potential involvement of oxidative stress in induction of

16
Vol. 8(25), pp. 685-700, 8 July, 2014 DOI: 10.5897/AJPP2013.3963 Article Number: E4D4F4346171 ISSN 1996-0816 Copyright © 2014 Author(s) retain the copyright of this article http://www.academicjournals.org/AJPP African Journal of Pharmacy and Pharmacology Review Potential involvement of oxidative stress in induction of neurodegenerative diseases: Actions, mechanisms and neurotherapeutic potential of natural antioxidants George Laylson da Silva Oliveira 1 , Francisco Rodrigo de Asevedo Mendes de Oliveira 1 and Rivelilson Mendes de Freitas 1 * Pharmaceutical Sciences, Federal University of Piaui, Teresina, Piaui, 64049-550, Brazil. Received 1 December, 2013; Accepted 2 July, 2014 Several studies have shown that oxidative damage to various organs of a given organism is involved in causing various diseases, and in this context, the brain is highly vulnerable to oxidative stress because of its high metabolic rate, low capacity of cell regeneration, small amounts of enzymatic and non- enzymatic antioxidants. Oxidative stress is caused by an imbalance in cell redox state, in which there is excessive production of reactive oxygen species and/or failure of antioxidant defense systems. Consequently, oxidative stress in brain has shown direct implications in the pathogenesis of several neuro-degenerative diseases such as Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis. In this review, the critical role of oxidative stress in neuro-degenerative diseases as well as some aspects of antioxidant compounds (phenolic compounds and vitamins) regarding prevention and/or treatment of Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis were evaluated. Key words: Natural antioxidants, brain, neurodegenerative diseases, oxidative stress. INTRODUCTION Oxidative stress causes damage to various cellular processes in a deleterious manner, and consequently plays important roles in the development of many diseases, including those that affect the central nervous system such as neuro-degenerative diseases. Thus, many studies have aimed at researching new treatment strategies using antioxidants as a therapy for neuro- degenerative diseases (Choi et al., 2012; Johri and Beal, 2012; Jin et al., 2013a; Li et al., 2014; Deslauriers et al., 2014). This review reports a general view on oxidative stress in pathophysiology of neuro-degenerative diseases and also discussed the use of antioxidants of natural origin as neurotherapy in diseases such as Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis. This review was made based on a literature search using Science Direct (http://www.sciencedirect.com), Scopus (http://www.scopus.com), Pub Med (http://www.ncbi.nlm.nih.gov/pubmed), Web of Science (http://wokinfo.com) and SciFinder (http://cas.org/products/scifindr/index.html). The search was conducted until May 2014 using the following terms: *Corresponding author. E-mail: [email protected]. Tel: +55 86 3215 5870. Fax: +55 86 3215 5870. Author(s) agree that this article remain permanently open access under the terms of the Creative Commons Attribution License 4.0 International License

Upload: others

Post on 07-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Potential involvement of oxidative stress in induction of

   Vol. 8(25), pp. 685-700, 8 July, 2014 DOI: 10.5897/AJPP2013.3963 Article Number: E4D4F4346171 ISSN 1996-0816 Copyright © 2014 Author(s) retain the copyright of this article http://www.academicjournals.org/AJPP

African Journal of Pharmacy and Pharmacology

Review

Potential involvement of oxidative stress in induction of neurodegenerative diseases: Actions, mechanisms and

neurotherapeutic potential of natural antioxidants

George Laylson da Silva Oliveira1, Francisco Rodrigo de Asevedo Mendes de Oliveira1 and Rivelilson Mendes de Freitas1*

Pharmaceutical Sciences, Federal University of Piaui, Teresina, Piaui, 64049-550, Brazil.

Received 1 December, 2013; Accepted 2 July, 2014

Several studies have shown that oxidative damage to various organs of a given organism is involved in causing various diseases, and in this context, the brain is highly vulnerable to oxidative stress because of its high metabolic rate, low capacity of cell regeneration, small amounts of enzymatic and non-enzymatic antioxidants. Oxidative stress is caused by an imbalance in cell redox state, in which there is excessive production of reactive oxygen species and/or failure of antioxidant defense systems. Consequently, oxidative stress in brain has shown direct implications in the pathogenesis of several neuro-degenerative diseases such as Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis. In this review, the critical role of oxidative stress in neuro-degenerative diseases as well as some aspects of antioxidant compounds (phenolic compounds and vitamins) regarding prevention and/or treatment of Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis were evaluated. Key words: Natural antioxidants, brain, neurodegenerative diseases, oxidative stress.

INTRODUCTION Oxidative stress causes damage to various cellular processes in a deleterious manner, and consequently plays important roles in the development of many diseases, including those that affect the central nervous system such as neuro-degenerative diseases. Thus, many studies have aimed at researching new treatment strategies using antioxidants as a therapy for neuro-degenerative diseases (Choi et al., 2012; Johri and Beal, 2012; Jin et al., 2013a; Li et al., 2014; Deslauriers et al., 2014). This review reports a general view on oxidative stress in pathophysiology of neuro-degenerative diseases

and also discussed the use of antioxidants of natural origin as neurotherapy in diseases such as Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis.

This review was made based on a literature search using Science Direct (http://www.sciencedirect.com), Scopus (http://www.scopus.com), Pub Med (http://www.ncbi.nlm.nih.gov/pubmed), Web of Science (http://wokinfo.com) and SciFinder (http://cas.org/products/scifindr/index.html). The search was conducted until May 2014 using the following terms:

*Corresponding author. E-mail: [email protected]. Tel: +55 86 3215 5870. Fax: +55 86 3215 5870. Author(s) agree that this article remain permanently open access under the terms of the Creative Commons Attribution License 4.0 International License

Page 2: Potential involvement of oxidative stress in induction of

686 Afr. J. Pharm. Pharmacol. oxidative stress, reactive oxygen species, reactive nitrogen species, brain, antioxidants, neurodegenerative diseases, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Schizophrenia, Huntington's disease, phenolic compounds and vitamins. These words were also used in various combinations, being considered valid original research article, full text and that provided the search terms in the title and/or abstract. The duplicate articles in scientific databases were excluded and the remaining ones were evaluated by their eligibility regarding the inclusion and exclusion criteria, by reading and analyzing title and abstract. Articles in English published up to 2014 were analyzed. OXIDATIVE STRESS IN BRAIN The chemical substances that have one or more unpaired electrons are considered free radicals, which have as main characteristic that facility to donate their electrons to other molecules causing chain reactions and oxidative damage (Liochev, 2013). Free radicals and related molecules are classified as reactive oxygen species (ROS) and reactive nitrogen species (RNS) and a wide variety of these radicals are produced during normal metabolism in biological systems, which are counter balanced by cellular antioxidant mechanisms. However, the imbalance by excessive ROS and RNS and decreased antioxidant defense systems at cellular level cause oxidative stress (Figure 1), which can be induced to damage by peroxidation of cellular structures, protein oxidation, DNA damage and inhibition of electron transport chain in mitochondria (Dasuri et al., 2013).

ROS are represented by a group of chemicals that include the superoxide anion (O2

•-), hydroxyl (HO•), peroxyl (RO2

•), hydroperoxyl (HO2•), lipid hydroperoxide

(LOOH), hypochlorous acid (HClO), singlet oxygen (O2) and hydrogen peroxide (H2O2). RNS produced in cells are represented by nitric oxide (NO) and peroxynitrite (ONOO•) (Roberts et al., 2010; Koskenkorva-Frank et al., 2013). The superoxide anion, hydroxyl and hydrogen peroxide are the main ROS. Among these, the superoxide anion is formed due to reduction of molecular oxygen by various enzymatic systems (enzymes in the electron transport chain in mitochondria, xanthine oxidase, cyclooxygenase and NADPH-oxidase). In turn, the enzyme superoxide dismutase (SOD) converts the superoxide anion into molecules of hydrogen peroxide. Hydrogen peroxide in other reactions, in the presence of transition metals (iron and copper) is converted into a very reactive ROS, the hydroxyl radical (Milenkovic et al., 2013; Koskenkorva-Frank et al., 2013). One of the main mechanisms responsible by production of ROS in cells of the brain tissue is illustrated in Figure 2.

Several studies have considered that oxidative damage to various organs of a given organism is involved in the onset of various diseases. In this context, the brain is an organ highly vulnerable to oxidative stress due to its high

metabolic rate, which accounts for 20% of oxygen consumption (Sultana et al., 2013). In addition, the brain has low capacity for cellular regeneration, small amount of antioxidant enzymes (catalase, superoxide dismutase, glutathione peroxidase and glutathione reductase) and antioxidant non-enzymatic (reduced glutathione, -tocopherol and ascorbic acid) and as in any other tissue, oxidative stress in brain can damage the neurons by peroxidation of poly-unsaturated fatty acids (arachidonic acid and docosahexaenoic acid) in cell membrane, oxidative damage to DNA, RNA, proteins and induction of apoptosis (Figure 3) (Nunomura et al., 2012; Smith et al., 2013).

Peroxidation of the cell membrane has as consequence the increase of stiffness, decreased activity of membrane bound enzymes, deficiency of membrane receptors and alters the permeability. ROS and RNS can also attack directly the proteins of the lipid membrane and cause fragmentation of proteins, cross-links between proteins and the formation of carbonyl, which contributes to the integrity of the cell membrane and interferes with various cellular processes. ROS and RNS can still attack the nucleic acids causing the crosslinking of the protein-DNA, modification of DNA bases, breakage and mutations in the DNA chain (Sultana et al., 2013). OXIDATIVE STRESS AND NEURODEGENERATIVE DISEASES Oxidative stress and Alzheimer's diseases Alzheimer's disease (AD) is clinically characterized by progressive memory loss and severe cognitive function decline. The accumulation of extracellular amyloid protein deposited senile plaques and intracellular neurofibrillary tangles made of abnormal and hyper-phosphorylated tau protein, regionalized neuronal death, loss of synaptic connections in selective brain regions, proliferation of astrocytes and microglia activation, and define neuro-pathologically the AD. In addition, the oxidative stress and mitochondrial dysfunction are related to the development of AD (Gubandru et al., 2013).

Mitochondria are responsible for ATP production, oxidative phosphorylation, cell cycle control, cell growth and apoptosis (Chaturvedi and Beal, 2013). Thus, a problem in energy metabolism contributes to reduced production of ATP, beyond the generation of excessive ROS, such as superoxide, hydroxyl radicals and hydrogen peroxide. The literature describes the mitochondrial damage induced by ROS and vascular hyperfusion as key initiators for the development of AD (Butterfield et al., 2006; Aliev et al., 2010; Massaad et al., 2009; Parihar and Brewer, 2007; Kovacic and Somanathan, 2012; Schrag et al., 2013).

Indeed, oxidative imbalance and significant increase of its bio-products have been consistently reported in AD. Thus, the products generated from oxidized biomolecules

Page 3: Potential involvement of oxidative stress in induction of

Oliveira et al. 687

Figure 1. Representation of oxidative stress.

Figure 2. Representation of ROS/RNS in cells of nervous tissue by reduction of molecular oxygen to water in mitochondrial electron transport chain, which consists of four multimeric complexes (I, II, III and IV) and two conveyors of electrons (Coenzyme Q and Cytochrome C). Complexes I and III are the major sources of production of free radicals.

Page 4: Potential involvement of oxidative stress in induction of

688 Afr. J. Pharm. Pharmacol.

Figure 3. Oxidative damage caused by ROS and RNS to various biomolecules.

are stable and used as markers of ROS. Moreover, exogenous antioxidants and antioxidant enzymes can indirectly measure the levels of ROS. In that sense, in lipid peroxidation, significant levels of reactive aldehydes may be evidenced, including 4-hydroxynonal, malondialdehyde (MDA), and 2-propenal (acrolein) and are chemically and metabolically stable iso-prostanoids including F2-isoprostanes and F4-neuroprostanes (Wang et al., 2013a; Gubandru et al., 2013; Sultana et al., 2013). Several investigations show that in the oxidation of proteins, the most investigated markers are carbonyls and 3-nitrotyrosine, final product of the interaction of peroxy nitrite with tyrosine residues (Aksenov et al., 2001; Castegna et al., 2002; Good et al., 1996; Tohgi et al., 1999; Butterfield et al., 2001; Castegna et al., 2003; Reed et al., 2009).

In the oxidation of the genetic material (DNA/RNA), the

hydroxyl deoxyguanosine 8-(8-OHdG) and 8-hydroxyguanosine (8-OHG) are the main markers, which are the products of oxidation of guanine (Mecocci et al., 1994; Nunomura et al., 1999; Shan et al., 2003; Ding et al., 2005; Honda et al., 2005). Besides the increase these toxic products of biomolecules, significant reduction of antioxidants and antioxidant enzymes can be verified. Study reveals that plasmatic levels of antioxidants such as albumin, bilirubin, uric acid, lycopene, vitamin A, vitamin C and vitamin E are decreased in patients with AD. Similar results can be shown with antioxidant enzymes such as superoxide dismutase, catalase, glutathione peroxidase and hemeoxigenase, in different brain regions (Wang et al., 2013; Foy et al., 1999; Kim et al., 2006). Despite continued efforts, current therapeutic strategies are limited to those that soften the symptoms without impeding the progress of the disease. Thus, there

Page 5: Potential involvement of oxidative stress in induction of

is growing demand for a treatment capable of protecting neurons and inhibiting oxidative damage, may be an effective alternative evidenced in natural antioxidants (Bonda et al., 2010). Oxidative stress and Parkinson's diseases Parkinson's disease (PD) is a chronic neurological progressive disease and associated with a loss of dopaminergic neurons in the compact part of the substantia nigra (SN), clinically characterized by cardinal symptoms, resting tremor, rigidity, brady kinesia and postural instability. In addition to dopamine depletion, PD is neuropathologically defined by the deposition of Lewis bodies and Lewis neurites in vulnerable neurons. The etio-pathogenesis of PD is still not fully understood. In most cases the disease is eventual: a multifactorial idiopathic disease, seems to arise from a combination of genetic susceptibility and environmental exposures (Perfeito et al., 2013; Dickson et al., 2009; Halliday et al., 2011; Hauser and Hastings, 2013).

Some environmental toxins are involved in the process of oxidative stress, including paraquat, rotetone, maneb and MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). The MPTP, for example, crosses the blood-brain barrier due to their lipophilicity and is oxidized to a toxic substance, MPP+ (1-methyl-4-phenylpyridinium) by monoamine oxidase in glial cells. MPP+ is accumulated in mitochondria of dopaminergic neurons. The MPP+ inhibits mitochondrial complex I, causing break in electron transport chain and as a result the reduction in ATP production and increased production of ROS (Subramaniam and Chesselet, 2013; Chen and Le, 2006).

Moreover, evidence shows an association between oxidative stress and mitochondrial DNA (mtDNA) mutations with the onset of PD. Associated with constant exposure to ROS, the lack of protection by histones, and limited DNA repair mechanisms, make mtDNA particu-larly vulnerable to oxidative damage, which can lead to harmful mutations. As a result, a positive feedback is generated by the accumulation of these mutations, which can gradually cause a reduction in the efficiency of the electron transport chain (ETC) stimulating decreased ATP production and increased production of ROS, followed by eventual cell death (Yan et al., 2013; Sanders and Greenamyre, 2013).

In addition, ROS can irreversibly damage protein, for example, amino groups in side chains of specific amino acids can be modified to carbonyl groups (Greenamyre and Sanders, 2013). It recently shows an association be-tween elevated levels of protein carbonyls and increased oxidative stress (Hyun et al., 2002). Another study showed damage of mitochondrial complex I subunit in PD brains related to high levels of protein carbonyl (Keeney et al., 2006).

Oliveira et al. 689

Similarly, the oxidized lipids can exercise deleterious effects on neuronal function and lead to PD. In general, lipids involved in membrane fluidity and permeability, can store energy that participate in inflammatory processes and in apoptosis signaling. The poly-unsaturated fatty acids are more prone to lipid peroxidation, particularly in the brain there is a large amount of two of these, the arachidonic acid and docosahexaenoic acid. After the adipose tissue, the organ with the highest lipid content is the brain and thus, it is very prone to lipid peroxidation (Ruipérez et al., 2010; Bochkov et al., 2002; Roberts II and Fessel, 2004; Chen et al., 2008). Evidence supports an association between lipid peroxidation and PD (Hyun et al., 2002; Hoepken et al., 2007; Lee et al., 2001). Oxidative stress and Huntington's diseases Huntington disease (HD) is a progressive neuro-degenerative disorder characterized by presence of emotional and movement disorders and dementia (Ayala-Peña, 2013). HD has autosomal dominant transmission with the mutant gene termed IT15, responsible for the disease located on chromosome 4. The mutation respon-sible by this disease consists in an excessive number of abnormal tri-nucleotide cytosine-adenine-guanosine (CAG). The mutant gene encodes a protein called huntingtin, which interacts with several proteins that are involved in transcription, cell signaling and intracellular transport. As a result of repeated CAG, may be polyglutamine expansion located near the N-terminus of protein molecule. The expanded poly-glutamine chains lead to fragmentation of protein, which tends to accumu-late inside the neuron. The aggregation of protein fragments causes changes in neuronal functioning and possibly plays a role in neuronal death process (Goldberg et al., 1994; DiMauro and Schon, 2008; Harjes and Wanker, 2003; Cattaneo et al., 2005; Ha et al., 2012).

It is known that oxidative stress is mediated by increased ROS, including superoxide, hydrogen peroxide and hydroxyl radical. These ROS impair cell function by degrading proteins, lipids and nucleic acids and consequently, there is a vicious cycle of mitochondrial oxidative damage in Huntington's diseases. In context of mutant Huntington expression, the ROS overproduction causes accumulation and exhaustion of mtDNA damage and reduced mitochondrial bioenergetics. Simultaneously, the mutant Huntington affects mtDNA repair, avoiding the accumulation of human apurinic/ apyrimidinic endonuclease (APE1) in mitochondria, which further contributes to mitochondrial damage. As a result, the mtDNA damage leads to exacerbated mitochondrial dysfunction and possible neuro-degenerative diseases. In addition, oxidative damage to nuclear DNA also contributes to mitochondrial dysfunction and contributes to activation of DNA repair processes that may lead to expansion of the CAG repeat (Mochel and Haller, 2011;

Page 6: Potential involvement of oxidative stress in induction of

690 Afr. J. Pharm. Pharmacol. Johri and Beal, 2012; Ayala-Peña, 2013). Furthermore, oxidative modification of proteins (protein carbonyl) and lipids (malondialdehyde and 4-hydroxynonenal) are also increased in HD in the brain (Beal and Browne, 2006).

Relevant characteristics that support the relationship between oxidative stress and HD, can be evidenced by accumulation of several markers, such as lipofuscin derived from peroxidation of unsaturated fatty acids, 3-nitrotyrosine acids (nitrated protein) as well as 8-hydroxy-2-deoxyguanosine (OH8dG) in mtDNA (Dhillon and Fenech, 2013). Oxidative stress and Schizophrenia Schizophrenia is a chronic debilitating neuro-degenerative disorder and is characterized by several positive symptoms such as delusions, hallucinations, disorganized speech and negative symptoms, including deficits in cognitive and social capacity and blunted affect (Yao and Keshavan, 2011). The initial pathophysiology of schizophrenia collaborates to increase the generation of reactive oxygen species in the brain (Bitanihirwe and Woo, 2011). In addition, genetic evidence demonstrates that decreased capacity of synthesizing antioxidant enzymes, such as glutathione (GSH) under conditions of oxidative stress favor the appearance of schizophrenia (Gysin et al., 2007; From et al., 2009). As previously discussed, excessive ROS cause damage to important macromolecules such as DNA, proteins and lipids. It can affect the genetic material by modifying gene expression, protein oxidation, making them non-functional and peroxidative damage in lipids, which results in damage to cell membrane and cell organelles (Wu et al., 2013). Additionally, dopamine, an auto-oxidizable neuro-transmitter, has a dihydroquinone structure that can be oxidized by molecular oxygen to form hydrogen peroxide and o-quinone. Thus, the catecholamines oxidation combined to deficient antioxidant system in the brain, produces an excess of free radicals resulting in oxidative stress (Bošković et al., 2011). Oxidative stress and amyotrophic lateral sclerosis Amyotrophic lateral sclerosis (ALS) is one of the major neurodegenerative disorders regarding the neuromuscular junction. The disease is characterized by selective degeneration of upper motor neurons (corticospinal) and lower (spinal and bulbar), which results in pathological mis-communication between nerve and muscle (Pansarasa et al., 2013). The majority of ALS cases are of unknown etiology (sporadic). However, the identification of mutant genes that predispose to these disorders has provided the means to better understand its pathogenesis. It is known that this condition occurs in a change mutant gene that encodes Cu/Zn superoxide

dismutase type 1 (SOD1), enzyme that removes free radicals and protects cells (motor neurons) from oxidative stress (Halter et al., 2010; Rosen et al., 1993). Studies have shown that SOD1 in its mutant version can cause mitochondrial dysfunction. The accumulation of SOD1 aggregates in membrane of mitochondria can cause blockage of proteins import, excessive ROS production and eventual cell death by apoptosis (Vijayvergiya et al., 2005; Mattiazzi et al., 2002; Takeuchi et al., 2002). NATURAL ANTIOXIDANTS WITH NEUROTHERAPEUTIC POTENTIAL Recently, natural antioxidants have gained great importance in treatment or prevention of diseases such as cancer, diabetes, cardiovascular and mainly neuro-degenerative disorders. Scientific studies have already developed and discussed in preceding paragraphs demonstrate that the generation of ROS and RNS have direct implications in neurodegenerative diseases such as Alzheimer's, Parkinson's, amyotrophic lateral sclerosis and Huntington's disease. Thus, there is great interest in studies aiming to investigate the therapeutic potential of natural antioxidants for preventing or controlling oxidative stress in neurodegenerative diseases, since it is known that brain tissue is vulnerable to oxidative damage by having abundant lipid content and relative scarcity of antioxidant enzymes when compared with other tissues (Figure 4).

Antioxidants such as vitamins, phenolic compounds and flavonoids have been extensively investigated as potential therapeutic agents in vitro and in vivo for prevention of neurodegenerative diseases (Ebrahimi and Schluesener, 2012; Choi et al., 2012). Most of these antioxidant compounds can be found in fruits, vegetables, plants or synthesized, and the antioxidant mechanisms can act by induction of gene expression of endogenous antioxidant defense systems (activation of nuclear factor kappa B, NF-kB), regulation of ROS and RNS, interaction with oxidative pathways and the capacity to chelate metal ions responsible for the formation of free radicals (Dajas, 2012) (Figure 5). VITAMINS Vitamins C, A, E and B6 shown in Figure 6, are substances with high capacity to remove ROS and RNS in vitro and in vivo studies, and therefore can act in prevention of oxidative stress and possible treatment for neurodegenerative diseases such as Alzheimer's and Parkinson's disease (Table 1). For example, studies performed by Ahmed (2012) in mice provided important evidence that dietary supplementation with vitamins slows the progression of neurodegenerative diseases like Alzheimer's.

Page 7: Potential involvement of oxidative stress in induction of

Oliveira et al. 691

Figure 4. Number of publications addressing antioxidants and neurodegenerative diseases, published between 2003 and 2013. Source: Pubmed and Science Direct.

Figure 5. Main antioxidant mechanisms against ROS/RNS.SOD (superoxide dismutase), CAT (catalase), GPx (glutathione peroxidase).

Vitamins C and E are natural compounds well known for their high antioxidant capacity in in vitro and in vivo experimental models. Once there is an involvement of ROS and RNS in neurodegenerative diseases, vitamins C and E have been considered as potential therapeutic agents for neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's and amyotrophic lateral sclerosis (Heo et al., 2013). For example, Miyake et al. (2011) investigated the relationship between intake

of antioxidant vitamins and risk of Parkinson's disease in Japan using data from a case-control study of hospital-based multicenter.This study included 249 patients with Parkinson's disease and results indicated that a higher intake of vitamin E might be associated with a reduced risk of Parkinson's disease. In another epidemiological study involving 6 patients bearer of Alzheimer's disease, Morris et al. (2005) demonstrated that ingestion of vitamin E could contribute to preventing the neurodegenerative

Page 8: Potential involvement of oxidative stress in induction of

692 Afr. J. Pharm. Pharmacol.

Figure 6. Chemical structures of vitamins C, B6, E and A.

disease.

Even considering the pharmacological potential of Vitamin C, there are conflicting scientific studies on its potential in treatment of various neurodegenerative diseases, including Alzheimer's disease. For example, in the study of Arlt et al. (2012) involving the 12 patients with Alzheimer's disease, was observed that supplemen-tation with vitamin C did not have significant effect on the prevention of Alzheimer's disease over 1 year. In another study involving 57 patients with Alzheimer's disease and treated with vitamin E, it has been observed that the oxidative stress was not inhibited when it was considering the oxidized glutathione levels (Lloret et al., 2009).

Vitamins addressed in Figure 6 are part of human diet and are considered substances with high antioxidant capacity. Thus, these antioxidant properties have attrac-ted great attention for the treatment of neurodegenerative diseases, but studies to date are contradictory regarding the therapeutic potential for neurodegenerative diseases. PHENOLIC COMPOUNDS Phenolic compounds have one or more hydroxyl groups bonded to a benzene ring, being chemical constituents found in a wide variety of plants and which are represented by a variety of classes of compounds that can be divided according to their chemical structures. Among these classes, there are phenolic acids (hydroxybenzoic acids (C6-C1) and hydroxycinnamic acids (C6-C3), flavonoids (C6-C3-C6, which includes antho-cyanins, flavonols, flavones, flavanones and isoflavones), stilbenes (C6-C2-C6), lignans (C6-C3-C6-C3) and curcuminoids (C3-C6-C3-C1-C6)) (Figure 7) (Cheynier et al.,

2013). Antioxidant capacity of phenolic compounds occurs

primarily by elimination of free radicals, chelation capacity and modulation of enzymes, as well as their effects on cell signaling pathways and gene expression, being these mechanisms dependent on the chemical characteristics of the compounds (De Mello and Fasolo, 2014). Animal studies, clinical and epidemiological, support the critical role of phenolic compounds in the prevention and treatment of various diseases such as neurodegenerative disorders (Table 2).

Among the studies of phenolic compounds approached in Figure 7, quercetin (flavonoid) has been shown to be promising in treating various neurodegenerative diseases (Jazvinšćak et al., 2012; Pandey et al., 2012; Denny Joseph and Muralidhara, 2013). In a model of induced Huntington's disease in rats, treatment with quercetin at a dose of 25 mg/kg was able to reverse the inhibition of mitochondrial electron transport chain, restore ATP levels, prevent the mitochondrial dysfunction by inhibiting oxidative stress and increase the SOD and CAT activities (Sandhir and Mehrotra, 2013). In other in vitro and in vivo studies, quercetin has demonstrated a role in therapeutic strategies to treatment of neurodegenerative diseases in clinical settings to provide neuroprotection related to suppression of oxidative stress, improvement in behavioral function, reduction in infarct volume, cerebral edema and cell injury. In contrast, some studies contra-dict the neuroprotective potential of quercetin and other flavonoids (Ossola et al., 2009; Huebbe et al., 2010).

In another study, Mansouri et al. (2013) examined the neuroprotective effect of gallic acid (phenolic acid) at a dose of 30 mg/kg (orally, once daily for 26 days) on cog-nitive changes and oxidative stress in the brain induced

Page 9: Potential involvement of oxidative stress in induction of

Oliveira et al. 693

Figure 7. Chemical structures of classes of phenolic compounds.

Page 10: Potential involvement of oxidative stress in induction of

694 Afr. J. Pharm. Pharmacol.

Table 1. Main vitamins with therapeutic potential for the treatment of neurodegenerative diseases.

Vitamin Neurodegenerative diseases Mechanisms involved References

1

Alzheimer's disease Parkinson's disease Huntington's disease Amyotrophic lateral sclerosis Schizophrenia

Elimination of ROS Chelating activity Lipid peroxidation inhibition Inhibition of amyloid-β (Aβ) aggregation

Ballaz et al. (2013), Heo et al. (2013), Engelhart et al. (2002), Kontush et al. (2001), Li et al. (2012), Quinn et al. (2003), Montilla-López et al. (2002), De Oliveira et al. (2012), Morales et al. (1989), Rebec et al. (2006), Heiser et al. (2010), Sivrioglu et al. (2007), Dadheech et al. (2006), Dakhale et al. (2005), and Arvindakshan et al. (2003)

2 Alzheimer's disease

Elimination of ROS Chelating activity

Hashim et al. (2011) and Nassiri-asl et al. (2012);

3

Alzheimer's disease Parkinson's disease Huntington's disease Schizophrenia

Elimination of ROS Regulation of antioxidant enzymes Lipid peroxidation inhibition

Morrissey and Hill (2011), Ricciarelli et al. (2007), Ahmed (2012), Morris et al. (2005), Viña et al. (2004), Guan et al. (2012), Kontush and Schekatolina (2004); Kalonia et al. (2010), Peyser et al. (1995), Rao et al. (2003), Dadheech et al. (2006), Sivrioglu et al. (2007) and Arvindakshan et al. (2003)

4 Alzheimer's disease Parkinson's disease

Elimination of ROS/RNS Chelating activity Inhibition of amyloid-β (Aβ) aggregation

Lee et al. (2009), Ono et al. (2004), Ono and Yamada (2012), Gackowski et al. (2008), Sutachan et al. (2012), King et al. (1992) and Rao et al. (2003)

Table 2. Phenolic compounds with therapeutic potential for the treatment of neurodegenerative diseases.

Phenolic compounds Neurodegenerative diseases Mechanisms involved References

1 Alzheimer's, Parkinson's, Huntington's and Schizophrenia

Elimination of ROS, chelating activity; Regulation of antioxidant enzymes; Lipid peroxidation inhibition; Inhibition of amyloid-β (Aβ) aggregation; Attenuation of deficits in motor coordination.

Liu et al. (2013), Sandhir and Mehrotra (2013), Zhu et al. (2013), Huebbe et al. (2010), Ansari et al. (2009), Lavoie et al. (2009), and Chakrabortyet al. (2013)

2 Alzheimer's and Parkinson's Elimination of ROS; Lipid peroxidation inhibition; Inhibition of amyloid-β (Aβ) aggregation

Wang et al. (2012), Javed et al. (2012), Moshahid et al. (2012), and Islam et al. (2012)

3 Alzheimer's, Parkinson's, Huntington's and Amyotrophic lateral sclerosis

Elimination of ROS, Inhibition of amyloid-β (Aβ); Modulation of cell signaling pathways

Zhang et al. (2013), Lee et al. (2013), He et al. (2012), Levites et al. (2001), Kim et al. (2010b), Avramovich-Tirosh et al. (2007), Ehrnhoefer et al. (2006), Xu et al. (2006), and Koh et al. (2006)

4 Alzheimer's, Parkinson's, and Schizophrenia

Elimination of ROS; Inhibition of amyloid-β (Aβ) aggregation; Inhibition of the aggregation of tau proteins; Lipid peroxidation inhibition.

George et al. (2013), Ejaz Ahmed et al. (2013), Teixeira et al. (2013), Dietrich-Muszalska et al. (2012), and Lim et al. (2013)

5 Alzheimer's and Parkinson's Elimination of ROS; Regulation of antioxidant enzymes; Inhibition of amyloid-β (Aβ) aggregation

Hamaguchi et al. (2009), Detoma et al. (2011), Zhang et al. (2011), and Molina et al. (2004)

Page 11: Potential involvement of oxidative stress in induction of

Oliveira et al. 695

Table 2. Contd.

6 Alzheimer's, Parkinson's, and Amyotrophic lateral sclerosis

Elimination of ROS; Inhibition of amyloid-β (Aβ) aggregation

Bagheri et al. (2011), Baluchne et al. (2009), Liu et al. (2008), Trieu and Uckun (1999)

7 Alzheimer's Elimination of ROS, chelating activity; Regulation of antioxidant enzymes; Inhibition of amyloid-β (Aβ) aggregation

Zhao et al. (2013a, b)

8 Alzheimer's and Parkinson's Elimination of ROS; Lipid peroxidation inhibition; Inhibition of amyloid-β (Aβ) aggregation

Heo et al. (2004), Khan et al. (2012), Ma et al. (2013), Zbarsky et al. (2005), and Sonia et al. (2013)

9 Alzheimer's, Parkinson's and Huntington's

Elimination of ROS; Inhibition of amyloid-β (Aβ) aggregation

Kim et al. (2010a), Sharoar et al. (2012), Filomeni et al., (2012), Li and Pu (2011), and Lagoa et al. (2009)

10 Alzheimer's, Parkinson's and Amyotrophic lateral sclerosis

Elimination of ROS, chelating activity; Lipid peroxidation inhibition; Inhibition of amyloid-β (Aβ) aggregation

Barros et al. (2013), Huang et al. (2013a), and Fontanilla et al. (2011)

11 Alzheimer's Elimination of ROS, chelating activity; Regulation of antioxidant enzymes; Inhibition of amyloid-β (Aβ) aggregation

Zhao et al. (2013a, b)

12 Alzheimer's Elimination of ROS; Inhibition of amyloid-β (Aβ) aggregation

Mori et al. (2013) and Pi et al. (2012)

13 Alzheimer's, Parkinson's, Huntington's and Schizophrenia

Elimination of ROS; chelating activity; Inhibition of amyloid-β (Aβ) aggregation

Wang et al. (2013b), Huang et al. (2013b), Li et al. (2013), Yu et al. (2013), Agrawal et al. (2012), Du et al. (2012), Mansouri et al. (2012), Sandhir et al. (2013), Hickey et al. (2012), Bishnoi et al. (2008) and Lavoie et al. (2009)

14 Alzheimer's, Parkinson's, Huntington's and Amyotrophic lateral sclerosis

Elimination of ROS; Inhibition of amyloid-β (Aβ) aggregation; Regulation of antioxidant enzymes; Chelating activity, regulating of expression of SIRT1

Khan et al. (2010), Jin et al. (2008b), Feng et al. (2013), Porquet et al. (2013), Ho et al. (2010), Wang et al. (2011), and Kim et al. (2007)

by streptozotocin (STZ) (model used to study the Alzheimer's disease) in rats and the results showed that the gallic acid attenuated the cognitive deficits, increased levels of enzymatic antioxidants (GPx, CAT and SOD) and inhibited lipid peroxidation by the reduction of the MDA levels in brain tissue.

Another substance with great therapeutic potential for treatment of neurodegenerative diseases is resveratrol (Figure 7), a phenolic non-flavonoid compound found in grapes and red

wine. This antioxidant compound has been investigated in different in vitro and in vivo studies to treatment of Parkinson's disease, Alzheimer's, Huntington's and amyotrophic lateral sclerosis. In a model of Parkinson's disease induced in rats, it was observed that resveratrol (20 mg/kg) in dopaminergic neurons showed neuroprotective effect by regulating glutathione, glutathione peroxidase, glutathione reductase, catalase, superoxide dismutase and decreasing levels of thiobarbituric acid reactive substances (TBARS).

Furthermore, it was observed decreased carbonyl protein levels, phospholipase A2 activity and improvement in content of dopamine (Khan et al., 2010). Resveratrol also showed neuroprotective effects in a cellular model of amyotrophic lateral sclerosis and in a model of Alzheimer's disease in mice, being observed in the hippocampus a significant increase of neuronal survival (Kim et al., de 2007).

Confirming the important role of phenolic compounds in complementary therapy and/or

Page 12: Potential involvement of oxidative stress in induction of

696 Afr. J. Pharm. Pharmacol. prevention of neurodegenerative diseases, the results of Bournival et al. (2009) have clearly demonstrated that resveratrol and quercetin are compounds that effectively protect neurons against apoptotic cascade induced by oxidative stress.

Most of the works discussed in this article are mainly supported by data obtained in animal models and thus more studies regarding the therapeutic potential for human health are needed to unravel the role of these antioxidant compounds, which are becoming increasingly important as alternative or complementary therapies to neurodegenerative diseases (Figure 4). CONCLUSION Based on the literature, there is good evidence that oxidative stress plays an important role in the pathogenesis of neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, Schizophrenia and amyotrophic lateral sclerosis. In this context, natural antioxidant compounds with different chemical structures that neutralize the damaging effects of ROS have been investigated in preventing neurodegenerative diseases. Most of the items discussed in this review demonstrated the efficacy of antioxidant compounds in treatment of neurodegenerative disorders using animal models or in small clinical studies. However, some studies showed contradictory results regarding the therapeutic potential of antioxidants of natural origin. Thus, new studies approa-ching clinical applications of antioxidants in the treatment or prevention of neurodegenerative diseases are needed. Conflict of interest The author(s) declare(s) that they have no conflicts of interest to disclose. REFERENCES Agrawal SS, Gullaiya S, Dubey V, Singh V, Kumar A, Nagar A, Tiwari P

(2012). Neurodegenerative Shielding by Curcumin and Its Derivatives on Brain Lesions Induced by 6-OHDA Model of Parkinson's Disease in Albino Wistar Rats. Cardiovasc. Psychiatry. Neurol. 1-8.

Ahmed HH (2012). Modulatory effects of vitamin E, acetyl-l-carnitine and α-lipoic acid on new potential biomarkers for Alzheimer's disease in rat model. Exp. Toxicol. Pathol. 64:549-556.

Aksenov MY, Aksenova MV, Butterfield DA, Geddes JW, Markesbery WR (2001). Protein oxidation in the brain in Alzheimer's disease. Neuroscience 103:373-383.

Aliev G, Palacios HH, Gasimov E, Obrenovich ME, Morales L, Leszak J, Bragin V, Herrera AS, Gokhman D (2010). Oxidative stress induced mitochondrial failure and vascular hyperfusion as a key initiator for the development of Alzheimer’s disease. Pharmaceuticals 3:158-187.

Ansari MA, Abdul HM, Joshi G, Opii WO, Butterfield DA (2009). Protective effect of quercetin in primary neurons against Aβ(1-42): relevance to Alzheimer's disease. J. Nutr. Biochem. 20:269-275.

Arlt S, Müller-Thomsen T, Beisiegel U, Kontush A (2012). Effect of One-Year Vitamin C- and E-Supplementation on Cerebrospinal Fluid Oxidation Parameters and Clinical Course in Alzheimer’s Disease.

Neurochem. Res. 37:2706-2714. Arvindakshan M, Ghate M, Ranjekar PK, Evans DR, Mahadik SP

(2003). Supplementation with a combination of ω-3 fatty acids and antioxidants (vitamins E and C) improves the outcome of schizophrenia. Schizophr. Res. 62:195-204.

Ayala-Peña S (2013). Role of oxidative DNA damage in mitochondrial dysfunction and Huntington’s disease pathogenesis. Free. Rad. Biol. Med. 62:102-110.

Bagheri M, Joghataei MT, Mohseni S, Roghani M (2011). Genistein ameliorates learning and memory deficits in amyloid β(1-40) rat model of Alzheimer’s disease. Neurobiol. Learn. Mem. 95:270-276.

Ballaz S, Morales I, Rodríguez M, Obeso JA (2013). Ascorbate prevents cell death from prolonged exposure to glutamate in an in vitro model of human dopaminergic neurons. J. Neurosci. Res. 91:1609-1617.

Barros Silva R, Santos NAG, Martins NM, Ferreira DAS, Barbosa Jr F, Oliveira Souza VC, Kinoshita Â, Baffa O, Del-Bel E, Santos AC (2013). Caffeic acid phenethyl ester protects against the dopaminergic neuronal loss induced by 6-hydroxydopamine in rats. Neuroscience 233:86-94.

Bishnoi M, Chopra K, Kulkarni SK (2008). Protective effect of Curcumin, the active principle of turmeric (Curcuma longa) in haloperidol-induced orofacial dyskinesia and associated behavioural, biochemical and neurochemical changes in rat brain. Pharmacol. Biochem. Behav. 88:511-522.

Bitanihirwe BKY, Woo TUW (2011). Oxidative stress in schizophrenia: An integrated approach. Neurosci. Biobehav. Rev. 35:878-893.

Bochkov VN, Kadl A, Huber J, Gruber F, Binder BR, Leitinger N (2002). Protective role of phospholipid oxidation products in endotoxin-induced tissue damage. Nature 419:77-81.

Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M, Zhu X, Smith MA (2010). Oxidative stress in Alzheimer disease: A possibility for prevention. Neuropharmacology 59:290-294.

Bošković M, Vovk T, Plesničar BK, Grabnar I (2011). Oxidative stress in schizophrenia. Curr. Neuropharmacol. 9:301-312.

Bournival J, Quessy P, Martinoli MG (2009). Protective Effects of Resveratrol and Quercetin Against MPP+ -Induced Oxidative Stress Act by Modulating Markers of Apoptotic Death in Dopaminergic Neurons. Cell. Mol. Neurobiol. 29:1169-1180.

Browne SE, Beal MF (2006). Oxidative damage in Huntington’s disease pathogenesis. Antioxid. Redox Signal 8:2061-2073.

Butterfield DA, Kanski J (2001). Brain protein oxidation in age-related neurodegenerative disorders that are associated with aggregated proteins. Mech. Ageing Dev. 122:945-962.

Butterfield DA, Perluigi M, Sultana R (2006). Oxidative stress in Alzheimer’s disease brain: new insights from redox proteomics. Eur. J. Pharmacol. 545:39-50.

Castegna A, Aksenov M, Thongboonkerd V, Klein JB, Pierce WM, Booze R, Markesbery WR, Butterfield DA (2002). Proteomic identification of oxidatively modified proteins in Alzheimer's disease brain. Part II: dihydropyrimidinase-related protein 2, alpha-enolase and heat shock cognate 71. J. Neurochem. 82:1524-1532.

Castegna A, Thongboonkerd V, Klein JB, Lynn B, Markesbery WR, Butterfield DA (2003). Proteomic identification of nitrated proteins in Alzheimer's disease brain. J. Neurochem. 85:1394-1401.

Cattaneo E, Zuccato C, Tartari M (2005). Normal Huntington function: an alternative approach to Huntington's disease. Nat. Rev. Neurosci. 6:919-30.

Chakraborty J, Singh R, Dutta D, Naskar A, Rajamma U, Mohanakumar KP (2013). Quercetin Improves Behavioral Deficiencies, Restores Astrocytes and Microglia, and Reduces Serotonin Metabolism in 3-Nitropropionic Acid-Induced Rat Model of Huntington's Disease. CNS. Neurosci. 1-10.

Chaturvedi RK, Flint Beal M (2013). Mitochondrial Diseases of the Brain. Free. Radic. Biol. Med. 63:1-29.

Chen CT, Green JT, Orr SK, Bazinet RP (2008). Regulation of brain polyunsaturated fatty acid uptake and turnover. Prostaglandins Leukot. Essent. Fatty Acids 79:85-91.

Chen S, Le W (2006). Neuroprotective therapy in Parkinson disease. Am. J. Ther. 13:445-457.

Cheynier V, Comte G, Davies KM, Lattanzio V, Martens S (2013). Plant phenolics: Recent advances on their biosynthesis, genetics, and ecophysiology. Plant. Physiol. Biochem. 72:1-20.

Page 13: Potential involvement of oxidative stress in induction of

Choi DY, Lee YJ, Hong JT, Lee, HJ (2012). Antioxidant properties of

natural polyphenols and their therapeutic potentials for Alzheimer's disease. Brain. Res. Bull. 87:144-153.

Dadheech G, Mishra S, Gautam S, Sharma P (2006). Oxidative stress, α-tocopherol, ascorbic acid and reduced glutathione status in schizophrenics. Indian J. Clin. Biochem. 21:34-38.

Dajas F (2012). Life or death: Neuroprotective and anticancer effects of quercetin. J. Ethnopharmacol. 143:383-396.

Dakhale GN, Khanzode SD, Khanzode SS, Saoji A (2005). Supplementation of vitamin C with atypical antipsychotics reduces oxidative stress and improves the outcome of schizophrenia. Psychopharmacology 182:494-498.

Dasuri K, Zhang L, Keller JN (2013). Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free. Radic. Biol. Med. 62:170-185.

De Mello AJM, Fasolo D (2014). Chapter 20 - Polyphenol Antioxidants from Natural Sources and Contribution to Health Promotion. In: Watson RR, Preedy VR, Zibadi S (eds) Polyphenols in Human Health and Disease. AcademicPress, San Diego. pp. 253-265.

De Oliveira BF, Veloso CA, Nogueira-Machado JA, de Moraes EN, dos Santos RR, Cintra MTG, Chaves MM (2012). Ascorbic acid, alpha-tocopherol, and beta-carotene reduce oxidative stress and proinflammatory cytokines in mononuclear cells of Alzheimer's disease patients. Nutr. Neurosci. 15:244-251.

De Toma AS, Choi J-S, Braymer JJ, Lim  MH (2011). Myricetin: A Naturally Occurring Regulator of Metal-Induced Amyloid-β Aggregation and Neurotoxicity. Chem. BioChem. 12:1198-1201.

Denny Joseph KM, Muralidhara (2013). Enhanced neuroprotective effect of fish oil in combination with quercetin against 3 nitropropionic acid induced oxidative stress in rat brain. Prog. Neuropsychopharmacol. Biol. Psychiatry 40:83-92.

Deslauriers J, Racine W, Sarret P, Grignon S (2014). Preventive effect of α-lipoic acid on prepulse inhibition deficits in a juvenile two-hit model of schizophrenia. Neuroscience 272:261-270.

Dhillon VS, Fenech M (2013). Mutations that affect mitochondrial functions and their association with neurodegenerative diseases. Mutat. Res-Rev. Mutat. 759:1-13.

Di Mauro S, Schon EA (2008). Mitochondrial disorders in the nervous system. Ann. Rev. Neurosci. 31:91-123.

Dickson DW, Braak H, Duda JE, Duyckaerts C, Gasser T, Halliday GM, Hardy J, Leverenz JB, Del Tredici K, Wszolek ZK, Litvan I (2009). Neuropathological assessment of Parkinson's disease: refining the diagnostic criteria. Lancet. Neurol. 8:1150-1157.

Dietrich-Muszalska A, Kontek B, Olas B, Rabe-Jabłońska J (2012). Epicatechin Inhibits Human Plasma Lipid Peroxidation Caused by Haloperidol In Vitro. Neurochem. Res. 37:557-562.

Ding QX, Markesbery WR, Chen QW, Li F, Keller JN (2005). Ribosome dysfunction is an early event in Alzheimer's disease. J. Neurosci. 25:9171-9175.

Ebrahimi A, Schluesener H (2012). Natural polyphenols against neurodegenerative disorders: Potentials and pitfalls. Ageing. Res. Rev. 11:329-345.

Ehrnhoefer DE, Duennwald M, Markovic P, Wacker JL, Engemann S, Roark M, Legleiter J, Marsh JL, Thompson LM, Lindquist S, Muchowski PJ, Wanker EE (2006). Green tea (−)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington's disease models. Hum. Mol. Genet. 15:2743-2751.

Ejaz Ahmed M, Khan MM, Javed H, Vaibhav K, Khan A, Tabassum R, Ashafaq M, Islam F, Safhi MM, Islam F (2013). Amelioration of cognitive impairment and neurodegeneration by catechin hydrate in rat model of streptozotocin-induced experimental dementia of Alzheimer’s type. Neurochem. Int. 62:492-501.

Engelhart MJ, Geerlings MI, Ruitenberg A, Van Swieten JC, Hofman A, Witteman JCM, Breteler MMB (2002). Dietary intake of antioxidants and risk of Alzheimer disease. JAMA 287:3223-3229.

Feng X, Liang N, Zhu D, Gao Q, Peng L, Dong H, Yue Q, Liu H, Bao L, Zhang J, Hao J, Gao Y, Yu X, Sun J (2013). Resveratrol Inhibits β-Amyloid-Induced Neuronal Apoptosis through Regulation of SIRT1-ROCK1 Signaling Pathway. PLoS ONE. 8:1-11.

Filomeni G, Graziani I, De Zio D, Dini L, Centonze D, Rotilio G, Ciriolo MR (2012). Neuroprotection of kaempferol by autophagy in models of

Oliveira et al. 697

rotenone-mediated acute toxicity: possible implications for Parkinson's disease. Neurobiol. Aging 33:767-785.

Fontanilla CV, Ma Z, Wei X, Klotsche J, Zhao L, Wisniowski P, Dodel RC, Farlow MR, Oertel WH, Du Y (2011). Caffeic acid phenethyl ester prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurodegeneration. Neuroscience. 188:135-141.

Foy CJ, Passmore AP, Vahidassr MD, Young IS, Lawson JT (1999). Plasma chain-breaking antioxidants in Alzheimer's disease, vascular dementia and Parkinson's disease. QJM 92:39-45.

Gackowski D, Rozalski R, Siomek A, Dziaman T, Nicpon K, Klimarczyk M, Araszkiewicz A, Olinski R (2008). Oxidative stress and oxidative DNA damage is characteristic for mixed Alzheimer disease/vascular dementia. J. Neurol. Sci. 266:57-62.

George RC, Lew J, Graves DJ (2013). Interaction of Cinnamaldehyde and Epicatechin with Tau: Implications of Beneficial Effects in Modulating Alzheimer's Disease Pathogenesis. J. Alzheimers Dis. 36:21-40.

Goldberg YP, Telenius H, Hayden MR (1994). The molecular genetics of Huntington’s disease. Curr. Opin. Neurol. 7:325-32.

Good PF, Werner P, Hsu A, Olanow CW, Perl DP (1996). Evidence of neuronal oxidative damage in Alzheimer's disease. Am. J. Pathol. 149:21-28.

Guan JZ, Guan WP, Maeda T, Makino N (2012). Effect of Vitamin E Administration on the Elevated Oxygen Stress and the Telomeric and Subtelomeric Status in Alzheimer’s Disease. Gerontology 58:62-69.

Gubandru M, Margina D, Tsitsimpikou C, Goutzourelas N, Tsarouhas K, Ilie M, Tsatsakis AM, Kouretas D (2013). Alzheimer’s disease treated patients showed different patterns for oxidative stress and inflammation markers. Food Chem. Toxicol. 61:209-214.

Gysin R, Kraftsik R, Sandell J, Bovet P, Chappuis C, Conus P, Deppen P, Preisig M, Ruiz V, Steullet P (2007). Impaired glutathione synthesis in schizophrenia: convergent genetic and functional evidence. Proc. Natl. Acad. Sci. 104:16621-16626.

Ha AD, Beck CA, Jankovic J (2012). Intermediate CAG repeats in Huntington's disease: analysis of COHORT. Tremor Other Hyperkinet. Mov. 2:1-7.

Halliday G, Lees A, Stern M (2011). Milestones in Parkinson's disease clinical and pathologic features. Mov. Disord. 26:1015-1021.

Halter B, Gonzalez de Aguilar J-L, Rene F, Petri S, Fricker B, Echaniz-Laguna A, Dupuis L, Larmet Y, Loeffler JP (2010). Oxidative stress in skeletal muscle stimulates early expression of Rad in a mouse model of amyotrophic lateral sclerosis. Free Radic. Biol. Med. 48:915-923.

Hamaguchi T, Ono K, Murase A, Yamada M (2009). Phenolic Compounds Prevent Alzheimer’s Pathology through Different Effects on the Amyloid-β Aggregation Pathway. Am. J. Pathol. 175:2557-2565.

Harjes P, Wanker EE (2003). The hunt for huntingtin function: interaction partners tell many different stories. Trends Biochem. Sci. 28:425-33.

Hashim A, Wang L, Juneja K, Ye Y, Zhao Y, Ming LJ (2011). Vitamin B6s inhibit oxidative stress caused by Alzheimer’s disease-related CuII-β-amyloid complexes-cooperative action of phospho-moiety. Bioorg. Med. Chem. Lett. 21:6430-6432.

Hauser DN, Hastings TG (2013). Mitochondrial dysfunction and oxidative stress in Parkinson's disease and monogenic Parkinsonism. Neurobiol. Dis. 51:35-42.

He M, Liu MY, Wang S, Tang QS, Yao WF, Zhao HS, Wei MJ (2012). Research on EGCG improving the degenerative changes of the brain in AD model mice induced with chemical drugs. Zhong Yao Cai. 35:1641-1644.

Heiser P, Sommer O, Schmidt A, Clement H, Hoinkes A, Hopt U, Schulz E, Krieg J, Dobschütz E (2010). Effects of antipsychotics and vitamin C on the formation of reactive oxygen species. J. Psychopharmacol. 24:1499-1504.

Heo HJ, Kim DO, Shin SC, Kim MJ, Kim BG, Shin DH (2004). Effect of Antioxidant Flavanone, Naringenin, from Citrus junos on Neuroprotection. J. Agric. Food Chem. 52:1520-1525.

Heo JH, Hyon-Lee, Lee KM (2013). The Possible Role of Antioxidant Vitamin C in Alzheimer’s Disease Treatment and Prevention. Am. J. Alzheimers. Dis. Other. Demen. 28:120-125.

Hickey M, Zhu C, Medvedeva V, Lerner R, Patassini S, Franich N, Maiti P, Frautschy S, Zeitlin S, Levine M, Chesselet MF (2012).

Page 14: Potential involvement of oxidative stress in induction of

698 Afr. J. Pharm. Pharmacol.

Improvement of neuropathology and transcriptional deficits in CAG 140 knock-in mice supports a beneficial effect of dietary curcumin in Huntington's disease. Mol. Neurodegen. 12:1-16.

Ho DJ, Calingasan NY, Wille E, Dumont M, Beal MF (2010). Resveratrol protects against peripheral deficits in a mouse model of Huntington's disease. Exp. Neurol. 225: 74-84.

Hoepken HH, Gispert S, Morales B, Wingerter O, Del Turco D, Mülsch A, Nussbaum RL, Müller K, Dröse S, Brandt U (2007). Mitochondrial dysfunction, peroxidation damage and changes in glutathione metabolism in PARK6. Neurobiol. Dis. 25:401-411.

Honda K, Smith MA, Zhu XW, Baus D, Merrick WC, Tartakoff AM, Hattier T, Harris PL, Siedlak SL, Fujioka H, Liu Q, Moreira PI, Miller FP, Nunomura A, Shimohama S, Perry G (2005). Ribosomal RNA in Alzheimer disease is oxidized by bound redox-active iron. J. Biol. Chem. 280:20978-20986.

Huang HC, Tang D, Xu K, Jiang ZF (2013b). Curcumin attenuates amyloid-β-induced tau hyperphosphorylation in human neuroblastoma SH-SY5Y cells involving PTEN/Akt/GSK-3β signaling pathway. J Recept. Signal. Transduct. Res. 1-12.

Huang Y, Jin M, Pi R, Zhang J, Chen M, Ouyang Y, Liu A, Chao X, Liu P, Liu J, Ramassamy C, Qin J (2013a). Protective effects of caffeic acid and caffeic acid phenethyl ester against acrolein-induced neurotoxicity in HT22 mouse hippocampal cells. Neurosci. Lett. 535:146-151.

Huebbe P, Wagner AE, Boesch-Saadatmandi C, Sellmer F, Wolffram S, Rimbach G (2010). Effect of dietary quercetin on brain quercetin levels and the expression of antioxidant and Alzheimer's disease relevant genes in mice. Pharmacol. Res. 61:242-246.

Hyun DH, Lee M, Hattori N, Kubo SI, Mizuno Y, Halliwell B, Jenner P (2002). Effect of wild-type or mutant Parkin on oxidative damage, nitric oxide, antioxidant defenses, and the proteasome. J. Biol. Chem. 277:28572-28577.

Islam F, Khan MM, Raza SS, Javed H, Ahmad A, Khan A, Safhi MM (2012). Rutin protects dopaminergic neurons from oxidative stress in an animal model of Parkinson's Disease. Neurotox. Res. 22:1-15.

Jazvinšćak Jembrek M, Vuković L, Puhović J, Erhardt J, Oršolić N (2012). Neuroprotective Effect of Quercetin Against Hydrogen Peroxide-induced Oxidative Injury in P19 Neurons. J. Mol. Neurosci. 47:286-299.

Jin F, Wu Q, Lu Y-F, Gong Q-H, Shi JS (2008b). Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson's disease in rats. Eur. J. Pharmacol. 600:78-82.

Jin H, Kanthasamy A, Ghosh A, Anantharam V, Kalyanaraman B, Kanthasamy AG (2013a). Mitochondria-targeted antioxidants for treatment of Parkinson's disease: Preclinical and clinical outcomes. Biochim. Biophys. Acta. 1842:1282-1294.

Johri A, Beal MF (2012). Antioxidants in Huntington's disease. Biochim. Biophys Acta. Dis. 1822:664-674. Kalonia H, Kumar P, Kumar A (2010). Targeting oxidative stress attenuates malonic acid induced Huntington like behavioral and mitochondrial alterations in rats. Eur. J. Pharmacol. 634:46-52.

Keeney PM, Xie J, Capaldi RA, Bennett JP (2006). Parkinson's disease brain mitochondrial complex I has oxidatively damaged subunits and is functionally impaired and misassembled. J. Neurosci. 26:5256-5264.

Khan MB, Khan MM, Khan A, Ahmed ME, Ishrat T, Tabassum R, Vaibhav K, Ahmad A, Islam F (2012). Naringenin ameliorates Alzheimer’s disease (AD)-type neurodegeneration with cognitive impairment (AD-TNDCI) caused by the intracerebroventricular-streptozotocin in rat model. Neurochem. Int. 61:1081-1093.

Khan MM, Ahmad A, Ishrat T, Khan MB, Hoda MN, Khuwaja G, Raza SS, Khan A, Javed H, Vaibhav K, Islam F (2010). Resveratrol attenuates 6-hydroxydopamine-induced oxidative damage and dopamine depletion in rat model of Parkinson's disease. Brain. Res. 1328:139-151.

Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers JT, Delalle I, Baur JA, Sui G, Armour SM, Puigserver P, Sinclair DA, Tsai LH (2007). SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. EMBO J. 26: 3169-3179.

Kim J, Choi S, Cho H, Hwang H, Kim Y, Lim S, Kim C, Kim H, Peterson S, Shin D (2010a). Protective effects of kaempferol (3,4',5,7-

tetrahydroxyflavone) against amyloid beta peptide (Abeta)-induced neurotoxicity in ICR mice. Biosci. Biotechnol. Biochem. 74:397-401.

Kim JS, Kim JM, O JJ, Jeon BS (2010b). Inhibition of inducible nitric oxide synthase expression and cell death by (−)-epigallocatechin-3-gallate, a green tea catechin, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. J. Clin. Neurosci. 17:1165-1168.

Kim TS, Pae CU, Yoon SJ, Jang WY, Lee NJ, Kim JJ, Lee SJ, Lee C, Paik IH, Lee CU (2006). Decreased plasma antioxidants in patients with Alzheimer's disease. Int. J. Geriatr. Psychiatry 21:344-348.

Koh SH, Lee SM, Kim HY, Lee K-Y, Lee YJ, Kim HT, Kim J, Kim M-H, Hwang MS, Song C, Yang KW, Lee KW, Kim SH, Kim OH (2006). The effect of epigallocatechin gallate on suppressing disease progression of ALS model mice. Neurosci. Lett. 395:103-107.

Kontush A, Mann U, Arlt S, Ujeyl A, Lührs C, Müller-Thomsen T, Beisiegel U (2001). Influence of vitamin E and C supplementation on lipoprotein oxidation in patients with Alzheimer’s disease. Free. Radic. Biol. Med. 31:345-354.

Kontush A, Schekatolina S (2004). Vitamin E in Neurodegenerative Disorders: Alzheimer's Disease. Ann. NY Acad. Sci. 1031:249-262.

Koskenkorva-Frank TS, Weiss G, Koppenol WH, Burckhardt S (2013). The complex interplay of iron metabolism, reactive oxygen species, and reactive nitrogen species: Insights into the potential of various iron therapies to induce oxidative and nitrosative stress. Free. Radic. Biol. Med. 65:1174-1194.

Kovacic P, Somanathan R (2012). Redox Processes in Neurodegenerative Disease Involving Reactive Oxygen Species. Curr. Neuropharmacol. 10:289-302.

Lagoa R, Lopez-Sanchez C, Samhan-Arias AK, Gañan CM, Garcia-Martinez V, Gutierrez-Merino C (2009). Kaempferol protects against rat striatal degeneration induced by 3-nitropropionic acid. J. Neurochem. 111:473-487.

Lavoie S, Chen Y, Dalton TP, Gysin R, Cuénod M, Steullet P, Do KQ (2009). Curcumin, quercetin, and tBHQ modulate glutathione levels in astrocytes and neurons: importance of the glutamate cysteine ligase modifier subunit. J. Neurochem. 108:1410-1422.

Lee HP, Casadesus G, Zhu X, Lee HG, Perry G, Smith MA, Gustaw-Rothenberg K, Lerner A (2009). All-trans retinoic acid as a novel therapeutic strategy for Alzheimer's disease. Expert. Rev. Neurother. 9:1615-1621.

Lee M, Hyun DH, Jenner P, Halliwell B (2001). Effect of proteasome inhibition on cellular oxidative damage, antioxidant defences and nitric oxide production. J. Neurochem. 78:32-41.

Lee MY, Choi EJ, Lee M-K, Lee JJ (2013). Epigallocatechin gallate attenuates L-DOPA-induced apoptosis in rat PC12 cells. Nutr. Res. Pract. 7:249-255.

Levites Y, Weinreb O, Maor G, Youdim MBH, Mandel S (2001) Green tea polyphenol (-)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine-induced dopaminergic neurodege-neration. J. Neurochem. 78:1073-1082.

Li FJ, Shen L, Ji HF (2012). Dietary Intakes of Vitamin E, Vitamin C, and β-Carotene and Risk of Alzheimer's Disease: A Meta-Analysis. J. Alzheimers. Dis. 31:253-258.

Li J, Han B, Zhang X, Lou J, Shuboy A, Wang L, Yin H, Wang Y (2013). Curcumin as a Potential Treatment for Alzheimer's Disease: A Study of the Effects of Curcumin on Hippocampal Expression of Glial Fibrillary Acidic Protein. Am. J. Chin. Med. 41:59-70.

Li S, Pu XP (2011). Neuroprotective Effect of Kaempferol against a 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Mouse Model of Parkinson's Disease. Biol. Pharm. Bull. 34:1291-1296.

Li SY, Wang XB, Kong LY (2014). Design, synthesis and biological evaluation of imine resveratrol derivatives as multi-targeted agents against Alzheimer's disease. Eur. J. Med. Chem. 71:36-45.

Lim HJ, Shim SB, Jee SW, Lee SH, Lim CJ, Hong JT, Sheen YY, Hwang DY (2013). Green tea catechin leads to global improvement among Alzheimer's disease-related phenotypes in NSE/hAPP-C105 Tg mice. J. Nutr. Biochem. 24:1302-1313.

Liochev SI (2013). Reactive oxygen species and the free radical theory of aging. Free. Radic. Biol. Med. 60:1-4.

Liu LX, Chen WF, Xie JX, Wong MS (2008). Neuroprotective effects of genistein on dopaminergic neurons in the mice model of Parkinson's disease. Neurosci. Res. 60:156-161.

Page 15: Potential involvement of oxidative stress in induction of

Liu R, Zhang TT, Zhou D, Bai XY, Zhou WL, Huang C, Song JK, Meng

FR, Wu CX, LI L, Du GH (2013). Quercetin protects against the Aβ25–35-induced amnesic injury: Involvement of inactivation of RAGE-mediated pathway and conservation of the NVU. Neuropharmacology 67:419-431.

Lloret A, Badía MC, Mora NJ, Pallardó FV, Alonso MD, Viña J (2009). Vitamin E Paradox in Alzheimer's Disease: It Does Not Prevent Loss of Cognition and May Even Be Detrimental. J. Alzheimers. Dis. 17:143-149.

Ma J, Yang WQ, Zha H, Yu HR (2013). Effect of naringenin on learning and memory ability on model rats with Alzheimer disease. Zhong. Yao. Cai. 36:271-276.

Mansouri MT, Naghizadeh B, Ghorbanzadeh B, Farbood Y, Sarkaki A, Bavarsad K (2013). Gallic acid prevents memory deficits and oxidative stress induced by intracerebroventricular injection of streptozotocin in rats. Pharmacol. Biochem. Behav. 111:90-96.

Mansouri Z, Sabetkasaei M, Moradi F, Masoudnia F, Ataie A (2012). Curcumin has Neuroprotection Effect on Homocysteine Rat Model of Parkinson. J. Mol. Neurosci. 47:234-242.

Massaad CA, Pautler RG, Klann E (2009). Mitochondrial superoxide a key player in Alzheimer’s disease. Aging 9:758-761.

Mattiazzi M, D'Aurelio M, Gajewski CD, Martushova K, Kiaei M, Beal MF, Manfredi G (2002). Mutated human SOD1 causes dysfunction of oxidative phosphorylation in mitochondria of transgenic mice. J. Biol. Chem. 277:29626-29633.

Mecocci P, Macgarvey U, Beal MF (1994). Oxidative damage to mitochondrial-DNA is increased in Alzheimer's disease. Ann. Neurol. 36:747-751.

Milenkovic D, Jude B, Morand C (2013). miRNA as molecular target of polyphenols underlying their biological effects. Free. Radic. Biol. Med. 64:40-51.

Miyake Y, Fukushima W, Tanaka K, Sasaki S, Kiyohara C, Tsuboi Y, Yamada T, Oeda T, Miki T, Kawamura N, Sakae N, Fukuyama H, Hirota Y, Nagai M, Fukuoka Kinki Parkinson’s Disease Study G (2011). Dietary intake of antioxidant vitamins and risk of Parkinson’s disease: a case–control study in Japan. Eur. J. Neurol. 18:106-113.

Mochel F, Haller RG (2011). Energy deficit in Huntington disease: why it matters. J. Clin. Invest. 121:493-499.

Molina-Jiménez MaF, Sánchez-Reus MaI, Andres D, Cascales Ma, Benedi J (2004). Neuroprotective effect of fraxetin and myricetin against rotenone-induced apoptosis in neuroblastoma cells. Brain. Res. 1009:9-16.

Montilla-López P, Muñoz-Águeda MC, Feijóo López M, Muñoz-Castañeda JR, Bujalance-Arenas I, Túnez-Fiñana I (2002). Comparison of melatonin versus vitamin C on oxidative stress and antioxidant enzyme activity in Alzheimer's disease induced by okadaic acid in neuroblastoma cells. Eur. J. Pharmacol. 451:237-243.

Morales LM, Estévez J, Suárez H, Villalobos R, Chacín de Bonilla L, Bonilla E (1989). Nutritional evaluation of Huntington disease patients. Am. J. Clin. Nutr. 50:145-150.

Mori T, Koyama N, Guillot-Sestier M-V, Tan J, Town T (2013). Ferulic Acid Is a Nutraceutical β-Secretase Modulator That Improves Behavioral Impairment and Alzheimer-like Pathology in Transgenic Mice. PLoS ONE. 8:1-16.

Morris MC, Evans DA, Tangney CC, Bienias JL, Wilson RS, Aggarwal NT, Scherr PA (2005). Relation of the tocopherol forms to incident Alzheimer disease and to cognitive change. Am. J. Clin. Nutr. 81:508-514.

Morrissey PA, Hill TR (2011). Vitamins|Vitamin E. In: Fuquay JW (ed) Encyclopedia of Dairy Sciences (Second Edition). Academic Press, San Diego. pp. 652-660.

Moshahid KM, Raza S, Javed H, Ahmad A, Khan A, Islam F, Safhi M, Islam F (2012). Rutin Protects Dopaminergic Neurons from Oxidative Stress in an Animal Model of Parkinson’s Disease. Neurotox. Res. 22:1-15.

Nassiri-Asl M, Sarookhani MR, Abbasi E, Zangivand AA, Shakiba E, Sedighi A, Rahbari M (2012). The effects of pre-treatment with vitamin B6 on memory retrieval in rats. Food. Chem. 132:1046-1048

Nunomura A, Moreira P, Castellani R, Lee H, Zhu X, Smith M, Perry G (2012). Oxidative Damage to RNA in Aging and Neurodegenerative Disorders. Neurotox. Res. 22:231-248.

Nunomura A, Perry G, Pappolla MA, Wade R, Hirai K, Chiba S, Smith

Oliveira et al. 699

MA (1999). RNA oxidation is a prominent feature of vulnerable neurons in Alzheimer's disease. J. Neurosci. 19:1959-1964.

Ono K, Yamada M (2012). Vitamin A and Alzheimer's disease. Geriatr.Gerontol. Int. 12:180-188.

Ossola B, Kääriäinen TM, Männistö PT (2009). The multiple faces of quercetin in neuroprotection. Expert. Opin. Drug Saf. 8:397-409.

Pandey AK, Patnaik R, Muresanu DF, Sharma A, Sharma HS (2012). Quercetin in Hypoxia-Induced Oxidative Stress: Novel Target for Neuroprotection. Int. Rev. Neurobiol. 102:107-146.

Pansarasa O, Rossi D, Berardinelli A, Cereda C (2013). Amyotrophic Lateral Sclerosis and Skeletal Muscle: An Update. Mol. Neurobiol. 49:984-990.

Parihar MS, Brewer GJ (2007). Microenergetic failure in Alzheimer’s disease. Am. J. Physiol. Cell. Physiol. 292:8-23.

Perfeito R, Cunha-Oliveira T, Rego AC (2013). Reprint of: Revisiting oxidative stress and mitochondrial dysfunction in the pathogenesis of Parkinson disease—resemblance to the effect of amphetamine drugs of abuse. Free. Radic. Biol. Med. 62:186-201.

Peyser CE, Folstein M, Chase GA, Starkstein S, Brandt J, Cockrell JR, Bylsma F, Coyle JT, McHugh PR, Folstein SE (1995). Trial of d-α-tocopherol in Huntington's disease. Am. J. Psychiatry 152:1771-1775.

Pi R, Mao X, Chao X, Cheng Z, Liu M, Duan X, Ye M, Chen X, Mei Z, Liu P, Li W, Han Y (2012). Tacrine-6-Ferulic Acid, a Novel Multi-functional Dimer, Inhibits Amyloid-β-Mediated Alzheimer's Disease-Associated Pathogenesis in vitro and in vivo. PLoS. ONE 7:1-8.

Porquet D, Casadesús G, Bayod S, Vicente A, Canudas A, Vilaplana J, Pelegrí C, Sanfeliu C, Camins A, Pallàs M, Valle J (2013). Dietary resveratrol prevents Alzheimer’s markers and increases life span in SAMP8. AGE 35:1851-1865.

Quinn J, Suh J, Moore MM, Kaye J, Frei B (2003). Antioxidants in Alzheimer's disease-vitamin C delivery to a demanding brain. J. Alzheimers. Dis. 5:309-313.

Rao A, Sudha K, Rao S (2003). Free radical toxicity and antioxidants in Parkinson’s disease. Neurology 51:60-62.

Rebec GV, Conroy SK, Barton SJ (2006). Hyperactive striatal neurons in symptomatic Huntington R6/2 mice: Variations with behavioral state and repeated ascorbate treatment. Neuroscience 137:327-336.

Reed TT, Pierce JWM, Turner DM, Markesbery WR, Butterfield DA (2009). Proteomic identification of nitrated brain prot0e0ins in early Alzheimer's disease inferior parietal lobule. J. Cell. Mol. Med. 13:2019-2029.

Ricciarelli R, Argellati F, Pronzato MA, Domenicotti C (2007). Vitamin E and neurodegenerative diseases. Mol. Aspects. Med. 28:591-606.

Roberts LJ, Fessel JP (2004). The biochemistry of the isoprostane, neuroprostane and isofuran pathways of lipid peroxidation. Chem. Phys. Lipids. 128:173-186.

Roberts RA, Smith RA, Safe S, Szabo C, Tjalkens RB, Robertson FM (2010). Toxicological and pathophysiological roles of reactive oxygen and nitrogen species. Toxicology. 276:85-94.

Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O'Regan JP, Deng HX (1993). Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362:59-62.

Ruipérez V, Darios F, Davletov B (2010). Alpha-synuclein, lipids and Parkinson’s disease. Prog. Lipid Res. 49:420-428.

Sanders LH, Greenamyre JT (2013). Oxidative damage to macromolecules in human Parkinson disease and the rotenone model. Free Radic. Bio. Med. 62:111-120.

Sandhir R, Mehrotra A (2013). Quercetin supplementation is effective in improving mitochondrial dysfunctions induced by 3-nitropropionic acid: Implications in Huntington's disease. Biochim. Biophys. Acta. 1832:421-430.

Sandhir R, Yadav A, Mehrotra A, Sunkaria A, Singh A, Sharma S (2013). Curcumin Nanoparticles Attenuate Neurochemical and Neurobehavioral Deficits in Experimental Model of Huntington’s Disease. Neuromol. Med. 6:1-13.

Schrag M, Mueller C, Zabel M, Crofton A, Kirsch WM, Ghribi O, Squitti R, Perry G (2013). Oxidative stress in blood in Alzheimer's disease and mild cognitive impairment: A meta-analysis. Neurobiol. Dis. 59:100-110.

Shan X, Tashiro H, Lin CLG (2003). The identification and characterization of oxidized RNAs in Alzheimer's disease. J.

Page 16: Potential involvement of oxidative stress in induction of

700 Afr. J. Pharm. Pharmacol.

Neurosci. 23:4913-4921. Sharoar M, Thapa A, Shahnawaz M, Ramasamy V, Woo ER, Shin S,

Park IS (2012). Keampferol-3-O-rhamnoside abrogates amyloid beta toxicity by modulating monomers and remodeling oligomers and fibrils to non-toxic aggregates. J. Biomed. Sci. 19:1-13.

Sivrioglu EY, Kirli S, Sipahioglu D, Gursoy B, Sarandöl E (2007). The impact of ω-3 fatty acids, vitamins E and C supplementation on treatment outcome and side effects in schizophrenia patients treated with haloperidol: An open-label pilot study. Prog. Neuropsychopharmacol. Biol. Psychiatry 31:1493-1499.

Smith JA, Park S, Krause JS, Banik NL (2013). Oxidative stress, DNA damage, and the telomeric complex as therapeutic targets in acute neurodegeneration. Neurochem. Int. 62:764-775.

Smith MA, Richey Harris PL, Sayre LM, Beckman JS, Perry G (1997). Widespread peroxynitrite-mediated damage in Alzheimer's disease. J. Neurosci. 17:2653-2657.

Sonia Angeline M, Sarkar A, Anand K, Ambasta RK, Kumar P (2013). Sesamol and naringenin reverse the effect of rotenone-induced PD rat model. Neuroscience 254:379-394.

Subramaniam SR, Chesselet MF (2013). Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Prog. Neurobiol. 107:17-32.

Sultana R, Perluigi M, Allan Butterfield D (2013). Lipid peroxidation triggers neurodegeneration: A redox proteomics view into the Alzheimer disease brain. Free Radic. Biol. Med. 62:157-169.

Sutachan JJ, Casas Z, Albarracin SL, Stab BR, Samudio I, Gonzalez J, Morales L, Barreto GE (2012). Cellular and molecular mechanisms of antioxidants in Parkinson's disease. Nutr. Neurosci. 15:120-126.

Takeuchi H, Kobayashi Y, Ishigaki S, Doyu M, Sobue G (2002). Mitochondrial localization of mutant superoxide dismutase 1 triggers caspase-dependent cell death in a cellular model of familial amyotrophic lateral sclerosis. J. Biol. Chem. 277:50966-50972.

Teixeira MDA, Souza CM, Menezes APF, Carmo MRS, Fonteles AA, Gurgel JP, Lima FAV, Viana GSB, Andrade GM (2013). Catechin attenuates behavioral neurotoxicity induced by 6-OHDA in rats. Pharmacol. Biochem. Behav. 110:1-7.

Tohgi H, Abe T, Yamazaki K, Murata T, Ishizaki E, Isobe C (1999). Alterations of 3-nitrotyrosine concentration in the cerebrospinal fluid during aging and in patients with Alzheimer's disease. Neurosci. Lett. 269:52-54.

Trieu VN, Uckun FM (1999). Genistein Is Neuroprotective in Murine Models of Familial Amyotrophic Lateral Sclerosis and Stroke. Biochem. Biophys. Res. Commun. 258:685-688.

Vijayvergiya C, Beal MF, Buck J, Manfredi G (2005). Mutant superoxide dismutase 1 forms aggregates in the brain mitochondrial matrix of amyotrophic lateral sclerosis mice. J. Neurosci. 25:2463-2470.

Viña J, Lloret A, Ortí R, Alonso D (2004). Molecular bases of the treatment of Alzheimer's disease with antioxidants: prevention of oxidative stress. Mol. Aspects Med. 25:117-123.

Wang J, Zhang Y, Tang L, Zhang N, Fan D (2011). Protective effects of resveratrol through the up-regulation of SIRT1 expression in the mutant hSOD1-G93A-bearing motor neuron-like cell culture model of amyotrophic lateral sclerosis. Neurosci. Lett. 503:250-255.

Wang P, Su C, Li R, Wang H, Ren Y, Sun H, Yang J, Sun J, Shi J, Tian

J, Jiang S (2013b). Mechanisms and effects of curcumin on spatial learning and memory improvement in APPswe/PS1dE9 mice. J. Neurosci. Res. 92: 218-231.

Wang SW, Wang YJ, Su YJ, Zhou W, Yang SG, Zhang R, Zhao M, Li YN, Zhang ZP, Zhan DW, Liu RT (2012). Rutin inhibits β-amyloid aggregation and cytotoxicity, attenuates oxidative stress, and decreases the production of nitric oxide and proinflammatory cytokines. Neurotoxicology 33:482-490.

Wang X, Wang W, Li L, Perry G, Lee H-g, Zhu X (2013a). Oxidative stress and mitochondrial dysfunction in Alzheimer's disease. Biochim. Biophys. Acta 1842:1240-1247.

Wu JQ, Kosten TR, Zhang XY (2013). Free radicals, antioxidant defense systems, and schizophrenia. Prog. Neuro-Psychoph. 46:200-206.

Xu Z, Chen S, Li X, Luo G, Li L, Le W (2006). Neuroprotective Effects of (-)-Epigallocatechin-3-gallate in a Transgenic Mouse Model of Amyotrophic Lateral Sclerosis. Neurochem. Res. 31:1263-1269.

Yan MH, Wang X, Zhu X (2013). Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic. Biol. Med. 62:90-101.

Yao JK, Keshavan MS (2011). Antioxidants, redox signaling, and pathophysiology in schizophrenia: an integrative view. Antioxid. Redox Signal 15:2011-2035.

Yu SY, Zhang M, Luo J, Zhang L, Shao Y, Li G (2013). Curcumin ameliorates memory deficits via neuronal nitric oxide synthase in aged mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 45:47-53.

Zbarsky V, Datla KP, Parkar S, Rai DK, Aruoma OI, Dexter DT (2005). Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkinson's disease. Free Radic. Res. 39:1119-1125.

Zhang K, Ma Z, Wang J, Xie A, Xie J (2011). Myricetin attenuated MPP+-induced cytotoxicity by anti-oxidation and inhibition of MKK4 and JNK activation in MES23.5 cells. Neuropharmacology 61:329-335.

Zhang X, Wu M, Lu F, Luo N, He Z-P, Yang H (2013). Involvement of α7 nAChR Signaling Cascade in Epigallocatechin Gallate Suppression of β-Amyloid-Induced Apoptotic Cortical Neuronal Insults. Mol. Neurobiol. 49:66-77.

Zhao L, Wang JL, Liu R, Li XX, Li JF, Zhang L (2013a). Neuroprotective, Anti-Amyloidogenic and Neurotrophic Effects of Apigenin in an Alzheimer’s Disease Mouse Model. Molecules 18:9949-9965.

Zhao L, Wang JL, Wang YR, Fa XZ (2013b). Apigenin attenuates copper-mediated β-amyloid neurotoxicity through antioxidation, mitochondrion protection and MAPK signal inactivation in an AD cell model. Brain Res. 1492:33-45.

Zhu M, Han S, Fink AL (2013). Oxidized quercetin inhibits α-synuclein fibrillization. Biochim. Biophys. Acta. 1830:2872-2881.