GET THE APP

Alpha-Tocopherol Counteracts Cognitive and Motor Deficits Induced
Biochemistry & Pharmacology: Open Access

Biochemistry & Pharmacology: Open Access
Open Access

ISSN: 2167-0501

+44-20-4587-4809

Research Article - (2015) Volume 4, Issue 1

Alpha-Tocopherol Counteracts Cognitive and Motor Deficits Induced by Repeated Treatment with Reserpine

Aldair José Sarmento-Silva1, Ramón Hypolito Lima1, Alicia Cabral1, Ywlliane Meurer1, Alessandra Mussi Ribeiro1,2 and Regina Helena Silva1,3*
1Memory Studies Laboratory, Physiology Department, Federal University of Rio Grande do Norte, Natal, Brazil
2Department of Biosciences, Federal University of São Paulo, Santos, Brazil
3Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
*Corresponding Author: Regina Helena Silva, Departamento de Farmacologia – UNIFESP, Rua Botucatu, 862, Edifício Leal Prado, 1º.andar, CEP 04023062 - São Paulo, SP, Brazil Email:

Abstract

Previous studies showed that chronic administration of the monoamine depleting agent reserpine in low doses promotes progressive cognitive and motor impairments in rats, and this protocol has been used as a pharmacological progressive model of Parkinson's disease. These behavioral alterations are accompanied by increased brain oxidative stress. We aimed to verify the effects of the concomitant treatment with the antioxidant agent alpha-tocopherol on the motor and cognitive deficits induced by chronic reserpine in rats. Rats were repeatedly treated with 0.1 mg/kg reserpine with or without a concomitant treatment with 40 mg/kg alpha-tocopherol. Across the treatment, motor and cognitive performances were evaluated by the catalepsy and novel object recognition tests, respectively. As expected, reserpinetreated rats showed progressively increased duration of catalepsy together with short-term memory deficits in the object recognition test. Importantly, these detrimental outcomes due to reserpine treatment were prevented by concomitant daily administration of the antioxidant agent alpha-tocopherol. The results show a preventive role of alpha-tocopherol on behavioral alterations induced by repeated reserpine treatment. This is relevant to the investigation of possible neuroprotective interventions in Parkinson’s disease.

<

Keywords: Reserpine; Parkinson’s disease; α-tocopherol; Motor impairment; Short-term memory impairment

Abbreviations

NOR: Novel Object Recognition; PD: Parkinson’s Disease; RES: Reserpine; ROS: Reactive Oxygen Species; TOC: Alpha- Tocopherol; VR: Vehicle For Reserpine; VT: Vehicle For Alpha- Tocopherol; PKC: Protein Kinase C

Introduction

Reserpine precludes the storage of monoamines through the blockage of the synaptic vesicles transporters [1]. Consequently, synaptic vesicles are still available but there is a reduction in the amount of dopamine in the synaptic cleft. Because an important loss of dopaminergic neurons is the core feature of Parkinson´s disease (PD) [2], reserpine administration to rodents is a valid approach to study this disease in animal models [3-5]. The acute administration of a high dose of reserpine (above 1.0 mg/kg) leads to severe motor impairment [4]. In addition, acute injection of reserpine in lower doses causes memory deficits in the absence of motor damage [6,7]. However, although both cognitive and motor impairments are symptoms of PD, their emergence shortly after an acute injection is not compatible with the gradual progression of symptoms found in the clinical situation. More recently, studies have shown that the chronic administration of reserpine in low doses can promote progressive cognitive and motor impairments, along with decreased tyrosine hydroxylase levels in the nigrostriatal pathway [8]. This protocol is suggested as a progressive pharmacological model of PD [8,9].

Besides its classical mechanism of action (i.e. blockage of the vesicular transport of monoamines), there is clear evidence that reserpine also causes an increase in cellular oxidative stress, possibly potentiated by the rise in the levels of dopamine in the cytoplasm, which undergoes oxidative metabolism [10]. In this respect, the central nervous system is quite vulnerable to reactive oxygen species (ROS), which play a very important function in the pathogenesis of neurodegenerative disorders, including PD [11]. For example, there is evidence that the inclusion of antioxidant agents in the pharmacological treatment of PD has advantages over the treatment based only in dopamine replacement [11-13]. In addition, the repeated treatment with reserpine that induces progressive features compatible with PD also leads to increased brain oxidative stress [9]. However, it is unclear if a possible oxidative damage is responsible for the behavioral deficits presented by animals repeatedly treated with reserpine.

Antioxidant agents mainly act as a reinforcement of endogenous antioxidant defenses. An important antioxidant agent is vitamin E (alpha-tocopherol; TOC), which plays an essential role in protecting the body against the damaging effects of ROS. Specifically, TOC blocks the propagation step of lipid peroxidation of polyunsaturated fatty acids in membranes and lipoproteins [14], mainly by neutralizing the effects of peroxides and oxygen free radicals [15].

The aim of this study was to evaluate the effects of the antioxidant agent TOC on motor, cognitive and neuronal parameters in animals submitted to a progressive pharmacological animal model of PD, i.e., the repeated treatment with a low dose of reserpine.

Materials and Methods

Animals

We used 75 five-month-old male Wistar rats (300-500 g). The animals were obtained from the Physiology Department at the Federal University of Rio Grande do Norte, and were housed in groups of four, in plastic cages, under controlled conditions of ventilation, temperature (23 ± 1ºC), and light/dark cycle (12h/12h, lights on 6:30 a.m.), with free access to water and food. The rats were handled according to the Brazilian law for the use of animals in scientific research (Law Number 11.794) and all the procedures described were approved by the local ethical committee (CEUA/UFRN nº 051/2011).

Drugs

Reserpine (RES; Sigma Chemical Co., St. Louis, MO) was dissolved in acetic acid and further diluted in distilled water at the concentration of 0.1 mg/mL, pH ≈ 6.5. We used this vehicle (glacial acetic acid diluted in water) as a control for reserpine treatment (VR). RES and VR were given s.c. on alternate days. The antioxidant alpha-tocopherol (TOC; Sigma Chemical Co., St. Louis, MO) was diluted in distilled water with Tween-80 at the concentration of 40 mg/mL. We used the vehicle used to dilute TOC (VT) as a control for TOC treatment. These solutions were injected i.p. daily. The volume of injection was 1 mL/kg of body weight in all cases. We prepared all solutions every 48 hours and kept them at 4ºC between administrations.

Experimental design

The rats were randomly assigned to the following groups: VR + VT (n=18), RES + VT (n=19), RES + TOC (n=19) and VR + TOC (n=19). Drug treatment lasted 30 days. Animals received 15 s.c. injections of RES (0.1 mg/kg) or VR every 48 hours, concomitantly to daily i.p. administration of TOC (40 mg/kg) or VT.

Before the beginning of the experiments, all animals were submitted to a daily 5-minute handling session for five consecutive days. Throughout the treatment, all the animals were subjected to catalepsy tests (performed daily) and part of the animals (n=35, 7-11 per group) went through the novel object recognition (NOR) tasks (days 2, 12 and 18 of treatment). The experimental design is shown in Figure 1. Both behavioral tests were performed as described in our previous study [8] and were conducted before the injections of that day. Thus, all behavioral evaluations were performed 48h after the last injection of reserpine in order to avoid acute effects of the drug. NOR sessions were recorded with a digital camera fixed above the arena and the behavior was analyzed through video-tracking software (Anymaze, Stoelting Co, Wood Dale, Illinois, and USA). Before each experimental procedure, the apparatuses were cleaned with a 5% alcohol solution, and the experimental groups were alternated across testing.

biochemistry-pharmacology-experimental-design

Figure 1: Schematic illustration of the experimental design.

Statistical analysis

We analyzed the performances in catalepsy test (total time spent in immobility until the animal removed both forepaws of the bar) by the two-way ANOVA with repeated measures followed by Tukey’s multiple comparison post hoc test. In the NOR task we conducted one-way ANOVA followed by Bonferroni’s multiple comparison post hoc test in order to compare old versus familiar object exploration. Analyses for the exploration ratio throughout test sessions and among experimental groups were conducted through two-way ANOVA followed by Tukey’s Post Hoc test.

Results

Catalepsy

Figure 2 shows that from day 15 onwards there was an increase in catalepsy behavior of the group RES + VT compared to all other groups (RM two-way ANOVA; days of treatment [F(29,2130) = 16.72, P < 0.0001], treatment [F(3,2130) = 211.0, P < 0.0001] and days of treatment × treatment interaction effects [F(87,2130) = 4.876, P < 0.0001]). This increase was not detected for the group RES+TOC.

biochemistry-pharmacology-Repeated-administration

Figure 2: Repeated administration of reserpine increases catalepsy duration and this effect is prevented by α-tocopherol. Animals were placed daily in a catalepsy bar and the latency to step-down was registered. Arrows indicate reserpine (RES; 0.1 mg/kg) or vehicle (VR) s.c. injections, while α-tocopherol (TOC; 40 mg/kg) or its vehicle (VT) were administered through daily i.p. injections. Data are expressed as mean + SEM; (*) P < 0.05 for RES + VT vs. RES+TOC; (#) P < 0.01 for RES + VT vs. VR+VT; (***) P < 0.001 and (****) P < 0.0001 for RES + VT vs. all experimental groups in Tukey’s multiple comparison post hoc test after RM two-way ANOVA.

Novel object recognition

We found that all animals spent more time exploring the new object in the second day of protocol (first test; Figure 3A; one-way ANOVA [F(7,62) = 11.23; P < 0.0001]). Reserpine treatment impaired short-term memory after the 12th day of protocol (second and third tests). Conversely, treatment with α-tocopherol was able to prevent the short-term memory impairment (Figure 3B; one-way ANOVA [F(7,74) = 6.864; P < 0.0001] and Figure 3C; one-way ANOVA [F(7,68) = 10.00; P < 0.0001]). We also performed statistical analyses in order to evaluate the effect of drug administration in objects exploration ratio throughout test sessions and among experimental groups. We found that in the third test session animals’ receiving RES differs on exploration rate of new (Table 1; two-way ANOVA [F(6,89) = 2.843; P < 0.05]) and old objects (Table 1; two-way ANOVA [F(6,89) = 2.843; P < 0.05]) when comparing to both VR + VT and RES + TOC. Yet, we found that only RES + VT group presented alterations in object discrimination across tests. More accurately, exploration of old and new objects increased and decreased, respectively, comparing first and second tests (Table 1; two-way ANOVA [F(3,89) = 2.760; P < 0.05]) and first and third tests (Table 1; two-way ANOVA [F(3,89) = 2.649; P < 0.05]).

    Groups
Tests Objects VR + VT RES + VT RES + TOC TOC
First Test
Post 1st injection
Old 36.94 ± 6.61 26.06 ± 5.01 34.11 ± 5.84 40.6 ± 4.91
New 63.06 ± 6.61 73.94 ± 5.01 65.89 ± 5.84 59.4 ± 4.91
Second Test
Post 6th injection
Old 34.69 ± 5.92 43.97 ± 5.31¥ 26.85 ± 5.87 39.17 ± 6.32
New 65.31 ± 5.92 56.03 ± 5.31¥ 73.15 ± 5.87 60.83 ± 6.32
Third Test
Post 9th injection
Old 27.71 ± 4.97 59.99 ± 6.95€ # 31.99 ± 6.43* 37.07 ± 3.25
New 72.28 ± 4.97 40.01 ± 6.95€ # 68.01 ± 6.43* 62.93 ± 3.25

Table 1: Exploration rate in the NOR task throughout the test sessions. Data are expressed as mean ± SEM. (*) P < 0.05 and (€) P < 0.01 when comparing RES + VT vs. RES + TOC and VR + VT vs. RES + VT respectively. (¥) P < 0.05 and (#) P < 0.001 when comparing the first vs. second test and first vs. third test respectively. All statistical analyses were conducted through two-way ANOVA followed by Tukey’s Post Hoc test.

biochemistry-pharmacology-one-hour-interval

Figure 3: Repeated administration of reserpine increases catalepsy duration and this effect is prevented by α-tocopherol. Animals were placed daily in a catalepsy bar and the latency to step-down was registered. Arrows indicate reserpine (RES; 0.1 mg/kg) or vehicle (VR) s.c. injections, while α-tocopherol (TOC; 40 mg/kg) or its vehicle (VT) were administered through daily i.p. injections. Data are expressed as mean + SEM; (*) P < 0.05 for RES + VT vs. RES+TOC; (#) P < 0.01 for RES + VT vs. VR+VT; (***) P < 0.001 and (****) P < 0.0001 for RES + VT vs. all experimental groups in Tukey’s multiple comparison post hoc test after RM two-way ANOVA.

Discussion

In this study, we investigated the effects of concomitant treatment with TOC on catalepsy behavior and NOR task in rats submitted to a chronic treatment with a low dosage of reserpine. We observed that the motor and cognitive impairments induced by chronic treatment with reserpine were prevented by treatment with TOC. These results can be seen in the evaluation of catalepsy behavior performed 48 h after each reserpine injection (Figure 2) as well as in the analysis of exploration time in the novel object recognition task (Figure 3 and Table 1).

As previously observed in studies by our group [8,9], repeated treatment with a low dose (0.1 mg/kg) of reserpine in rats induced the progressive appearance of motor impairment. This impairment is marked by a gradual increase in the duration of catalepsy behavior. Indeed, as one can see in Figure 2, reserpine-treated (RES + VT) animals start differing from control subjects after 7 reserpine s.c. injections. It is well documented that catalepsy in rodents indicates akinesia and rigidity that are important symptoms of PD [16-18]. Importantly, we did not observe this impairment in the group that was concomitantly treated with TOC. Indeed, the group RES + TOC (Figure 2) presented catalepsy duration similar to control across the treatment.

Besides motor assessment, the protocol used in the present study includes the cognitive evaluation. Cognitive deficits have been reported as symptoms of PD, and can even appear before the motor deficits. In a previous study, we have shown that the protocol of reserpine treatment used here induces short-term memory deficits before the appearance of increased catalepsy behavior and other motor signs [8]. The present study corroborates those findings. We used the NOR task, which involves recognition memory and executive functions, both functions that can be impaired in PD [19,20]. Our results corroborated the previous study showing that animals treated with reserpine failed to discriminate the objects in the test session (in the second and third tests, Figure 3). Further, similarly to that described for motor evaluations, the deficit was prevented by TOC administration. Indeed, animals treated with both reserpine and TOC presented increased novel object exploration in all tests, similarly to control subjects. In addition, comparisons among experimental groups showed that animals treated with RES had worse object discrimination compared to both control and RES + TOC groups in the third test. Finally, when performances across the three tests were analyzed, only the group treated with reserpine alone presented discrimination deficits in the second and third tests compared to the first test (Table 1). These additional analyses reinforce the prevention of the reserpine-induced object recognition impairment by co-treatment with TOC.

As mentioned, reserpine is a non-selective inhibitor of the vesicular monoamine transporter [1]. Thus, one could raise the possibility that the behavioral alterations induced by reserpine treatment are related exclusively to the dopamine depletion caused by this blockage. In other words, the alterations could be a consequence of an additive effect on dopaminergic function. However, there is evidence that favors the hypothesis that the progressive effect of the repeated treatment with reserpine is due to oxidative damage. First, a previous study has shown that the classical acute treatment (with a dose 10 times higher than the one we used) did not cause a reduction in tyrosine hydroxylase staining (an indicative of dopaminergic neuronal function), although causing an important motor impairment [21]. Conversely, the protocol used here (repeated treatment with a low dose) reduced tyrosine hydroxylase staining in the substantia nigra and striatum, and part of the alterations induced by the treatment were not recovered after 30 days of treatment withdrawal [8]. Second, it has been shown that reserpine treatment increases brain oxidative stress and this alteration is accompanied by behavioral deficits [10,22,23]. In addition, in a previous study [9] the repeated treatment with a low dose of reserpine induced an increase in striatal level of lipid peroxidation, which occurred concomitantly to the motor impairment. These results lead us to question if cotreatment with TOC would prevent the progressive motor and cognitive alterations induced by the repeated treatment with a low dose of reserpine. As discussed above, treatment with TOC was able to prevent these deficits. This preventive effect might be explained by a neuroprotection mechanism, probably by a reduction the in neurotoxic dopamine oxidation bioproducts [24].

Despite the well-known antioxidant properties of vitamin E, it is important to mention that tocopherol and other antioxidant agents can have pro-oxidant effects as well. Indeed, the ability of these compounds to accept and donate electrons enables them to cause oxidative damage under certain conditions [25]. However, this prooxidant action is mainly found in vitro, and under high concentrations [26,27]. Some in vivo studies have also shown pro-oxidant effects of classical antioxidants, but they are variable depending on substance, concentration, age of the subject and target molecules [25,28-30]. Further, it seems that their preferential action is antioxidant when an oxidant insult from another source is present [31]. In the case of the present results, there was no evidence of a pro-oxidant action regarding possible behavioral alterations.

Nevertheless, an antioxidant role of vitamin E in ameliorating neurodegeneration in PD has been consistently proposed by in vitro and animal studies [32-37]. On the other hand, despite strong evidence favoring an antioxidant effect, the exact mechanism of action of vitamin E in Parkinson´s disease is still under investigation [32]. There is evidence that vitamin E, particularly alpha-tocopherol, can act through other mechanisms not related to modulation of oxidative stress. For example, studies showed that alpha-tocopherol regulates the expression of several genes [38,39] and inhibits protein kinase C (PKC) activity [40,41]. The later could be related to the neuroprotective action of this compound, because PKC activation has been implicated in cell death signaling pathways related to PD [42]. This relationship was found in studies with animal models of PD induced by the toxins 1-methyl-4-phenylpyridinium [43] and paraquat [44]. If PKC activation is also relevant for reserpine-induced Parkinsonism it is still unknown.

Regardless of the specific mechanism related to the prevention of behavioral alterations found in the present study, there is evidence that increased oxidative stress underlies the physiopathology of neurodegenerative diseases such as PD [45-48]. Further, clinical data suggest that neuroprotective treatments based on increasing antioxidant defenses are able to delay the progression of the pathology [49-56]. Thus, a neuroprotective intervention could be a relevant line of investigation in animal models of this disease. However, the usual acute pharmacological models include severe motor impairment upon a single injection of reserpine or specific neurotoxins [4,57-60]. This approach is not suitable for the investigation for testing neuroprotective interventions because they usually present a preventive and/or a neurodegeneration delaying profile. Further, most of the previous studies investigating the effects of vitamin E treatments on PD models did not investigate progressive behavioral deficits related to the clinical symptoms of the disease [33,35-37]. In this sense, the need for animal models of PD more compatible with clinical outcomes when investigating neuroprotective therapies has been pointed out. Thus, the present findings reinforce the idea that the protocol of progressive Parkinsonism induction with reserpine is suitable for investigating possible neuroprotective interventions in animal models of PD.

In conclusion, concomitant treatment with alpha-tocopherol prevents behavioral alterations induced by repeated reserpine. Although the antioxidant action of vitamin E is probably related, the exact mechanism underlying this preventive effect remains to be investigated. Finally, the progressive behavioral motor and cognitive alterations induced by repeated reserpine treatment seems an adequate protocol to investigate possible neuroprotective interventions for PD.

Acknowledgements

The authors would like to thank Antonio Carlos Queiroz de Aquino for capable technical assistance. This study was supported by grants from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, Brazil), Fundação de Amparo a Pesquisa do Estado do Rio Grande do Norte (FAPERN, Brazil), and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, Brazil).

References

  1. Henry JP, Sagné C, Botton D, Isambert MF, Gasnier B (1998) Molecular pharmacology of the vesicular monoamine transporter. Adv Pharmacol 42: 236-239.
  2. Dauer W, Przedborski S (2003) Parkinson's disease: mechanisms and models. Neuron 39: 889-909.
  3. Alves CS, Andreatini R, da Cunha C, Tufik S, Vital MA (2000) Phosphatidylserine reverses reserpine-induced amnesia. Eur J Pharmacol 404: 161-167.
  4. Colpaert FC (1987) Pharmacological characteristics of tremor, rigidity and hypokinesia induced by reserpine in rat. Neuropharmacology 26: 1431-1440.
  5. Skalisz LL, Beijamini V, Joca SL, Vital MA, Da Cunha C, et al. (2002) Evaluation of the face validity of reserpine administration as an animal model of depression--Parkinson's disease association. Prog Neuropsychopharmacol Biol Psychiatry 26: 879-883.
  6. Carvalho RC, Patti CC, Takatsu-Coleman AL, Kameda SR, Souza CF, et al. (2006) Effects of reserpine on the plus-maze discriminative avoidance task: dissociation between memory and motor impairments. Brain Res 1122: 179-183.
  7. Fernandes VS, Ribeiro AM, Melo TG, Godinho M, Barbosa FF, et al. (2008) Memory impairment induced by low doses of reserpine in rats: possible relationship with emotional processing deficits in Parkinson disease. Prog Neuropsychopharmacol Biol Psychiatry 32: 1479-1483.
  8. Santos JR, Cunha JA, Dierschnabel AL, Campêlo CL, Leão AH, et al. (2013) Cognitive, motor and tyrosine hydroxylase temporal impairment in a model of parkinsonism induced by reserpine. Behav Brain Res 253: 68-77.
  9. Fernandes VS, Santos JR, Leão AH, Medeiros AM, Melo TG, et al. (2012) Repeated treatment with a low dose of reserpine as a progressive model of Parkinson's disease. Behav Brain Res 231: 154-163.
  10. Abílio VC, Araujo CC, Bergamo M, Calvente PR, D’Almeida V, et al. (2003) Vitamin E attenuates reserpine-induced oral dyskinesia and striatal oxidized glutathione/reduced glutathione ratio (GSSG/GSH) enhancement in rats. Prog Neuro-Psychopharmacol Biological Psychiatry 27:109–114.
  11. Ebadi M, Srinivasan SK, Baxi MD (1996) Oxidative stress and antioxidant therapy in Parkinson's disease. Prog Neurobiol 48: 1-19.
  12. Bavarsad Shahripour R, Harrigan MR, Alexandrov AV (2014) N-acetylcysteine (NAC) in neurological disorders: mechanisms of action and therapeutic opportunities. Brain Behav 4: 108-122.
  13. Pérez-H J, Carrillo-S C, García E, Ruiz-Mar G, Pérez-Tamayo R, et al. (2014) Neuroprotective effect of silymarin in a MPTP mouse model of Parkinson's disease. Toxicology 319: 38-43.
  14. Halliwell B, Gutteridge JM (2007) Free Radicals in Biology and Medicine. Oxford University Press.
  15. Jiang Q (2014) Natural forms of vitamin E: metabolism, antioxidant, and anti-inflammatory activities and their role in disease prevention and therapy. Free Radic Biol Med 72: 76-90.
  16. Sanberg PR, Bunsey MD, Giordano M, Norman AB (1988) The catalepsy test: its ups and downs. Behav Neurosci 102: 748-759.
  17. de Lau LM, Breteler MM (2006) Epidemiology of Parkinson's disease. Lancet Neurol 5: 525-535.
  18. Duty S, Jenner P (2011) Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol 164: 1357-1391.
  19. Higginson CI, Wheelock VL, Carroll KE, Sigvardt KA (2005) Recognition memory in Parkinson's disease with and without dementia: evidence inconsistent with the retrieval deficit hypothesis. J Clin Exp Neuropsychol 27: 516-528.
  20. Lewis SJ, Dove A, Robbins TW, Barker RA, Owen AM (2003) Cognitive impairments in early Parkinson's disease are accompanied by reductions in activity in frontostriatal neural circuitry. J Neurosci 23: 6351-6356.
  21. Caudle WM, Colebrooke RE, Emson PC, Miller GW (2008) Altered vesicular dopamine storage in Parkinson's disease: a premature demise. Trends Neurosci 31: 303-308.
  22. Bergström T, Ersson C, Bergman J, Möller L (2012) Vitamins at physiological levels cause oxidation to the DNA nucleoside deoxyguanosine and to DNA--alone or in synergism with metals. Mutagenesis 27: 511-517.
  23. Osiecki M, Ghanavi P, Atkinson K, Nielsen LK, Doran MR (2010) The ascorbic acid paradox. Biochem Biophys Res Commun 400: 466-470.
  24. Palozza P, Calviello G, Serini S, Maggiano N, Lanza P, et al. (2001) Beta-carotene at high concentrations induces apoptosis by enhancing oxy-radical production in human adenocarcinoma cells. Free Radic Biol Med 30: 1000-1007.
  25. de Oliveira BF, Veloso CA, Nogueira-Machado JA, Martins Chaves M (2012) High doses of in vitro beta-carotene, alpha-tocopherol and ascorbic acid induce oxidative stress and secretion of IL-6 in peripheral blood mononuclear cells from healthy donors. Curr Aging Sci 5: 148-156.
  26. Winterbone MS, Sampson MJ, Saha S, Hughes JC, Hughes DA (2007) Pro-oxidant effect of alpha-tocopherol in patients with type 2 diabetes after an oral glucose tolerance test--a randomised controlled trial. Cardiovasc Diabetol 22: 6-8.
  27. Nadeem N, Woodside JV, Kelly S, Allister R, Young IS, et al. (2012) The two faces of α- and γ-tocopherols: an in vitro and ex vivo investigation into VLDL, LDL and HDL oxidation. J Nutr Biochem 23: 845-851.
  28. Reich EE, Montine KS, Gross MD, Roberts LJ 2nd, Swift LL, et al. (2001) Interactions between apolipoprotein E gene and dietary alpha-tocopherol influence cerebral oxidative damage in aged mice. J Neurosci 21: 5993-5999.
  29. Vatassery GT (1992) Vitamin E. Neurochemistry and implications for neurodegeneration in Parkinson's disease. Ann N Y Acad Sci 669: 97-109.
  30. Casani S, Gómez-Pastor R, Matallana E, Paricio N (2013) Antioxidant compound supplementation prevents oxidative damage in a Drosophila model of Parkinson's disease. Free Radic Biol Med 61: 151-160.
  31. Fariss MW, Zhang JG (2003) Vitamin E therapy in Parkinson's disease. Toxicology 189: 129-146.
  32. Miklya I, Knoll B, Knoll J (2003) A pharmacological analysis elucidating why, in contrast to (-)-deprenyl (selegiline), alpha-tocopherol was ineffective in the DATATOP study. Life Sci 72: 2641-2648.
  33. Butterfield DA, Castegna A, Drake J, Scapagnini G, Calabrese V (2002) Vitamin E and neurodegenerative disorders associated with oxidative stress. Nutr Neurosci 5: 229-239.
  34. Roghani M, Behzadi G (2001) Neuroprotective effect of vitamin E on the early model of Parkinson's disease in rat: behavioral and histochemical evidence. Brain Res 892: 211-217.
  35. Azzi A (2007) Molecular mechanism of alpha-tocopherol action. Free Radic Biol Med 43: 16-21.
  36. Azzi A, Gysin R, Kempná P, Munteanu A, Negis Y, et al. (2004) Vitamin E mediates cell signaling and regulation of gene expression. Ann N Y Acad Sci 1031: 86-95.
  37. Ferri P, Cecchini T, Ambrogini P, Betti M, Cuppini R, et al. (2006) alpha-Tocopherol affects neuronal plasticity in adult rat dentate gyrus: the possible role of PKCdelta. J Neurobiol 66: 793-810.
  38. Azzi A, Ricciarelli R, Zingg JM (2002) Non-antioxidant molecular functions of alpha-tocopherol (vitamin E). FEBS Lett 519: 8-10.
  39. Kanthasamy A, Jin H, Mehrotra S, Mishra R, Kanthasamy A, et al. (2010) Novel cell death signaling pathways in neurotoxicity models of dopaminergic degeneration: relevance to oxidative stress and neuroinflammation in Parkinson's disease. Neurotoxicology 31: 555-561.
  40. Chalimoniuk M, Stolecka A, Zieminska E, Stepien A, Langfort J, Strosznajder JB (2009) Involvement of multiple protein kinases in cPLA2 phosphorylation, arachidonic acid release, and cell death in in vivo and in vitro models of 1-methyl-4-phenylpyridinium-induced parkinsonism--the possible key role of PKG. J Neurochem 110: 307-317.
  41. Cristóvão AC, Barata J, Je G, Kim YS (2013) PKCδ mediates paraquat-induced Nox1 expression in dopaminergic neurons. Biochem Biophys Res Commun 437: 380-385.
  42. Beal MF (2002) Oxidatively modified proteins in aging and disease. Free Radic Biol Med 32: 797-803.
  43. Beal MF (2003) Mitochondria, oxidative damage, and inflammation in Parkinson's disease. Ann N Y Acad Sci 991: 120-131.
  44. Cadenas E, Davies KJ (2000) Mitochondrial free radical generation, oxidative stress, and aging. Free Radic Biol Med 29: 222-230.
  45. Younes-Mhenni S, Frih-Ayed M, Kerkeni A, Bost M, Chazot G (2007) Peripheral blood markers of oxidative stress in Parkinson's disease. Eur Neurol 58: 78-83.
  46. Abdel-Salam OM (2008) Drugs used to treat Parkinson's disease, present status and future directions. CNS Neurol Disord Drug Targets 7: 321-342.
  47. Beal MF (2009) Therapeutic approaches to mitochondrial dysfunction in Parkinson's disease. Parkinsonism Relat Disord 15 Suppl 3: S189-194.
  48. Chen JJ, Ly AV (2006) Rasagiline: A second-generation monoamine oxidase type-B inhibitor for the treatment of Parkinson's disease. Am J Health Syst Pharm 63: 915-928.
  49. De Araújo DP, Lobato Rde F, Cavalcanti JR, Sampaio LR, Araújo PV, et al. (2011) The contributions of antioxidant activity of lipoic acid in reducing neurogenerative progression of Parkinson's disease: a review. Int J Neurosci 121: 51-57.
  50. Magyar K, Pálfi M, Tábi T, Kalász H, Szende B, et al. (2004) Pharmacological aspects of (-)-deprenyl. Curr Med Chem 11: 2017-2031.
  51. Mayo JC, Sainz RM, Tan DX, Antolín I, Rodríguez C, et al. (2005) Melatonin and Parkinson's disease. Endocrine 27: 169-178.
  52. Weber CA, Ernst ME (2006) Antioxidants, supplements, and Parkinson's disease. Ann Pharmacother 40: 935-938.
  53. Weinreb O, Amit T, Bar-Am O, Youdim MB (2010) Rasagiline: a novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol 92: 330-344.
  54. Hsieh MH, Gu SL, Ho SC, Pawlak CR, Lin CL, et al. (2012) Effects of MK-801 on recognition and neurodegeneration in an MPTP-induced Parkinson's rat model. Behav Brain Res 229: 41-47.
  55. Marin C, Aguilar E (2011) In vivo 6-OHDA-induced neurodegeneration and nigral autophagic markers expression. Neurochem Int 58: 521-526.
  56. Salamone J, Baskin P (1996) Vacuous jaw movements induced by acute reserpine and low-dose apomorphine: possible model of parkinsonian tremor. Pharmacol Biochem Behav 53: 179-183.
  57. Salamone JD, Ishiwari K, Betz AJ, Farrar AM, Mingote SM et al. (2008) Dopamine/adenosine interactions related to locomotion and tremor in animal models: possible relevance to parkinsonism. Parkinsonism Relat Disord 14: S130-134.
  58. Tetrud JW, Langston JW (1989) MPTP-induced parkinsonism as a model for Parkinson's disease. Acta Neurol Scand Suppl 126: 35-40.
  59. Itoh N, Masuo Y, Yoshida Y, Cynshi O, Jishage K, et al. (2006) gamma-Tocopherol attenuates MPTP-induced dopamine loss more efficiently than alpha-tocopherol in mouse brain. Neurosci Lett 403: 136-140.
  60. Kamat CD, Gadal S, Mhatre M, Williamson KS, Pye QN, et al. (2008) Antioxidants in central nervous system diseases: preclinical promise and translational challenges. J Alzheimers Dis 15: 473-493.
Citation: Sarmento-Silva AJ, Lima RH, Cabral A, Meurer Y, Ribeiro AM, et al. (2014) Alpha-Tocopherol Counteracts Cognitive and Motor Deficits Induced by Repeated Treatment with Reserpine. Biochem Pharmacol (Los Angel) 4:153.

Copyright: © 2014 Sarmento-Silva AJ et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top