This study examined whether ethyl pyruvate (EP) promotes the survival of nigrostriatal dopaminergic (DA) neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson’s disease. MPTP induced degeneration of nigrostriatal DA neurons and glial activation as visualized by tyrosine hydroxylase, macrophage Ag complex-1, and/or glial fibrillary acidic protein immunoreactivity. Western blotting and immunohistochemistry showed activation of microglial NADPH oxidase and astroglial myeloperoxidase (MPO) and subsequent reactive oxygen species/reactive nitrogen species production and oxidative DNA damage in the MPTP-treated substantia nigra. Treatment with EP prevented degeneration of nigrostriatal DA neurons, increased striatal dopamine levels, and improved motor function. This neuroprotection afforded by EP was associated with the suppression of astroglial MPO expression, NADPH oxidase-, and/or inducible NO synthase-derived reactive oxygen species/reactive nitrogen species production by activated microglia. Interestingly, EP was found to protect DA neurons from 1-methyl-4-phenyl-pyridinium neurotoxicity in cocultures of mesencephalic neurons and microglia but not in neuron-enriched mesencephalic cultures devoid of microglia. The present findings show that EP may inhibit glial-mediated oxidative stress, suggesting that EP may have therapeutic value in the treatment of aspects of Parkinson’s disease related to glia-derived oxidative damage.

Parkinson’s disease (PD) is a neurodegenerative disorder characterized by motor symptoms, including resting tremor, muscle rigidity, bradykinesia (slowness of movement), and postural instability (1, 2). The pathological hallmark of PD is the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) that project to the striatum (STR), which plays an essential role in normal motor function (2). Clinical parkinsonian symptoms arise when a majority (∼60–70%) of SNpc DA neurons are lost, resulting in reduced dopamine levels in the nigrostriatal system (3). Although PD is a sporadic disease of unknown pathogenesis, accumulating evidence suggests that glial activation-derived oxidative stress increases the risk of developing PD (4). In vivo and in vitro 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) models of PD have shown that key enzymes involved in the production of reactive oxygen species (ROS)/reactive nitrogen species (RNS), such as microglial NAPDH oxidase and inducible NO synthase (iNOS), and astroglial myeloperoxidase (MPO), are upregulated in damaged areas and contribute to DA neuronal death (58).

Ethyl pyruvate (EP), a derivative of pyruvate, is an effective scavenger of ROS, especially hydrogen peroxide, by virtue of its nonenzymatic oxidative decarboxylation reaction. Because it is a simple ester, EP might be more stable in aqueous solution than the pyruvate anion (9). EP has been reported to exert pharmacological effects, such as scavenging of ROS, suppression of inflammation, inhibition of apoptosis, and support of cellular ATP synthesis (10). Several lines of evidence have shown that systemic administration of EP decreases infarct volume in the ischemic brain (11) and decreases kainic acid-induced neuronal cell death in the hippocampus (12). EP was also found to protect PC12 cells from dopamine-induced apoptosis (13). However, little is known about the effects of EP related to inflammation in the nigrostriatal DA system in the context of PD. Thus, the current study sought to determine whether EP could prevent the degeneration of nigrostriatal DA neurons by regulating glial activation and reducing oxidative stress in the MPTP model of PD.

Experimental protocol (KHUASP[SE]-10-030) was approved by the Institutional Animal Care and Use Committee of the Kyung Hee University. All experiments conducted with 8- to 10-wk-old male C57BL/6 mice (22–24 g; Charles River Breeding Laboratory) in a room maintained at 20–22°C on a 12-h light/dark cycle with food and water available ad libitum. For MPTP intoxication, mice received four i.p. injection of MPTP-HCl (20 mg/kg; Sigma-Aldrich) dissolved in PBS at 2-h intervals in accordance with published guidelines (14). For EP, in a Ringer’s-type Ca2+- and K+-containing balanced salt solution (9), mice received a single injection per day of EP (10, 25, and 50 mg/kg body weight; Sigma-Aldrich) into the peritoneum at 12 h after the last MPTP injection. Some mice were injected with EP alone as a control. For sodium pyruvate (Py) in PBS, mice received a single injection per day of Py (500 mg/kg body weight; Sigma-Aldrich) into the peritoneum at 12 h after the last MPTP injection. Some mice were injected with Py alone or vehicle as a control.

Animals were transcardially perfused with a saline solution containing 0.5% sodium nitrate and heparin (10 U/ml) and then fixed with 4% paraformaldehyde dissolved in 0.1 M phosphate buffer. Brains were dissected from the skull and postfixed overnight in buffered 4% paraformaldehyde at 4°C. And then, the brains were stored in a 30% sucrose solution for 2–3 d at 4°C until they sank and were frozen sectioned on a sliding microtome in 30-μm-thick coronal sections. All sections were collected in six separate series and processed for immunohistochemical staining as described previously (15). The primary Abs included those directed against tyrosine hydroxylase (TH, 1:2000; Pel-freez), macrophage Ag complex-1 (MAC-1, 1:200; Serotec), ionized calcium-binding adaptor molecule 1 (Iba-1, 1:1000; Wako Chemicals), CD68 (clone ED1 CD68 microglial/macrophage marker [ED-1] 1:1000; Serotec), glial fibrillary acidic protein (GFAP, 1:5000; Neuromics), MPO (1:500; Thermo Scientific), 8-hydroxy-2′-deoxyguanosine (8-OHdG, 1:200; JaICA), and nitrotyrosine (1:100; Millipore). Stained cells were viewed and analyzed under a bright-field microscope (Nikon) or viewed with a confocal laser-scanning microscope (Olympus). For Nissl staining, substantia nigra (SN) tissues immunostained with 8-OHdG or nitrotyrosine Ab were mounted on gelatin-coated slide and dried for 1 h at room temperature and stained with 0.5% cresyl violet (Sigma-Aldrich).

At 2 d after the last MPTP injection, animals were sacrificed, and the brains were harvested for fluorojade-C staining (Chemicon). The staining was performed according to the manufacturer’s protocol. Briefly, 30-μm-thick coronal sections were mounted on gelatin-coated slides, dried for 20 min at room temperature, immersed in 1% sodium hydroxide in 80% ethanol, rinsed in 70% ethanol for 2 min and in distilled water for 2 min, and then incubated in 0.06% potassium permanganate solution for 10 min. After the rinse with distilled water, slides were incubated in 0.0001% fluorojade-C solution for 10 min. After the rinse with distilled water, slides were coverslipped with Vectashield medium and viewed using an LSM confocal laser scanning microscope (Carl Zeiss). To assess the extent of neuronal injury in the sections, the number of fluorojade-C–immunopositive (ip) cells per area was counted.

For double-immunofluorescence staining, sections were processed as described recently (15). Briefly, 30-μm-thick coronal sections were mounted on gelatin-coated slides, dried for 20 min at room temperature, and washed twice in PBST (PBS containing 1% Tween 20). Slides were incubated overnight at 4°C in a combination of an anti-GFAP Ab (1:5000; Neuromics), anti–MAC-1 Ab (1:200; Serotec) and anti-MPO Ab (1:500; Thermo Scientific). After washing in PBST, the sections were incubated for 1 h at room temperature with a mixture of FITC-conjugated anti-mouse IgG (Vector Laboratories) and/or Cy3-counjugated anti-rabbit IgG (Millipore). Slides were coverslipped with Vectashield medium and viewed using an LSM confocal laser scanning microscope (Carl Zeiss). To analyze the localization of different Ags in double-stained samples, images were obtained from the same area and merged using interactive software. For Nissl and GAD67 double-immunofluorescence staining, SN tissues were obtained at 7 d after last MPTP injection and 30-μm-thick coronal sections were mounted on gelatin-coated slides, dried for 20 min at room temperature, and washed twice in PBST (PBS containing 1% Tween 20). Slides were incubated overnight at 4°C in a combination of an anti-GAD67 Ab (1:200; Chemicon). After washing in PBST, the sections were incubated for 1 h at room temperature with a Cy3-counjugated anti-mouse IgG (Millipore). After washing in PBS, the sections were incubated for 1 h at room temperature with NeuroTrace staining solution (1:100; Molecular Probes). The sections were washed with PBST and then incubated overnight at 4°C in PBS. Slides were coverslipped with Vectashield medium and viewed using an LSM confocal laser scanning microscope (Carl Zeiss). To analyze the localization of different Ags in double-stained samples, images were merged the same area and obtained by the computed mask of the colocalized pixels.

The unbiased stereological estimation of the total number of the TH-ip neurons, MAC-1–ip cells, Iba-1–ip cells, and ED-1–ip cells in the substantia nigra (SN) was made using the optical fractionator method performed on an Olympus Computer-Assisted Stereological Toolbox system version 2.1.4 (Olympus) as described recently (15). The sections used for counting covered the entire SN from the rostral tip of the pars compacta back to the caudal end of the pars reticulate (anterioposterior, −2.06 to −4.16 mm from bregma) (16). The SN was delineated at a ×1.25 objective and generated counting grid of 150 × 150 μm. An unbiased counting frame of known area (47.87 × 36.19 μm = 1733 μm2) superimposed on the image was placed randomly on the first counting area and systemically moved through all counting areas until the entire delineated area was sampled. Actual counting was performed using a ×100 objective. The estimate of the total number of neurons was calculated according to the Optical Fractionator Equation (17).

As previously described (15), an average of 17 coronal sections of the STR, starting from the rostral anteroposterior (+1.60 mm) to anteroposterior (0.00 mm), according to bregma of the brain atlas (16), were examined at a ×5 magnification using the IMAGE PRO PLUS system (version 4.0; Media Cybernetics) on a computer attached to a light microscope (Zeiss Axioskop) interfaced with a charge-coupled device video camera (Kodak Mega Plus model 1.4 I). To determine the density of the TH-immunoreactive staining in the STR, a square frame of 700 × 700 μm was placed in the dorsal part of the STR. A second square frame of 200 × 200 μm was placed in the region of the corpus callosum to measure background values. To control for variations in background illumination, the average of the background density readings from the corpus callosum was subtracted from the average of density readings of the STR for each section as described recently (15). Then, the average of all sections of each animal was calculated separately before the data were processed statistically.

An accelerating rotarod (five-lane accelerating rotarod; Ugo Basile) was used to measure forelimb and hindlimb motor dysfunctions as described recently (15). The rotarod unit consisted of a rotating spindle (diameter, 3 cm) where mice were challenged for speed. For training, mice were placed on the rotarod at 10 rpm for 20 min, 7 consecutive days before MPTP treatment. Mice that stayed on the rod without falling during training were selected and randomly divided into experimental groups. Seven days after the last MPTP treatment, animals receiving various treatment regimes were placed in a separate compartment on the rotating rod and tested at 20 rpm for 20 min. The latency to fall was automatically recorded by magnetic trip plates.

To measure contents of dopamine in STR, we performed the reversed-phase HPLC with electrochemical detector as described recently (15). Dissected striatal tissues were homogenized with 0.1 M perchloric acid and 0.1 mM EDTA buffer and centrifuged at 9000 rpm for 20 min. The supernatant was injected into an autosampler at 4°C (Waters 717 plus autosampler) and eluted through μBondapak C18 column (3.9 × 300 mm × 10 μm, ESA) with mobile phase for catecholamine analyses (Chromosystems). The peaks of dopamine content were analyzed by ESA Coulochem II electrochemical detector and integrated using a commercially available program (Breeze). All samples were normalized for protein content as spectrophotometrically determined using the Bio-Rad protein assay kit (Bio-Rad).

The extracted striatal tissues were frozen and homogenized with a tissue crusher and sonicated for 10 min in an ice bath. After sonication, the samples were centrifuged at 13,000 × g for 15 min. The upper layers were taken for solid-phase extraction cleanup. The process of solid-phase extraction was performed using mixed-mode strong cation-exchange (1 ml, 30 mg) cartridges. Norepinephrine was eluted with 2 ml 5% NH4OH, and the extracts were dried under the gentle stream of nitrogen gas and reconstituted with 70 μl ethyl acetate. Prior to injection, extracts were derivatived. The sample analysis was carried out with an Agilent GC/MS (Palo Alto, CA) equipped with DB-5MS fused-silica capillary column (30 × 0.25 mm i.d., film thickness 0.25 μm; J&W Scientific, Folsom, CA). The oven temperature was held at 150°C for 2 min, elevated to 195°C at 10°C/min and to 210°C at 5°C/min, increased to 250°C at 20°C/min and elevated from 250 to 300°C at 40°C/min, and held for 3 min. The mass spectra of derivatives were obtained in the mass range from 100 to 600 amu. For the monitoring and confirmation analysis, the selected ion monitoring mode was used, and the dwell time of each ion was set at 50 ms.

Hydroethidine histochemistry was performed for in situ visualization of O2 and O2-derived oxidants (15). Three days after the last MPTP injection, hydroethidine (1 mg/ml in PBS containing 1% DMSO; Molecular Probes) was administered i.p. After 15 min, the animals were transcardially perfused with a saline solution containing 0.5% sodium nitrate and heparin (10 U/ml) and then fixed with 4% paraformaldehyde in 0.1 M phosphate buffer. After fixation, the brains were cut into 30-μm slices using a sliding microtome. Sections were mounted on gelatin-coated slides, and the oxidized hydroethidine product ethidium was examined by IX71 confocal microscop (Olympus Optical). To quantify, obtained images were analyzed by the Computer Imaging Device and accompanying software (Fujifilm).

For immunoprecipitation, we dissected SN tissues from the animals at 2 d after injection of MPTP. As recently described (15), SN samples were homogenized with using a glass homogenizer in ice-cold buffer consisting of the following: 50 mM Tris (pH 8), 150 mM NaCl, 2 mM EDTA, 1% Nonidet P-40, 0.1% SDS, 0.5% deoxycholate, and protease inhibitor mixture (Roche). Homogenates were centrifuged for 5 min at 500 × g at 4°C, and supernatants were collected and centrifuged for 20 min at 13,000 × g at 4°C. Equal amounts of proteins (240 μg) were immunoprecipitated with anti-p47phox Ab (2.4 μg; Santa Cruz Biotechnology) or anti-TH Ab (2.4 μg; Millipore). The immunoprecipitated proteins were separated on a 12% SDS-PAGE gel and were then electroblotted onto polyvinylidene difluoride membrane and blocked for 1 h at room temperature with TBS containing 5% nonfat dried milk. The membranes were immunoblotted with anti-phosphoserine Ab (1:1000; Invitrogen Life Technologies), anti-p47phox Ab (1:1000; Santa Cruz Biotechnology), anti-gp91phox Ab (1:1000; BD Biosciences), anti-nitrotyrosine Ab (1:1000), and anti-TH Ab (1:2000). For semiquantitative analyses, the densities of whole bands on immunoblots were measured with the Computer Imaging Device and accompanying software (Fujifilm).

We dissected SN tissues from the animals at 3 d after injection of MPTP. As previously described (18, 19), SN samples were homogenized with using a glass homogenizer in ice-cold buffer consisting of the following: 20 mM HEPES, 250 mM sucrose, 10 mM KCl, 1.5 mM MgCl2, 2 mM EDTA, and protease inhibitor mixture (Roche). Homogenates were centrifuged for 5 min at 500 × g at 4°C, and supernatants were collected and centrifuged for 20 min at 13,000 × g at 4°C. Equal amounts of proteins (30 μg) were separated by 10% SDS-PAGE gels, transferred to polyvinylidene difluoride membrane (Millipore) using an electrophoretic transfer system (Bio-Rad). The membranes were immunoblotted with anti–β-tubulin Ab (1:1000; Santa Cruz Biotechnology), anti-nitrotyrosine Ab (1:1000; Millipore), anti-MPO Ab (1:000; Thermo Scientific), and anti-iNOS Ab (1:500; BD Biosciences). For semiquantitative analyses, the densities of bands on immunoblots were measured with the Computer Imaging Device and accompanying software (Fujifilm).

Neuron-enriched mesencephalic cultures were prepared as described previously (20, 21). In brief, dissociated cells from the ventral mesencephalons of embryonic day (E) 14 Sprague–Dawley rats were seeded on 12-mm round aclar plastic coverslips (1.2 × 105 cells/coverslip) housed in 24-well culture plates. To suppress the proliferation of glial cells, at 2 d in vitro (DIV), the medium was replaced with a chemically defined serum-free medium. At DIV 4, cultures were pretreated with EP for 30 min and then treated with vehicle or 20 μM 1-methyl-4-phenyl-pyridinium (MPP+) in RF medium ([DMEM; Life Technologies, Rockville, MD] supplemented with 6 mg/ml gllucose, 204 μg/ml l-glutamine, 100 U/ml penicillin/streptomycin, and 2% FBS [Hyclone]) for 48 h. These cultures were processed for immunocytochemistry. When cultures were immunostained with cell-type specific Abs (see above immunohistochemistry), the cell composition included ∼5% astrocytes and less than ∼1% microglia (21). The remaining cells were presumed to be neurons, 1–1.2 and 9.5–11% of which were TH-ip and γ amino butyric acid-ip neurons, respectively (22). For methyl green staining, cells were stained for 5 min at room temperature with 0.5% methyl green solution (Sigma-Aldrich) after immunostained with TH Ab.

Mesencephalic microglia cultures were prepared from the ventral mesencephalons of E14 Sprague–Dawley rat brains as described previously (23). Tissues were triturated and plated in 75-cm2 T flasks precoated with poly-d-lysine at a density of 1 × 107 cells/flask and then maintained in DMEM supplemented with 10% FBS. After 2–3 wk, microglia were detached from the flasks, applied to a nylon mesh to remove astrocytes, and then plated into 24-well plates at a density of 5 × 104 cells/well. At 1 h after plating, unattached cells were removed, and at 24 h after plating, the cells were pretreated with EP for 30 min and then treated with MPP+ or vehicle.

At DIV 3, neuron-enriched mesencephalic cultures plated on 12-mm round aclar plastic coverslips (1.2 × 105 cells/coverslip) housed in 24-well plates were supplemented with 5 × 104 mesencephalic microglia/well. After 1 h, the culture medium was replaced with neuronal culture medium (DMEM containing 2% FBS); 24 h later, the cocultures were pretreated with, EP for 30 min at indicated doses and then treated with MPP+ or vehicle for 48 h and were processed for measurement of dopamine uptake or for immunocytochemistry.

Measurement of dopamine uptake was performed as described previously (24). Briefly, cultures were washed twice with an incubation solution (HBSS containing 10 mM HEPES, 0.6% glucose, 0.2 mM pargyline, and 0.01% ascorbic acid), tritiated dopamine ([3H]DA; 444 GBq/mmol) was added to a final concentration of 83.3 nM, and the cultures were incubated at 37°C for 20 min. Blanks were obtained by incubating cultures at 0°C. The reaction was stopped by rinsing the cultures with ice-cold incubation solution. The cultures were then lysed with 0.2 M NaOH containing 0.2% Triton X-100 and transferred to scintillation vials for counting.

Intracellular ATP contents were measured using luciferin–luciferase reaction with CellTiter-Glo Luminescent Cell ViabilityAssay kit (Promega) according to the manufacturer’s instructions. Briefly, 20 μl cell media was mixed with 20 μl CellTiter-Glo Luminescent Cell ViabilityCellTiter-Glo Reagent and incubated at 20°C for 10 min. We measured the luminescence signal with an LB 9501 Lumatluminometer (Berthold, German). We subtracted a background luminescence value in control wells containing medium without cells from the signal. The amounts of ATP contents were normalized to protein concentration. The ROS generation was measured using 5,6-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate, acetyl ester (Molecular Probes). The cells in phenol red-free media without serumin black 96-well culture plate were incubated with 1 μM 5,6-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate, acetyl ester at 37 °C for 1 h and then incubated with 0.5 μM Hoechst 33342 at room temperature for 10 min. The fluorescence intensities at 485/535 nm for diacetate, acetyl ester were normalized by Hoechst intensity at 355/460 nm. All data were presented as percent of control after calculation.

All values are expressed as mean ± SEM. Statistical significance (p < 0.05 for all analyses) was assessed by ANOVA using Instat 3.05 (GraphPad, San Diego, CA), followed by Student–Newman–Keuls analyses.

The mice in each group received four i.p. injections of MPTP (20 mg/kg) or PBS (control) at 2-h intervals. Seven days later, brains were removed, and sections were immunostained for TH to specifically detect DA neurons. Consistent with our recent report (15), there was a significant loss of TH-ip cell bodies in the SN (Fig. 1B) and fibers in the STR (Fig. 2B) at 7 d in MPTP-injected mice compared with those in PBS-treated control mice (Figs. 1A, 2A). TH-ip cells in the SN and their nerve terminals in the STR were quantified by stereological counts and densitometric analyses, respectively. MPTP treatment decreased the number of TH-ip neurons by 69% in the SN (Fig. 1E) and reduced the OD of TH-ip fibers by 68% in the STR (Fig. 2E) compared with those of PBS-treated mice.

FIGURE 1.

EP and Py prevent MPTP-induced neurotoxicity in the SN of the mouse brain. Animals receiving PBS as a control (A), MPTP (B), MPTP and EP (50 mg/kg) (C), or MPTP and Py (500 mg/kg) (D) were sacrificed 7 d after the last MPTP injection. Insets, Four-fold higher magnifications of A–D. The brain tissues were cut into 30-μm-thick coronal sections using a sliding microtome and immunostained with an Ab against the DA neuronal marker TH. Scale bar, 200 μm. E, Bars represent the numbers of TH-ip neurons in the SN after treatment with the indicated dose of EP (10–50 mg/kg) or Py (500 mg/kg). Four to nine animals were used for each experimental group. #p < 0.001, compared with PBS, EP, or Py alone; *p < 0.01, **p < 0.001, $p < 0.001, compared with MPTP only.

FIGURE 1.

EP and Py prevent MPTP-induced neurotoxicity in the SN of the mouse brain. Animals receiving PBS as a control (A), MPTP (B), MPTP and EP (50 mg/kg) (C), or MPTP and Py (500 mg/kg) (D) were sacrificed 7 d after the last MPTP injection. Insets, Four-fold higher magnifications of A–D. The brain tissues were cut into 30-μm-thick coronal sections using a sliding microtome and immunostained with an Ab against the DA neuronal marker TH. Scale bar, 200 μm. E, Bars represent the numbers of TH-ip neurons in the SN after treatment with the indicated dose of EP (10–50 mg/kg) or Py (500 mg/kg). Four to nine animals were used for each experimental group. #p < 0.001, compared with PBS, EP, or Py alone; *p < 0.01, **p < 0.001, $p < 0.001, compared with MPTP only.

Close modal
FIGURE 2.

EP and Py prevent MPTP-induced neurotoxicity in the STR of the mouse brain. Striatal tissues obtained from the same animals used in Fig. 1 were immunostained with an anti-TH Ab for DA fibers. Animals were treated with PBS as a control (A), MPTP (B), MPTP and EP (50 mg/kg) (C), or MPTP and Py (500 mg/kg) (D). E, Bars represent the OD of TH-ip fibers in the STR at the indicated dose of EP (10–50 mg/kg) or Py (500 mg/kg) 7 d after the last MPTP administration. Four to nine animals were used for each experimental group. #p < 0.001, compared with PBS, EP, or Py alone; *p < 0.01, **p < 0.001, $p < 0.001, compared with MPTP only. Scale bar, 1 mm. F, Effects of EP on motor behavior and striatal dopamine levels. After the rotarod test, striatal tissues were obtained for determination of dopamine levels. Four to six animals were used for each experimental group. #p < 0.001, compared with PBS or EP alone; *p < 0.01, compared with MPTP only. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg).

FIGURE 2.

EP and Py prevent MPTP-induced neurotoxicity in the STR of the mouse brain. Striatal tissues obtained from the same animals used in Fig. 1 were immunostained with an anti-TH Ab for DA fibers. Animals were treated with PBS as a control (A), MPTP (B), MPTP and EP (50 mg/kg) (C), or MPTP and Py (500 mg/kg) (D). E, Bars represent the OD of TH-ip fibers in the STR at the indicated dose of EP (10–50 mg/kg) or Py (500 mg/kg) 7 d after the last MPTP administration. Four to nine animals were used for each experimental group. #p < 0.001, compared with PBS, EP, or Py alone; *p < 0.01, **p < 0.001, $p < 0.001, compared with MPTP only. Scale bar, 1 mm. F, Effects of EP on motor behavior and striatal dopamine levels. After the rotarod test, striatal tissues were obtained for determination of dopamine levels. Four to six animals were used for each experimental group. #p < 0.001, compared with PBS or EP alone; *p < 0.01, compared with MPTP only. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg).

Close modal

To investigate whether EP altered MPTP-induced neurotoxicity of nigrostriatal DA neurons, we administered EP (10–50 mg/kg body weight) 12 h after the last MPTP or PBS injection. We chose a 12-h time point because MPTP is completely converted into MPP+ and sufficiently accumulates in DA neurons prior to 12 h posttreatment (15); thus, its effects could not be attributed to the diminished metabolism of MPTP to MPP+ or uptake of MPP+ into DA neurons. The results of TH immunohistochemistry showed that treatment with 50 mg/kg EP effectively reduced MPTP-induced loss of DA neurons in the SN (Fig. 1C) and their nerve terminals in the STR (Fig. 2C). Quantification of TH immunostaining showed that 25 and 50 mg/kg EP increased the number of TH-ip neurons in the SN by 13% (p < 0.01; Fig. 1E) and 25% (p < 0.001; Fig. 1E), respectively, and produced a corresponding increase in the OD of TH-ip fibers in the STR of 23% (p < 0.01; Fig. 2E) and 35% (p < 0.001; Fig. 2E) compared with controls. Treatment with 10 mg/kg EP did not increase the number of TH-ip neurons. EP alone had no effect on the number of neurons in the SN (Fig. 1E) or DA fibers in the STR (Fig. 2E).

To investigate whether Py altered MPTP-induced neurotoxicity of nigrostriatal DA neurons, we administered 500 mg/kg Py 12 h after the last MPTP or PBS injection. Quantification of TH immunostaining showed that 500 mg/kg Py increased the number of TH-ip neurons in the SN by 32% (p < 0.001; Fig. 1D, 1E) and produced a corresponding increase in the OD of TH-ip fibers in the STR of 49% (p < 0.001; Fig. 2D, 2E) compared with controls.

To verify neuroprotective effect of EP on GABA neurons, SN tissues were double immunostaining with Nissl and GAD67 Ab. MPTP did not induce loss of Nissl+/GABA neurons. In addition, EP had no effect on the number of Nissl+/GABA neurons with or without MPTP (Supplemental Fig. 2J–N).

We next determined whether EP improves MPTP-induced motor deficits by testing performance on a rotarod apparatus, as described recently (15). Animals receiving various treatment regimens were evaluated 7 d after the last MPTP injection by measuring the latency to falling. MPTP treatment decreased sustained rotarod time to 11.04 ± 0.4 min, a 61% decrease compared with that of PBS treatment (p < 0.01; Fig. 2F). The behavioral dysfunction in MPTP-treated mice was recovered by EP treatment, which increased the latency to falling to 14.59 ± 0.83 min (p < 0.05; Fig. 2F). EP alone had no effect on motor behavior.

After rotarod performance tests, the mice were sacrificed for biochemical assessments. In parallel with behavioral dysfunction, HPLC analyses revealed that MPTP reduced striatal dopamine levels to 71% of those in PBS controls (Fig. 2F). By contrast, treatment of MPTP-injected mice with EP increased dopamine levels by 25% compared with MPTP only (p < 0.05; Fig. 2F). EP alone had no effect on striatal dopamine levels.

The loss of norepinephrine may aggravate the MPTP-mediated damage of the nigrostriatal DA system (25, 26). gas chromatography–mass spectrometry analysis showed that norepinephrine levels were unchanged in the STR with or without MPTP (Supplemental Fig. 1H). This observation is comparable to a recent report that striatal norepinephrine level was decreased by acute MPTP treatment (40 mg/kg per dose, twice), and these effects of acute MPTP on norepinephrine may be transient (26).

Accumulating evidence suggests that activated microglia play an important role in DA neuronal cell death in the MPTP model (27). Thus, we next examined whether the neuroprotective effect of EP resulted from inhibition of microglial activation in the SN. At 3 d after the last MPTP injection, brain tissues from mice treated with or without EP were processed for immunostaining with Abs against the markers for activated microglia, MAC-1, Iba-1, and ED-1. In contrast to the SN of control mice treated with PBS (Fig. 3A) or EP (Fig. 3B) alone, where relatively few positive microglia were observed, the SN of MPTP-treated mice contained numerous robustly immunoreactive MAC-1–positive (activated) microglia (Fig. 3C). EP treatment dramatically reduced the number of MAC-1–ip microglia in the MPTP-treated SN (Fig. 3D, 3M), indicating that EP suppressed MPTP-induced microglial activation.

FIGURE 3.

EP blocks microglial activation in the SN in vivo. Animals receiving PBS as a control (A, E, I), EP (50 mg/kg) alone (B, F, J), MPTP (C, G, K), or MPTP and EP (50 mg/kg) (D, H, L) were sacrificed 3 d after the last MPTP administration. Mouse SN tissues were immunostained with Abs against MAC-1, Iba-1, and ED-1 to identify microglia. Insets, Four-fold higher magnifications of A–L. Dotted lines indicate the SNpc. Scale bar, 100 μm. M, Bars represent the numbers of MAC-1-ip, Iba-1-ip, or ED-1-ip cells in the SN, respectively. Five animals were used for each experimental group. #p < 0.001, compared with PBS or EP alone; *p < 0.001, compared with MPTP only. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg).

FIGURE 3.

EP blocks microglial activation in the SN in vivo. Animals receiving PBS as a control (A, E, I), EP (50 mg/kg) alone (B, F, J), MPTP (C, G, K), or MPTP and EP (50 mg/kg) (D, H, L) were sacrificed 3 d after the last MPTP administration. Mouse SN tissues were immunostained with Abs against MAC-1, Iba-1, and ED-1 to identify microglia. Insets, Four-fold higher magnifications of A–L. Dotted lines indicate the SNpc. Scale bar, 100 μm. M, Bars represent the numbers of MAC-1-ip, Iba-1-ip, or ED-1-ip cells in the SN, respectively. Five animals were used for each experimental group. #p < 0.001, compared with PBS or EP alone; *p < 0.001, compared with MPTP only. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg).

Close modal

Consistent with MAC-1 staining result, Iba-1–positive microglia exhibited the typical ramified morphology of resting microglia in the PBS-treated (Fig. 3E) and EP-treated (Fig. 3F) control SN. In contrast, the majority of Iba-1–ip microglia in the MPTP-treated SN displayed an activated morphology, including larger cell bodies with short and thick process (Fig. 3G). Treatment with EP mitigated these effects of MPTP, dramatically decreasing the number of activated microglia in the MPTP-treated SN (Fig. 3H, 3M).

Microglia in the MPTP-treated SN appeared to reach a state similar to that of active phagocytes (Fig. 3K), as determined by ED-1 immunostaining, which labels phagocytes (28, 29). The majority of ED-1–ip microglia in the MPTP-treated SN displayed an activated morphology, including larger cell bodies with short and thick process or no processes (Fig. 3K). Treatment with EP mitigated these effects of MPTP, dramatically decreasing the number of activated microglia in the MPTP-treated SN (Fig. 3L, 3M). The numbers of ED-1–ip cells were not detectable in the SN of mice treated with PBS (Fig. 3I) or EP (Fig. 3J) only.

Few such ED-1–ip cells were observed in the MPTP and EP-treated SN (Fig. 3L), similar to the results of MAC-1 immunostaining. ED-1–ip cells were not detectable in the SN of mice treated with PBS (Fig. 3I) or EP (Fig. 3J) only.

The production of ROS, such as H2O2 and O2, is increased in the midbrain of MPTP-treated mice, and oxidants originating from microglia are thought to mediate the loss of DA neurons in the SN (30). Thus, we examined whether EP rescued nigral DA neurons by inhibiting MPTP-induced oxidant production. The fluorescent product of oxidized hydroethidine (i.e., ethidium) was significantly increased in the MPTP-injected SN 3 d after administration (Fig. 4C, 4E) compared with that of the PBS-injected SN (Fig. 4A, 4E). EP dramatically decreased MPTP-induced oxidant production in the SN in vivo (Fig. 4D, 4E) but had no effect alone (Fig. 4B, 4E).

FIGURE 4.

EP inhibits MPTP-induced oxidant production, activation of NADPH oxidase, and oxidative DNA damage in the SN in vivo. Animals receiving PBS as a control (A), EP (50 mg/kg) alone (B), MPTP (C), or MPTP and EP (50 mg/kg) (D) were sacrificed 3 d after the last MPTP injection. A–D, SN tissues were prepared for hydroethidine histochemistry to detect oxidant production. Five to seven animals were used for each experimental group. Scale bar (A–D), 20 μm. E, The graph represents OD of hydroethidine staining analyzed by the Computer Imaging Device and accompanying software (Fujifilm). #p < 0.001, compared with PBS or EP only; *p < 0.001, compared with MPTP only. F, Western blot analyses show phosphorylation of cytosolic subunits p47phox and p47phox–gp9phox binding 2 d after MPTP injection, indicating activation of NADPH oxidase in the SN. Animals were sacrificed 2 d after the last MPTP administration, and SN tissues were isolated. Tissue lysates were immunoprecipitated with an anti-p47phox Ab and analyzed by immunoblotting with Abs against p47phox, gp91phox, and phospho-serine. Total p47phox level indicates protein-loading control. G, The graph represents phospho-serine and gp91phox levels expressed relative to total p47phox. The results represent the mean ± SEM for four or five mice per group from three independent experiments. #p < 0.01, compared with PBS or EP only; *p < 0.01, compared with MPTP only. Animals receiving PBS as a control (H), EP (50 mg/kg) alone (I), MPTP (J), or MPTP and EP (50 mg/kg) (K) were sacrificed 3 d after the last MPTP injection. H–K, SN tissues obtained from the same animals used in A–D were stained for 8-OHdG to detect oxidative DNA damage and counterstained with Nissl. Insets, Four-fold higher magnifications of H–K. Dotted lines indicate the SNpc. Five animals were used for each experimental group. Scale bar (G–J), 100 μm. CON, control; EP, 50 mg/kg EP; IP, immunoprecipitation; M, MPTP; MEP, MPTP, and EP (50 mg/kg).

FIGURE 4.

EP inhibits MPTP-induced oxidant production, activation of NADPH oxidase, and oxidative DNA damage in the SN in vivo. Animals receiving PBS as a control (A), EP (50 mg/kg) alone (B), MPTP (C), or MPTP and EP (50 mg/kg) (D) were sacrificed 3 d after the last MPTP injection. A–D, SN tissues were prepared for hydroethidine histochemistry to detect oxidant production. Five to seven animals were used for each experimental group. Scale bar (A–D), 20 μm. E, The graph represents OD of hydroethidine staining analyzed by the Computer Imaging Device and accompanying software (Fujifilm). #p < 0.001, compared with PBS or EP only; *p < 0.001, compared with MPTP only. F, Western blot analyses show phosphorylation of cytosolic subunits p47phox and p47phox–gp9phox binding 2 d after MPTP injection, indicating activation of NADPH oxidase in the SN. Animals were sacrificed 2 d after the last MPTP administration, and SN tissues were isolated. Tissue lysates were immunoprecipitated with an anti-p47phox Ab and analyzed by immunoblotting with Abs against p47phox, gp91phox, and phospho-serine. Total p47phox level indicates protein-loading control. G, The graph represents phospho-serine and gp91phox levels expressed relative to total p47phox. The results represent the mean ± SEM for four or five mice per group from three independent experiments. #p < 0.01, compared with PBS or EP only; *p < 0.01, compared with MPTP only. Animals receiving PBS as a control (H), EP (50 mg/kg) alone (I), MPTP (J), or MPTP and EP (50 mg/kg) (K) were sacrificed 3 d after the last MPTP injection. H–K, SN tissues obtained from the same animals used in A–D were stained for 8-OHdG to detect oxidative DNA damage and counterstained with Nissl. Insets, Four-fold higher magnifications of H–K. Dotted lines indicate the SNpc. Five animals were used for each experimental group. Scale bar (G–J), 100 μm. CON, control; EP, 50 mg/kg EP; IP, immunoprecipitation; M, MPTP; MEP, MPTP, and EP (50 mg/kg).

Close modal

We then examined the assembly and activation of NADPH oxidase as a source of ROS production and resultant oxidative damage in the SN 2 d after the last MPTP injection by immunoprecipitating with an anti-p47phox Ab and then immunoblotting with anti-phosphoserine, anti-gp91phox and anti-p47phox Abs. In MPTP-treated SN tissues, p47phox phosphorylation and p47phox–gp91phox complexes were significantly increased compared with PBS-treated SN samples (p < 0.01; Fig. 4F, 4G), indicating activation of NADPH oxidase. By contrast, treatment with EP after the MPTP injection significantly decreased phosphorylation of p47phox and the level of p47phox–gp91phox complexes in the SN compared with MPTP alone (p < 0.01; Fig. 4F, 4G). EP alone had no effect on the level of phospho-p47phox or p47phox–gp91phox complex formation (Fig. 4F, 4G). These results suggest that MPTP-induced activation of NADPH oxidase is inhibited by EP. As shown in our recent report (15), p47phox immunoreactivity was detected exclusively in activate microglia but not astrocytes and neurons (data not shown).

We have recently shown that, in MPTP mice, DA neuronal cell death is accompanied by increased levels of 8-OHdG, a marker of oxidative nucleic acid damage (15). To determine whether EP prevents MPTP-induced oxidative damage to DNA in the SN, we immunostained for 8-OHdG. The present results showed that the levels of 8-OHdG were dramatically increased in the SN 3 d after MPTP injection (Fig. 4J) compared with those in the PBS-treated SN (Fig. 4H). This MPTP-induced oxidative DNA damage was dramatically inhibited by EP (Fig. 4K). EP alone had no effect on DNA damage (Fig. 4I).

It has been shown that transgenic mice deficient in iNOS are resistant to MPTP-induced neurotoxicity (5). Thus, we examined whether EP might modulate DA neuronal survival by affecting MPTP-induced expression of iNOS in the SN. Two days after the final MPTP injection, SN tissues were dissected and prepared for Western blot analyses. The results showed that EP attenuated MPTP-induced expression of iNOS protein in the SN (p < 0.0.1; Fig. 5A, 5B).

FIGURE 5.

EP decreases MPTP-induced NO damage in the SN in vivo. A, Animals were sacrificed 2 d after the last MPTP administration and SN tissues were isolated. Tissue lysates were analyzed by Western blotting using Abs against iNOS, nitro-Tyr, and TH. An anti–β-tubulin Ab was used as a loading control. B, The histogram represents iNOS, nitro-Tyr, and TH levels expressed relative to β-tubulin. The results represent the mean ± SEM of three separate experiments. Four to six animals were used for each experimental group. #p < 0.05, compared with PBS or EP only; *p < 0.05, compared with MPTP only. C–F, SN tissues obtained from the same animals used in Fig. 4A–D were stained for nitro-Tyr to detect oxidative damage to proteins involving modification of tyrosine residues and then were counterstained with Nissl. G, Western blot analyses show nitration of TH after EP treatment with or without MPTP. Animals were sacrificed 2 d after the last MPTP administration, and SN tissues were isolated. Tissue lysates were immunoprecipitated with an anti-TH Ab and analyzed by immunoblotting with Abs against nitro-Tyr and TH. TH level indicates protein-loading control. Animals received PBS as a control (C), EP (50 mg/kg) alone (D), MPTP (E), or MPTP and EP (F). Dotted lines indicate the SNpc. Insets, Four-fold higher magnifications of C–F. Scale bar, 100 μm. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg); Nitro-Tyr, nitro-tyrosine.

FIGURE 5.

EP decreases MPTP-induced NO damage in the SN in vivo. A, Animals were sacrificed 2 d after the last MPTP administration and SN tissues were isolated. Tissue lysates were analyzed by Western blotting using Abs against iNOS, nitro-Tyr, and TH. An anti–β-tubulin Ab was used as a loading control. B, The histogram represents iNOS, nitro-Tyr, and TH levels expressed relative to β-tubulin. The results represent the mean ± SEM of three separate experiments. Four to six animals were used for each experimental group. #p < 0.05, compared with PBS or EP only; *p < 0.05, compared with MPTP only. C–F, SN tissues obtained from the same animals used in Fig. 4A–D were stained for nitro-Tyr to detect oxidative damage to proteins involving modification of tyrosine residues and then were counterstained with Nissl. G, Western blot analyses show nitration of TH after EP treatment with or without MPTP. Animals were sacrificed 2 d after the last MPTP administration, and SN tissues were isolated. Tissue lysates were immunoprecipitated with an anti-TH Ab and analyzed by immunoblotting with Abs against nitro-Tyr and TH. TH level indicates protein-loading control. Animals received PBS as a control (C), EP (50 mg/kg) alone (D), MPTP (E), or MPTP and EP (F). Dotted lines indicate the SNpc. Insets, Four-fold higher magnifications of C–F. Scale bar, 100 μm. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg); Nitro-Tyr, nitro-tyrosine.

Close modal

Nitration of protein tyrosine residues, a well-known marker of oxidative stress in PD patients (31), is mediated by iNOS-derived oxidants (32). To measure the extent of iNOS-mediated oxidative damage to proteins, we immunostained the SN for nitrotyrosine 3 d after MPTP injection, with or without EP treatment. We found that the levels of nitrotyrosine were dramatically increased in the MPTP-injected SN (Fig. 5E) compared with the PBS-treated SN (Fig. 5C). This MPTP-induced nitration of protein was dramatically inhibited by EP (Fig. 5F), which had no effect alone (Fig. 5D). Consistent with this immunocytochemical staining, Western blot analyses also demonstrated that the levels of nitrotyrosine were significantly increased in the MPTP-treated SN compared with those in the PBS-treated SN (p < 0.01; Fig. 5A, 5B). EP significantly reduced the levels of MPTP-induced nitrotyrosine and increased expression of TH proteins in the MPTP-treated SN (p < 0.01; Fig. 5A, 5B). TH is a target for MPTP-induced nitrotyrosine, leading to loss of TH (33, 34). Additional Western blot analysis revealed that EP significantly attenuated MPTP-induced nitrotyrosine of TH (Supplemental Fig. 1G), indicating that EP protects TH from nitrosative damage.

Because EP was administered 12 h after the final injection of MPTP in all in vivo experiments, its effects could not be attributed to reduced metabolism of MPTP to MPP+ or MPP+ uptake into DA neurons (15). However, the possibility remained that EP might promote neuronal survival by preventing MPP+-induced blockade of mitochondrial respiration in neurons. To test this hypothesis, we performed additional experiments with mesencephalic neurons cultured alone or cocultured with microglia. In microglia-free, neuron-enriched mesencephalic cultures, pretreatment with 0.5–1 mM EP (30 min before MPP+ treatment) had no protective effects (Fig. 6A–D, 6I), and 5 mM EP alone was neurotoxic (data not shown). By contrast, in cocultures of mesencephalic neurons and microglia, 1 mM EP blocked MPP+-induced death of DA neurons (p < 0.05; Fig. 6E–H, 6I).

FIGURE 6.

EP protects against neuronal cell death induced by MPP+ in cocultures of mesencephalic neurons and microglia. A and E, Phase-contrast images of cells in cultures of mesencephalic neurons (A) and cocultures of neurons and microglia (E) prior to MPP+ treatment. Arrowheads indicate microglia. B–D, TH-ip neurons in neuron-enriched mesencephalic cultures treated with PBS as a control (B) or MPP+ (20 μM) for 48 h in the absence (C) or presence (D) of EP (1 mM) pretreatment for 30 min. F–H, TH-ip neurons in cocultures of mesencephalic neurons and microglia treated with PBS as a control (F) or MPP+ (20 μM) for 48 h in the absence (G) or presence (H) of EP (1 mM) pretreatment for 30 min. Methyl green was used as a counterstaining. I, Numbers of TH-ip neurons were counted in neuron-enriched cultures and cocultures. $p < 0.001, compared with PBS or EP alone in neuron-enriched cultures; #p < 0.001, compared with PBS or EP alone in cocultures; *p < 0.05, compared with MPP+ only in cocultures. The results for [3H] dopamine uptake are expressed as a percentage of that in PBS-treated control cocultures. Scale bar (A, E), 20 μm; scale bar (BD, FH), 10 μm. Results are the mean ± SEM of triplicate cultures from three separate plates. CON, control; EP, 1 mM EP; M, MPP+; MEP, MPP+ and EP (1 mM).

FIGURE 6.

EP protects against neuronal cell death induced by MPP+ in cocultures of mesencephalic neurons and microglia. A and E, Phase-contrast images of cells in cultures of mesencephalic neurons (A) and cocultures of neurons and microglia (E) prior to MPP+ treatment. Arrowheads indicate microglia. B–D, TH-ip neurons in neuron-enriched mesencephalic cultures treated with PBS as a control (B) or MPP+ (20 μM) for 48 h in the absence (C) or presence (D) of EP (1 mM) pretreatment for 30 min. F–H, TH-ip neurons in cocultures of mesencephalic neurons and microglia treated with PBS as a control (F) or MPP+ (20 μM) for 48 h in the absence (G) or presence (H) of EP (1 mM) pretreatment for 30 min. Methyl green was used as a counterstaining. I, Numbers of TH-ip neurons were counted in neuron-enriched cultures and cocultures. $p < 0.001, compared with PBS or EP alone in neuron-enriched cultures; #p < 0.001, compared with PBS or EP alone in cocultures; *p < 0.05, compared with MPP+ only in cocultures. The results for [3H] dopamine uptake are expressed as a percentage of that in PBS-treated control cocultures. Scale bar (A, E), 20 μm; scale bar (BD, FH), 10 μm. Results are the mean ± SEM of triplicate cultures from three separate plates. CON, control; EP, 1 mM EP; M, MPP+; MEP, MPP+ and EP (1 mM).

Close modal

GFAP-immunostained tissues adjacent to those used for MAC-1 immunostaining confirmed that resting astrocytes (small somas with thin processes; Fig. 7A, 7B) were transformed into active astrocytes (enlarged cell bodies with short or thick processes; Fig. 7C, 7D) in MPTP-treated mice. Interestingly, treatment with EP (50 mg/kg) after MPTP injection sustained the activated state of GFAP–ip astrocytes (Fig. 7E, 7F), but alone had no effect (data not shown).

FIGURE 7.

Effect of EP on MPTP-activated astrocytes in the SN in vivo. A–F, Animals receiving PBS as a control (A, B), MPTP (C, D), or MPTP and EP (50 mg/kg, i.p.) (E, F) were sacrificed 3 d after the last MPTP injection. SN tissues were immunostained with an anti-GFAP Ab to identify astrocytes. Dotted lines indicate the SNpc. Scale bar, 50 μm. G, Animals were sacrificed 3 d after the last MPTP administration, and SN tissues were isolated. Tissue lysates were analyzed by Western blotting with Abs against MPO and β-tubulin (used as a loading control). H, The histogram represents MPO protein levels expressed relative to β-tubulin levels and the number of double-ip (MPO+/GFAP+) cells in the SNpc. The results represent the mean ± SEM of three separate experiments. Three to six animals were used for each experimental group: #p < 0.001, compared with PBS or EP alone; *p < 0.001, compared with MPTP only. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg).

FIGURE 7.

Effect of EP on MPTP-activated astrocytes in the SN in vivo. A–F, Animals receiving PBS as a control (A, B), MPTP (C, D), or MPTP and EP (50 mg/kg, i.p.) (E, F) were sacrificed 3 d after the last MPTP injection. SN tissues were immunostained with an anti-GFAP Ab to identify astrocytes. Dotted lines indicate the SNpc. Scale bar, 50 μm. G, Animals were sacrificed 3 d after the last MPTP administration, and SN tissues were isolated. Tissue lysates were analyzed by Western blotting with Abs against MPO and β-tubulin (used as a loading control). H, The histogram represents MPO protein levels expressed relative to β-tubulin levels and the number of double-ip (MPO+/GFAP+) cells in the SNpc. The results represent the mean ± SEM of three separate experiments. Three to six animals were used for each experimental group: #p < 0.001, compared with PBS or EP alone; *p < 0.001, compared with MPTP only. CON, control; EP, 50 mg/kg EP; M, MPTP; MEP, MPTP and EP (50 mg/kg).

Close modal

It has been shown that MPO is upregulated in astrocytes in the SN of PD patients and MPTP-treated mice; furthermore, mice deficient in MPO are resistant to MPTP neurotoxicity (35). Thus, we examined whether EP inhibited MPTP-induced expression of MPO in the SN, which would be predicted to enhance neuronal survival. Western blot analyses showed that the levels of MPO were 4-fold higher in the MPTP-treated SN than that in the SN of PBS-treated controls (p < 0.001; Fig. 7G, 7H). Treatment with EP after MPTP injection dramatically decreased the level of MPO protein expression by 61% (p < 0.001; Fig. 7G, 7H) but alone had no effect. Double immunostaining showed MPO expression in GFAP–ip astrocytes (Supplemental Fig. 1A–C) but not in microglia (Supplemental Fig. 1D–F). The number of MPO and GFAP double-ip (MPO+/GFAP+) cells in the MPTP-treated SN was 4-fold higher than in PSB- or EP-treated controls (p < 0.001; Fig. 7H).

In this study, we demonstrated that EP protects nigrostriatal DA neurons from MPTP neurotoxicity in vivo by regulating glial activation and the resultant oxidative stress. We showed that EP suppressed MPTP-induced ROS/RNS generation and reduced oxidative damage to nucleic acids by inhibiting microglia-derived NADPH oxidase and/or iNOS and astrocyte-derived MPO, leading to survival of nigrostriatal DA neurons, recovery of striatal dopamine depletion in vivo, and reversal of motor dysfunction. To our knowledge, this is the first study to show that EP rescues nigrostriatal DA neurons through regulation of glial activation in the MPTP model of PD.

It is well-known that EP increases the level of intracellular Py and tricarboxylic acid metabolites because EP is cleaved into ethanol and Py by numerous esterases (36). Several lines of evidence have shown that Py decreases infarct volume in the ischemic brain (11) and protects neuroblastoma cells against MPP+ or 6-hydroxydopamine neurotoxicity (37). To verify whether neuroprotective effects of EP are associated with metabolic recovery, additional experiments were performed to assess neuroprotective effects of Py in MPTP mice. In the present study, we showed that Py protects nigrostriatal DA neurons from MPTP neurotoxicity as evidenced by TH immunostaining, indicating that neuroprotective effects of EP may be due to metabolic protection.

Microglia, resident immunocompetent, and phagocytic cells play an important role in supporting neurons in the CNS (38). However, in the presence of adverse stimuli, they may become activated and contribute to chronic, damaging inflammation and, ultimately, to neurodegeneration. Many studies have reported the presence of reactive microglia in the SN of PD patients (39, 40) and in the SN of animal models of PD produced by administration of MPTP (15, 41). Activated microglia produce NADPH oxidase- and/or iNOS-derived ROS/RNS, which trigger or exacerbate neurotoxicity by inducing oxidative stress to neurons (19, 35).

Activated NADPH oxidase is evident in the SN of PD patients (7) and in the SN of MPTP mouse PD models (15). NADPH oxidase is composed of the cytosolic components p47phox, p67phox, and Rac1 as well as the membrane components gp91phox and p22phox (42, 43). Upon stimulation, the oxidase complex is activated, a process that involves phosphorylation of p47phox on serine residues and recruitment of the cytosolic components to the membrane-bound components, including gp91phox, to assemble the active NADPH oxidase (44, 45). Thus, both phosphorylation of p47phox and binding of p47phox and gp91phox are thought to be important aspects of NADPH oxidase activation (46); the resulting activation leads to ROS generation (18). The generation of ROS by NADPH oxidase is increased in the SN of PD patients (7) and in the SN of MPTP-treated mice (15), and oxidants originating from microglia are thought to mediate the loss of DA neurons in the SN (47). Several studies have provided evidence for oxidative stress in PD patients and in MPTP-treated mice, including high levels of oxidative nucleic acid damage, as visualized by 8-OHdG immunostaining in the SN (15, 48). The results of the current study show that MPTP activated NADPH oxidase, as demonstrated by increases in phosphorylation of p47phox serine residue and binding of phospho-p47phox and gp91phox. NADPH oxidase activation resulted in increased O2 and O2-derived oxidants and DNA damage, as visualized by hydroethidine staining and 8-OHdG immunostaining in the SN, respectively. Treatment with EP not only inhibited microglial NADPH oxidase activation but also mitigated ROS production and nucleic acid oxidation. These results verify that EP inhibited MPTP-induced activation of NADPH oxidase and oxidative damage, thereby resulting in neuroprotection in the MPTP model.

RNS, another major contributor to oxidative stress, has been implicated in the pathogenesis and progression of PD (49). Moreover, NO, synthesized by iNOS, reacts with superoxide to form peroxynitrate, which subsequently induces oxidative stress to proteins by nitration of tyrosine residues (31, 32, 50). Several studies have demonstrated that reactive glia that express iNOS and/or increased levels of nitrotyrosine are present in the midbrains of PD patients (51, 52). In the MPTP mouse model of PD, iNOS contributes to DA neuronal death through MPTP-induced RNS and increases in nitrotyrosine levels (5, 53), although the existence of iNOS in human microglia remains controversial (54). Treatment with EP not only reduced iNOS expression but also decreased the levels of TH nitrotyrosine (Supplemental Fig. 1G) in the SN of MPTP-treated mice. Collectively, these results suggest that EP decreases MPTP-induced expression of iNOS and oxidative stress, leading to increased neuronal survival.

Although the role of activated astrocytes remains controversial, accumulating evidence obtained from the MPTP model suggests that activation of astrocytes can contribute to the loss of nigrostriatal DA neurons via production of neurotoxic substances (6, 55, 56). MPO, which is expressed in activated astrocytes, is the key enzyme involved in the generation of cytotoxic ROS/RNS (57, 58). These astroglial MPO-derived oxidants can be released and subsequently exert toxic effects on adjacent mesencephalic DA neurons in the MPTP model (15). A role for this mechanism is supported by evidence that increased levels of 3-chlorotyrosine- and hypochlorous acid-modified proteins—MPO-derived oxidative stress markers—are present in the MPTP-treated SN (6). Alternatively, MPO secretion may participate in DA neuronal death in the MPTP model of PD via microglia activation (59). Although potential cytokine-like effects of MPO are still not well-known, this enzyme may play an important role in the signaling pathway of microglial activation that results in neuronal death (15, 59). This is consistent with our present data showing that MPO expression in activated astrocytes was increased in the MPTP-treated SN, as evidenced by an increase in the number of MPO+/GFAP+ cells. Western blot analyses also confirmed increases in MPO expression. Notably, EP significantly attenuated the levels of MPO expression in the SN. Similar to our recent report (15), the present findings suggest that EP rescues DA neuronal death via suppression of MPO expression in activated astrocytes. Interestingly, in spinal cord injury, EP reduces MPO expression originating in neutrophils but not in astrocytes (60). This discrepancy may reflect differences in the animal models used (PD versus spinal cord injury) and/or regions studied (SN versus spinal cord) in each experiment.

In the current study, we found that EP failed to protect DA neurons from MPP+ neurotoxicity in neuron-enriched mesencephalic cultures, as assessed by TH immunostaining. By contrast, recent data have demonstrated that the same dose of EP as used in our experiments (1 mM) decreases neuronal cell death caused by MPP+ neurotoxicity in SH–SY5Y neuroblastoma cells, as assessed using the tetrazolium-based MTT assay (61). The apparent discrepancy in experimental results between these two studies is probably a result of differences in the cell types used (primary mesencephalic neurons versus neuroblastoma cells). The neuroprotective effects of EP in cocultures of mesencephalic microglia and neurons also provide further evidence that EP acts through microglia to mediate its neuroprotective effects under our experimental conditions. Although not tested directly, these results provide indirect evidence that the neuroprotective effects of EP do not involve blocking MPP+ entry into DA neurons.

EP has been known to reinforce mitochondrial functions such as the production of ATP in cerebrocortical slice cultures (36) and ROS in PC12 cells and cortical microglia cultures (13, 62), which results in neuronal survival. Taking this into consideration, the current data show that EP aids neuronal survival by affecting mitochondrial functions (increased ATP production and decreased ROS levels) in the cocultures of mesencephanlic microglia and neurons (Supplemental Fig. 3A, 3B). The data carefully suggest that the effect of EP on mitochondria is possibly an additional protective mechanism. However, this might be irrelevant for our study because even though EP can produce similar amounts of ATP and ROS (Supplemental Fig. 3A, 3B), it fails to show neuroprotection in neuron-enriched cultures. Therefore, it is likely that under our experimental conditions, the effect of EP on mitochondrial functions is an indirect contribution to neuronal survival. Although we do not provide direct evidence, the speculation is that it does so by regulating microglial-mediated inflammation.

The most prominent biochemical change in the STR of PD patients and MPTP-treated mice is a decrease in the levels of dopamine (2, 14). In our study, such deficits in striatal dopamine in MPTP-treated mice led to a decreased latency to fall from an accelerating rotarod apparatus, reflecting diminished coordination and balance (15). EP was found to increase striatal dopamine levels and to ameliorate motor deficits in MPTP-treated mice. These behavioral and in vivo biochemical effects of EP on the lesioned nigrostriatal DA system, together with the present finding of inhibitory actions of EP on glia-mediated oxidative stress, suggest that EP may be beneficial in treating PD related to neuroinflammation.

It is worth considering whether the neuroprotection afforded by EP results in the partial restoration or prevention of further decreases in DA neurons in the nigrostriatal pathway. Our recent study revealed that MPTP reduced DA neurons by 17% in the SN and the OD of striatal TH-ip fibers by 15% at 12 h after the last MPTP injection, indicating that a majority of nigrostriatal TH-ip neurons are viable (15). Therefore, it is likely that the observed neuroprotective effects are due to the prevention of further decreases in nigrostriatal DA neurons. Although we did not provide direct evidence, it is also possible that partial restoration may account for the neuroprotective actions of EP.

In conclusion, in view of the clinical safety of EP and its ability to effectively penetrate the blood–brain barrier (63), we propose that EP may have therapeutic value in the treatment of aspects of PD and other disorders associated with neuroinflammation and glial activation-mediated oxidative damage.

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education, Science, and Technology (Grant 20090063274).

The online version of this article contains supplemental material.

Abbreviations used in this article:

DA

dopaminergic

DIV

day in vitro

E

embryonic day

ED-1

clone ED1 CD68 microglial/macrophage marker

EP

ethyl pyruvate

GFAP

glial fibrillary acidic protein

Iba-1

ionized calcium-binding adaptor molecule 1

iNOS

inducible NO synthase

ip

immunopositive

MAC-1

macrophage Ag complex-1

MPO

myeloperoxidase

MPP+

1-methyl-4-phenyl-pyridinium

8-OHdG

8-hydroxy-2′-deoxyguanosine

PD

Parkinson’s disease

Py

sodium pyruvate

RNS

reactive nitrogen species

ROS

reactive oxygen species

SN

substantia nigra

SNpc

substantia nigra pars compacta

STR

striatum

TH

tyrosine hydroxylase.

1
Dauer
W.
,
Przedborski
S.
.
2003
.
Parkinson’s disease: mechanisms and models.
Neuron
39
:
889
909
.
2
Savitt
J. M.
,
Dawson
V. L.
,
Dawson
T. M.
.
2006
.
Diagnosis and treatment of Parkinson disease: molecules to medicine.
J. Clin. Invest.
116
:
1744
1754
.
3
Lang
A. E.
,
Lozano
A. M.
.
1998
.
Parkinson’s disease: first of two parts.
N. Engl. J. Med.
339
:
1044
1053
.
4
Hirsch
E. C.
,
Hunot
S.
.
2009
.
Neuroinflammation in Parkinson’s disease: a target for neuroprotection?
Lancet Neurol.
8
:
382
397
.
5
Liberatore
G. T.
,
Jackson-Lewis
V.
,
Vukosavic
S.
,
Mandir
A. S.
,
Vila
M.
,
McAuliffe
W. G.
,
Dawson
V. L.
,
Dawson
T. M.
,
Przedborski
S.
.
1999
.
Inducible nitric oxide synthase stimulates dopaminergic neurodegeneration in the MPTP model of Parkinson disease.
Nat. Med.
5
:
1403
1409
.
6
Choi
D. K.
,
Pennathur
S.
,
Perier
C.
,
Tieu
K.
,
Teismann
P.
,
Wu
D. C.
,
Jackson-Lewis
V.
,
Vila
M.
,
Vonsattel
J. P.
,
Heinecke
J. W.
,
Przedborski
S.
.
2005
.
Ablation of the inflammatory enzyme myeloperoxidase mitigates features of Parkinson’s disease in mice.
J. Neurosci.
25
:
6594
6600
.
7
Wu
D. C.
,
Teismann
P.
,
Tieu
K.
,
Vila
M.
,
Jackson-Lewis
V.
,
Ischiropoulos
H.
,
Przedborski
S.
.
2003
.
NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease.
Proc. Natl. Acad. Sci. USA
100
:
6145
6150
.
8
Gao
H. M.
,
Liu
B.
,
Zhang
W.
,
Hong
J. S.
.
2003
.
Critical role of microglial NADPH oxidase-derived free radicals in the in vitro MPTP model of Parkinson’s disease.
FASEB J.
17
:
1954
1956
.
9
Sims
C. A.
,
Wattanasirichaigoon
S.
,
Menconi
M. J.
,
Ajami
A. M.
,
Fink
M. P.
.
2001
.
Ringer’s ethyl pyruvate solution ameliorates ischemia/reperfusion-induced intestinal mucosal injury in rats.
Crit. Care Med.
29
:
1513
1518
.
10
Kao
K. K.
,
Fink
M. P.
.
2010
.
The biochemical basis for the anti-inflammatory and cytoprotective actions of ethyl pyruvate and related compounds.
Biochem. Pharmacol.
80
:
151
159
.
11
Yu
Y. M.
,
Kim
J. B.
,
Lee
K. W.
,
Kim
S. Y.
,
Han
P. L.
,
Lee
J. K.
.
2005
.
Inhibition of the cerebral ischemic injury by ethyl pyruvate with a wide therapeutic window.
Stroke
36
:
2238
2243
.
12
Cho
I. H.
,
Kim
S. W.
,
Kim
J. B.
,
Kim
T. K.
,
Lee
K. W.
,
Han
P. L.
,
Lee
J. K.
.
2006
.
Ethyl pyruvate attenuates kainic acid-induced neuronal cell death in the mouse hippocampus.
J. Neurosci. Res.
84
:
1505
1511
.
13
Wang
L. Z.
,
Sun
W. C.
,
Zhu
X. Z.
.
2005
.
Ethyl pyruvate protects PC12 cells from dopamine-induced apoptosis.
Eur. J. Pharmacol.
508
:
57
68
.
14
Jackson-Lewis
V.
,
Przedborski
S.
.
2007
.
Protocol for the MPTP mouse model of Parkinson’s disease.
Nat. Protoc.
2
:
141
151
.
15
Chung
Y. C.
,
Kim
S. R.
,
Jin
B. K.
.
2010
.
Paroxetine prevents loss of nigrostriatal dopaminergic neurons by inhibiting brain inflammation and oxidative stress in an experimental model of Parkinson’s disease.
J. Immunol.
185
:
1230
1237
.
16
Paxinos
G.
,
Franklin
K.
.
2001
.
The Mouse Brain in Stereotaxic Coordinates.
Academic Press
,
San Diego, CA
.
17
West
M. J.
,
Slomianka
L.
,
Gundersen
H. J.
.
1991
.
Unbiased stereological estimation of the total number of neurons in thesubdivisions of the rat hippocampus using the optical fractionator.
Anat. Rec.
231
:
482
497
.
18
Park
K. W.
,
Jin
B. K.
.
2008
.
Thrombin-induced oxidative stress contributes to the death of hippocampal neurons: role of neuronal NADPH oxidase.
J. Neurosci. Res.
86
:
1053
1063
.
19
Choi
S. H.
,
Lee
D. Y.
,
Chung
E. S.
,
Hong
Y. B.
,
Kim
S. U.
,
Jin
B. K.
.
2005
.
Inhibition of thrombin-induced microglial activation and NADPH oxidase by minocycline protects dopaminergic neurons in the substantia nigra in vivo.
J. Neurochem.
95
:
1755
1765
.
20
Han
B. S.
,
Hong
H. S.
,
Choi
W. S.
,
Markelonis
G. J.
,
Oh
T. H.
,
Oh
Y. J.
.
2003
.
Caspase-dependent and -independent cell death pathways in primary cultures of mesencephalic dopaminergic neurons after neurotoxin treatment.
J. Neurosci.
23
:
5069
5078
.
21
Choi
S. H.
,
Lee
D. Y.
,
Ryu
J. K.
,
Kim
J.
,
Joe
E. H.
,
Jin
B. K.
.
2003
.
Thrombin induces nigral dopaminergic neurodegeneration in vivo by altering expression of death-related proteins.
Neurobiol. Dis.
14
:
181
193
.
22
Chung
E. S.
,
Jin
B. K.
.
2006
.
Disialogangliosides induce neurodegeneration in rat mesencephalic cultures.
Biochem. Biophys. Res. Commun.
346
:
572
577
.
23
Kim
W. G.
,
Mohney
R. P.
,
Wilson
B.
,
Jeohn
G. H.
,
Liu
B.
,
Hong
J. S.
.
2000
.
Regional difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat brain: role of microglia.
J. Neurosci.
20
:
6309
6316
.
24
Lee
D. Y.
,
Oh
Y. J.
,
Jin
B. K.
.
2005
.
Thrombin-activated microglia contribute to death of dopaminergic neurons in rat mesencephalic cultures: dual roles of mitogen-activated protein kinase signaling pathways.
Glia
51
:
98
110
.
25
Nayyar
T.
,
Bubser
M.
,
Ferguson
M. C.
,
Neely
M. D.
,
Shawn Goodwin
J.
,
Montine
T. J.
,
Deutch
A. Y.
,
Ansah
T. A.
.
2009
.
Cortical serotonin and norepinephrine denervation in parkinsonism: preferential loss of the beaded serotonin innervation.
Eur. J. Neurosci.
30
:
207
216
.
26
Luchtman
D. W.
,
Shao
D.
,
Song
C.
.
2009
.
Behavior, neurotransmitters and inflammation in three regimens of the MPTP mouse model of Parkinson’s disease.
Physiol. Behav.
98
:
130
138
.
27
Block
M. L.
,
Zecca
L.
,
Hong
J. S.
.
2007
.
Microglia-mediated neurotoxicity: uncovering the molecular mechanisms.
Nat. Rev. Neurosci.
8
:
57
69
.
28
Henkel
J. S.
,
Beers
D. R.
,
Siklós
L.
,
Appel
S. H.
.
2006
.
The chemokine MCP-1 and the dendritic and myeloid cells it attracts are increased in the mSOD1 mouse model of ALS.
Mol. Cell. Neurosci.
31
:
427
437
.
29
Travaglione
S.
,
Falzano
L.
,
Fabbri
A.
,
Stringaro
A.
,
Fais
S.
,
Fiorentini
C.
.
2002
.
Epithelial cells and expression of the phagocytic marker CD68: scavenging of apoptotic bodies following Rho activation.
Toxicol. In Vitro
16
:
405
411
.
30
Zhou
C.
,
Huang
Y.
,
Przedborski
S.
.
2008
.
Oxidative stress in Parkinson’s disease: a mechanism of pathogenic and therapeutic significance.
Ann. N. Y. Acad. Sci.
1147
:
93
104
.
31
Dawson
T. M.
,
Dawson
V. L.
.
2003
.
Molecular pathways of neurodegeneration in Parkinson’s disease.
Science
302
:
819
822
.
32
Liang
L. P.
,
Huang
J.
,
Fulton
R.
,
Day
B. J.
,
Patel
M.
.
2007
.
An orally active catalytic metalloporphyrin protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity in vivo.
J. Neurosci.
27
:
4326
4333
.
33
Ara
J.
,
Przedborski
S.
,
Naini
A. B.
,
Jackson-Lewis
V.
,
Trifiletti
R. R.
,
Horwitz
J.
,
Ischiropoulos
H.
.
1998
.
Inactivation of tyrosine hydroxylase by nitration following exposure to peroxynitrite and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP).
Proc. Natl. Acad. Sci. USA
95
:
7659
7663
.
34
Blanchard-Fillion
B.
,
Souza
J. M.
,
Friel
T.
,
Jiang
G. C.
,
Vrana
K.
,
Sharov
V.
,
Barrón
L.
,
Schöneich
C.
,
Quijano
C.
,
Alvarez
B.
, et al
.
2001
.
Nitration and inactivation of tyrosine hydroxylase by peroxynitrite.
J. Biol. Chem.
276
:
46017
46023
.
35
Choi
S. H.
,
Lee
D. Y.
,
Kim
S. U.
,
Jin
B. K.
.
2005
.
Thrombin-induced oxidative stress contributes to the death of hippocampal neurons in vivo: role of microglial NADPH oxidase.
J. Neurosci.
25
:
4082
4090
.
36
Zeng
J.
,
Liu
J.
,
Yang
G. Y.
,
Kelly
M. J.
,
James
T. L.
,
Litt
L.
.
2007
.
Exogenous ethyl pyruvate versus pyruvate during metabolic recovery after oxidative stress in neonatal rat cerebrocortical slices.
Anesthesiology
107
:
630
640
.
37
Mazzio
E.
,
Soliman
K. F.
.
2003
.
Pyruvic acid cytoprotection against 1-methyl-4-phenylpyridinium, 6-hydroxydopamine and hydrogen peroxide toxicities in vitro.
Neurosci. Lett.
337
:
77
80
.
38
Graeber
M. B.
,
Streit
W. J.
.
2010
.
Microglia: biology and pathology.
Acta Neuropathol.
119
:
89
105
.
39
McGeer
P. L.
,
McGeer
E. G.
.
2008
.
Glial reactions in Parkinson’s disease.
Mov. Disord.
23
:
474
483
.
40
Mirza
B.
,
Hadberg
H.
,
Thomsen
P.
,
Moos
T.
.
2000
.
The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson’s disease.
Neuroscience
95
:
425
432
.
41
Vroon
A.
,
Drukarch
B.
,
Bol
J. G.
,
Cras
P.
,
Brevé
J. J.
,
Allan
S. M.
,
Relton
J. K.
,
Hoogland
P. V.
,
Van Dam
A. M.
.
2007
.
Neuroinflammation in Parkinson’s patients and MPTP-treated mice is not restricted to the nigrostriatal system: microgliosis and differential expression of interleukin-1 receptors in the olfactory bulb.
Exp. Gerontol.
42
:
762
771
.
42
Babior
B. M.
1999
.
NADPH oxidase: an update.
Blood
93
:
1464
1476
.
43
Cross
A. R.
,
Segal
A. W.
.
2004
.
The NADPH oxidase of professional phagocytes—prototype of the NOX electron transport chain systems.
Biochim. Biophys. Acta
1657
:
1
22
.
44
Lambeth
J. D.
2004
.
NOX enzymes and the biology of reactive oxygen.
Nat. Rev. Immunol.
4
:
181
189
.
45
Groemping
Y.
,
Rittinger
K.
.
2005
.
Activation and assembly of the NADPH oxidase: a structural perspective.
Biochem. J.
386
:
401
416
.
46
Fan
J.
,
Li
Y.
,
Levy
R. M.
,
Fan
J. J.
,
Hackam
D. J.
,
Vodovotz
Y.
,
Yang
H.
,
Tracey
K. J.
,
Billiar
T. R.
,
Wilson
M. A.
.
2007
.
Hemorrhagic shock induces NAD(P)H oxidase activation in neutrophils: role of HMGB1-TLR4 signaling.
J. Immunol.
178
:
6573
6580
.
47
Wu
D. C.
,
Jackson-Lewis
V.
,
Vila
M.
,
Tieu
K.
,
Teismann
P.
,
Vadseth
C.
,
Choi
D. K.
,
Ischiropoulos
H.
,
Przedborski
S.
.
2002
.
Blockade of microglial activation is neuroprotective in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson disease.
J. Neurosci.
22
:
1763
1771
.
48
Nakabeppu
Y.
,
Tsuchimoto
D.
,
Yamaguchi
H.
,
Sakumi
K.
.
2007
.
Oxidative damage in nucleic acids and Parkinson’s disease.
J. Neurosci. Res.
85
:
919
934
.
49
Aquilano
K.
,
Baldelli
S.
,
Rotilio
G.
,
Ciriolo
M. R.
.
2008
.
Role of nitric oxide synthases in Parkinson’s disease: a review on the antioxidant and anti-inflammatory activity of polyphenols.
Neurochem. Res.
33
:
2416
2426
.
50
Kavya
R.
,
Saluja
R.
,
Singh
S.
,
Dikshit
M.
.
2006
.
Nitric oxide synthase regulation and diversity: implications in Parkinson’s disease.
Nitric Oxide
15
:
280
294
.
51
Giasson
B. I.
,
Duda
J. E.
,
Murray
I. V.
,
Chen
Q.
,
Souza
J. M.
,
Hurtig
H. I.
,
Ischiropoulos
H.
,
Trojanowski
J. Q.
,
Lee
V. M.
.
2000
.
Oxidative damage linked to neurodegeneration by selective α-synuclein nitration in synucleinopathy lesions.
Science
290
:
985
989
.
52
Hunot
S.
,
Boissière
F.
,
Faucheux
B.
,
Brugg
B.
,
Mouatt-Prigent
A.
,
Agid
Y.
,
Hirsch
E. C.
.
1996
.
Nitric oxide synthase and neuronal vulnerability in Parkinson’s disease.
Neuroscience
72
:
355
363
.
53
Dehmer
T.
,
Lindenau
J.
,
Haid
S.
,
Dichgans
J.
,
Schulz
J. B.
.
2000
.
Deficiency of inducible nitric oxide synthase protects against MPTP toxicity in vivo.
J. Neurochem.
74
:
2213
2216
.
54
Heneka
M. T.
,
Wiesinger
H.
,
Dumitrescu-Ozimek
L.
,
Riederer
P.
,
Feinstein
D. L.
,
Klockgether
T.
.
2001
.
Neuronal and glial coexpression of argininosuccinate synthetase and inducible nitric oxide synthase in Alzheimer disease.
J. Neuropathol. Exp. Neurol.
60
:
906
916
.
55
Oki
C.
,
Watanabe
Y.
,
Yokoyama
H.
,
Shimoda
T.
,
Kato
H.
,
Araki
T.
.
2008
.
Delayed treatment with arundic acid reduces the MPTP-induced neurotoxicity in mice.
Cell. Mol. Neurobiol.
28
:
417
430
.
56
Bolin
L. M.
,
Strycharska-Orczyk
I.
,
Murray
R.
,
Langston
J. W.
,
Di Monte
D.
.
2002
.
Increased vulnerability of dopaminergic neurons in MPTP-lesioned interleukin-6 deficient mice.
J. Neurochem.
83
:
167
175
.
57
Arnhold
J.
,
Flemmig
J.
.
2010
.
Human myeloperoxidase in innate and acquired immunity.
Arch. Biochem. Biophys.
500
:
92
106
.
58
Hampton
M. B.
,
Kettle
A. J.
,
Winterbourn
C. C.
.
1998
.
Inside the neutrophil phagosome: oxidants, myeloperoxidase, and bacterial killing.
Blood
92
:
3007
3017
.
59
Lau
D.
,
Mollnau
H.
,
Eiserich
J. P.
,
Freeman
B. A.
,
Daiber
A.
,
Gehling
U. M.
,
Brümmer
J.
,
Rudolph
V.
,
Münzel
T.
,
Heitzer
T.
, et al
.
2005
.
Myeloperoxidase mediates neutrophil activation by association with CD11b/CD18 integrins.
Proc. Natl. Acad. Sci. USA
102
:
431
436
.
60
Genovese
T.
,
Esposito
E.
,
Mazzon
E.
,
Di Paola
R.
,
Meli
R.
,
Caminiti
R.
,
Bramanti
P.
,
Fink
M. P.
,
Cuzzocrea
S.
.
2009
.
Beneficial effects of ethyl pyruvate in a mouse model of spinal cord injury.
Shock
32
:
217
227
.
61
Choi
J. S.
,
Lee
M. S.
,
Jeong
J. W.
.
2010
.
Ethyl pyruvate has a neuroprotective effect through activation of extracellular signal-regulated kinase in Parkinson’s disease model.
Biochem. Biophys. Res. Commun.
394
:
854
858
.
62
Kim
J. B.
,
Yu
Y. M.
,
Kim
S. W.
,
Lee
J. K.
.
2005
.
Anti-inflammatory mechanism is involved in ethyl pyruvate-mediated efficacious neuroprotection in the postischemic brain.
Brain Res.
1060
:
188
192
.
63
Hurd
R. E.
,
Yen
Y. F.
,
Mayer
D.
,
Chen
A.
,
Wilson
D.
,
Kohler
S.
,
Bok
R.
,
Vigneron
D.
,
Kurhanewicz
J.
,
Tropp
J.
, et al
.
2010
.
Metabolic imaging in the anesthetized rat brain using hyperpolarized [1-13C] pyruvate and [1-13C] ethyl pyruvate.
Magn. Reson. Med.
63
:
1137
1143
.

The authors have no financial conflicts of interest.