache F, Gruber G, Anderer P (2008) Is there a link between sleep changes and memory in Alzheimer’s disease? Neuroreport 19, 1159-1162. Dugan LL, Turetsky DM, Du C, Lobner D, Wheeler M, Almli CR, Shen CK, Luh TY, Choi DW, Lin TS (1997) Carboxyfullerenes as neuroprotective agents. Proc Natl Acad Sci U S A 94, 9434-9439. Dugan LL, Lovett EG, Quick KL, Lotharius J, Lin TT, O’Malley KL (2001) Fullerene-based antioxidants and neurodegenerative disorders. Parkinsonism Relat Disord 7, 243-246. Kordonets OL, Godukhin OV, Podolski IYa (2008) Effect of fullerenes C60 on the activity of pyramidal neurons in the

V. Vorobyov et al. / Fullerene and EEG in AD Model

[54]

[55] [56] [57]

[58] [59]

[60]

[61] [62]

CA1 field of rat hippocampal slices. Dokl Biol Sci 423, 1-3 (in Russian). Bland BH, Oddie SD (2001) Theta band oscillation and synchrony in the hippocampal formation and associated structures: The case for its role in sensorimotor integration. Behav Brain Res 127, 119-136. Lisman JE, Jensen O (2013) The ␪ − ␥ neural code. Neuron 77, 1002-1016. O’Keefe J (1993) Hippocampus, theta, and spatial memory. Curr Opin Neurobiol 3, 917-924. Quick KL, Ali SS, Arch R, Xiong C, Wozniak D, Dugan LL (2008) A carboxyfullerene SOD mimetic improves cognition and extends the lifespan of mice. Neurobiol Aging 29, 117128. Tamminga CA (2002) Partial dopamine agonists in the treatment of psychosis. J Neural Transm 109, 411-420. Bagetta G, De Sarro G, Priolo E, Nistic, ograve G (1988) Ventral tegmental area: Site through which dopamine D2receptor agonists evoke behavioural and electrocortical sleep in rats. Br J Pharmacol 95, 860-866. Sebban C, Zhang XQ, Tesolin-Decros B, Millan MJ, Spedding M (1999) Changes in EEG spectral power in the prefrontal cortex of conscious rats elicited by drugs interacting with dopaminergic and noradrenergic transmission. Br J Pharmacol 128, 1045-1054. Lee M (2013) Neurotransmitters and microglial-mediated neuroinflammation. Curr Protein Pept Sci 14, 21-32. McCormick AV, Wheeler JM, Guthrie CR, Liachko NF, Kraemer BC (2013) Dopamine D2 receptor antagonism

[63]

[64]

[65]

[66]

[67]

[68]

233

suppresses tau aggregation and neurotoxicity. Biol Psychiatry 73, 464-471. Martorana A, Di Lorenzo F, Esposito Z, Lo Giudice T, Bernardi G, Caltagirone C, Koch G (2013) Dopamine D--2 agonist rotigotine effects on cortical excitability and central cholinergic transmission in Alzheimer’s disease patients. Neuropharmacology 64, 108-113. Wisman LA, Sahin G, Maingay M, Leanza G, Kirik D (2008) Functional convergence of dopaminergic and cholinergic input is critical for hippocampus-dependent working memory. J Neurosci 28, 7797-7807. Millan MJ, Maiofiss L, Cussac D, Audinot V, Boutin JA, Newman-Tancredi A (2002) Differential actions of antiparkinson agents at multiple classes of monoaminergic receptor. I. A multivariate analysis of the binding profiles of 14 drugs at 21 native and cloned human receptor subtypes. J Pharmacol Exp Ther 303, 791-804. Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Hoi Yu AC (2011) Astrocytes: Implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Curr Alzheimer Res 8, 67-80. Yang X, Ebrahimi A, Li J, Cui Q (2014) Fullerenebiomolecule conjugates and their biomedicinal applications. Int J Nanomedicine 9, 77-92. Ubhi K, Masliah E (2013) Alzheimer’s disease: Recent advances and future perspectives. J Alzheimers Dis 33(Suppl. 1), 185-194.

Neuroprotective effects of hydrated fullerene C60: cortical and hippocampal EEG interplay in an amyloid-infused rat model of Alzheimer's disease.

We studied the effects of fullerene C60 nanoparticles, namely hydrated fullerene C60 (C60HyFn), on interrelations between EEG frequency spectra from t...
1MB Sizes 2 Downloads 5 Views