Abstract
Neurological diseases like Alzheimer’s disease, epilepsy, parkinsonism, depression, Huntington’s disease and amyotrophic lateral sclerosis prevailing globally are considered to be deeply influenced by oxidative stress-based changes in the biochemical settings of the organs. The excess oxygen concentration triggers the production of reactive oxygen species, and even the intrinsic antioxidant enzyme system, i.e. SOD, CAT and GSHPx, fails to manage their levels and keep them under desirable limits. This consequently leads to oxidation of protein, lipids and nucleic acids in the brain resulting in apoptosis, proteopathy, proteasomes and mitochondrion dysfunction, glial cell activation as well as neuroinflammation. The present exploration deals with the evidence-based mechanism of oxidative stress towards development of key neurological diseases along with the involved biomechanistics and biomaterials.
Introduction
The brain comprises approximately 2% of human body weight, yet it consumes about 20% of the total inhaled oxygen (Clarke and Sokoloff, 1999). The neurons and glial cells are the main types of brain cells extremely accountable for high oxygen consumption (Malonek and Grinvald, 1996; Thompson et al., 2003). However, the oxygen is indispensable for the biological functioning of all the living cells, but its surplus leads to yield reactive oxygen species (ROS) (Yu, 1994), which recompense oxidative stress (OS) trailing to neurodegeneration (Uttara et al., 2009). The free radicals and ROS are normally produced inside the brain and are dedicated for the oxidations of protein, nucleic acids and lipids’ peroxidation (Lobo et al., 2010). The concentration of ROS in the brain is regulated by endogenous antioxidant defense (EAS) system, which is composed of several enzymes, mainly copper-zinc superoxide dismutase (CuZn SOD), manganese superoxide dismutase (MnSOD), catalase and glutathione peroxidases (GSHPx) (Sies, 1997; Halliwell and Gutteridge, 1999). The reduced EAS and increased ROS are entitled for the induction of OS in the system. The neurodegeneration causes aggravation of several neurological diseases, among them Alzheimer’s disease (Rosini et al., 2014), epilepsy (Shin et al., 2011), parkinsonism (Hwang, 2013), depression (Michel et al., 2012), Huntington’s disease (Gil-Mohapel et al., 2014) and amyotrophic lateral sclerosis (Barber et al., 2006) are the prominent ones encountered globally. The biomechanics involving free radicals generation, the damage due to DNA oxidation, the mitochondrial DNA aberrations as well as apoptosis dysregulation are some of the common connection points in these diseases (Jabir et al., 2015).
Concept of free radicals and their generation
In an atomic orbital, the electron pairs always occur to neutralize their spins, i.e. clockwise (ms=+1/2) and anticlockwise (ms=−1/2) (Merzbacher, 1998), but in the case of free radicals, the spin of the unpaired electron is not neutralized, which transforms them to more reactive and damaging entities (Lobo et al., 2010). Several external factors have also been reported for spin impairment of electrons, and among them, age, malnutrition, stress, chemicals, radiation, pollution and drugs are important (Aseervatham et al., 2013). The main free radicals found inside the OS cascade are oxygen free radical (O˙), superoxide free radical (O2˙−), hydroxyl (OH˙) and nitric oxide (NO˙) (Gibson and Lilley, 1997). The external factors trigger both the enzymes, i.e. xanthine oxidase (XO) and NADPH oxidase-2 (NOX-2). The xanthine oxidase or xanthine dehydrogenases are interconvertible oxidoreductase molybdoflavoenzyme responsible for purine catabolism to form a superoxide, hydrogen peroxide and uric acid by catalyzing the dismutation of the SOD enzyme. Typically, this enzyme exists in xanthine dehydrogenase, but Ca+2-induced proteases convert it into XO. The low oxygen levels lead to ATP metabolism and hypoxanthine accumulation, but reperfusion of oxygen triggers XO causing the reduction of hypoxanthine to xanthine molecule and producing the superoxide and H2O2, (Auscher et al., 1979; Kennedy et al., 1989). Moreover, the NADPH oxidase (NOX−2) enzyme, which is a complex of cytochrome b558 (p22PHOX, gp91PHOX), cytosolic proteins (p40PHOX, p47PHOX and p67PHOX) and Rac G-protein (Granger and Kvietys, 2015), produce superoxide free radical as the result of cytosolic protein’s phosphorylation and Rac activation processes. The SOD enzyme exist in three isoforms: Cu/ZnSOD (copper-zinc superoxide dismutase), MnSOD (manganese superoxide dismutase) and ECSOD (extracellular superoxide dismutase) encoded by genes sod1, sod2 and sod3 (Weydert and Cullen, 2010). Additionally, the neuronal nitric oxide synthases (NOS) are found in both central and peripheral nervous systems and are responsible for the metabolism of arginine to NO but in conjunction with superoxide result in the formation of ONOO−. H2O2, ONOO− or both are transformed to OH˙ free radical, and with NO˙, they together act as prominent agents for the neurodegeneration.
The production of OH˙ free radical readily oxidizes (a) structural proteins, (b) nucleic acid and (c) lipids of the neuronal cells. They also disrupt their biological functioning (Pacher et al., 2007; Kong and Lin, 2010). The enzyme, glutathione peroxidase (GPX), uses reduced glutathione (GSH) to remove hydrogen peroxide as H2O (H2O2+2GSH→GS-SG+2H2O) (Ng et al., 2007). Simultaneously, the catalase enzyme, which is localized in peroxisomes, works by sensing the higher concentrations of the hydrogen peroxide by converting it into water and oxygen. Additionally, the monoamine oxidase-B, a type of brain redox enzyme, accountable for metabolism of monoamines into aldehyde, H2O2 and ammonia (RCH2NH2+FAD+O2+H2O→RCHO+FADH2+HOOH+NH3), is also responsible for producing an additional load of H2O2 (Girgin et al., 2004; Yusuf et al., 2008; Weinreb et al., 2016).
Influence of OS on cellular mechanisms
The OS critically affects apoptosis, proteopathy, proteasome dysfunction, mitochondrion dysfunction, glial cell activation, neuroinflammation and vice versa. (Guo et al., 2013). The apoptosis is a physiological process of programmed cell death for destroying unwanted cells overloading, thus maintaining the tissue homeostasis (Hengartner, 2000). The apoptosis can proceed extrinsically and inducted by death receptors or intrinsically by the cytochrome c, caspase-8, 9 releases and formation of apoptosome complex in the mitochondrion (Kajta, 2004; Lavrik et al., 2005). Both the pathways synchronize to release caspase-3, which further activates the caspase-activated DNase, nuclease (CAD) or DNA fragmentation factor 40, a heterodimer of 40 kDa, resulting into the specific DNA fragmentations and apoptotic cell death (Engel and Henshall, 2009; Panickar and Anderson, 2011). This has been evidenced experimentally in animals for seizure-induced neuronal death (Chuang et al., 2007). The apoptotic cells can be distinguished by certain biochemical and morphological changes, such as chromatin condensation, cell shrinkage and internucleosomal DNA fragmentation (Hengartner, 2000) (Figure 1).

The oxidative stress cascade illustrating production of reactive oxygen species (ROS), and their involvement in neurodegeneration.
The OS-induced ROS and mitochondrial Ca2+ efficiently initiates the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) linked channel opening, thereby resulting in free diffusion of the cytochrome c release through the inner and outer membranes of the mitochondria to the cytoplasm where it activates caspase-9 and further the caspase-3 (Emerit et al., 2004; Lavrik et al., 2005). The MPTP opening is also favored by certain mitochondrial-linked proapoptotic (Bax, Bad and Bim) and antiapoptotic (Bcl-xl, Bcl-2 and Bcl-w) proteins (Kajta, 2004; Engel and Henshall, 2009; Henshall and Engel, 2013). The pro-apoptotic signal enhances the release of other mitochondrial-linked protein from the intermembrane space of the mitochondrion called apoptosis-inducing factor (AIF). The AIF triggers DNA fragmentation and also participates in the activation of caspase-9 in the cytoplasm (Kajta, 2004).
The abnormal structure and function of proteins are the main characteristics of proteopathy (Walker and LeVine, 2000). The misfolding and peculiar polymerization of β-amyloid, including the misfolding of the tau protein in the nervous system, has been primarily attributed to the pathogenesis of Alzheimer’s disease (Jucker and Walker, 2011). The repeated triplet codes occurrence in Huntington’s disease (Walker, 2007), protein structure changes in SOD 1 in the case of amyotrophic lateral sclerosis (Rosen et al., 1993) and conformational conversion of α-helix-rich prion protein (PrPC) into PrPSc (β-structure-rich insoluble) (Eghiaian et al., 2004) as well as mutations of α-synuclein have been thought to be responsible in Parkinsonism (Stefanis, 2012). The OS induces protein misfolding via DNA and mtDNA (mitochondrial DNA) mutations (Ling and Söll, 2010).
Among other mechanistic details, the ubiquitin-proteasome system (UPS), which is a foremost pathway for protein and is needed for cellular protein homeostasis by eliminating misfolded or damaged proteins, their oxidative modifications reduces their functions and stability (Droge, 2002; Martindale and Holbrook, 2002). The proteasomal activity declines with aging, resulting in the decreased expression and oxidative modifications (Keller et al., 2000; Carrard et al., 2002). The proteasomal dysfunction causes accumulation of proteins aggregates and is considered to be a hallmark of aging and neurodegeneration. The proteasome inhibition may cause cell death via several factors; among them is the lack of free amino acids produced in the proteasomal degradation (Suraweera et al., 2012). The interaction between protein aggregate-induced ROS generation and redox-active metal ions is the defining key causes (Tabner et al., 2005; Allsop et al., 2008; Jomova et al., 2010).
Besides the oxidative phosphorylation, the mitochondria also assists in apoptosis, ROS signaling and Ca2+ homeostasis in neurons and influence neurodegeneration (Danial and Korsmeyer, 2004; Lin and Beal, 2006). The mitochondrial intracellular ROS production via electron leakage as a by-product of oxidative phosphorylation is an important step. The mitochondrial antioxidative enzymes, such as MnSOD, and glutathione peroxidase maintain the level of ROS, but their excess production can lead to mitochondrial damage and dysfunction. The mitochondrial components, i.e. the electron transport chain (ETC), and mtDNA are very susceptible to ROS linked oxidative damage (Sohal et al., 1995; Lin and Beal, 2006). The mtDNA is 10 times more prone to mutations as compared to nuclear DNA (Mecocci et al., 1993). The mitochondrial dysfunction progresses with age (Turner and Schapira, 2001; Trifunovic and Larsson, 2008) and results in affecting the ATP-dependent processes, such as receptors function, ion channels, vesicle release, pumps and neurotransmitter recycling. During neuron degeneration, the glial cells assist in regulating the metabolism of stressed neurons (Zabel and Kirsch, 2013), and this can be achieved by the up-regulation of glial cell activities, which in itself is a promising indicator of stress and confirms the onset of degeneration and chemical imbalances (Kamal et al., 2014). The frenzied glial stimulation and neuroinflammation critically add to the brain damage, and it was reported in cyanide-induced toxicity (Skaper et al., 2013). The ROS-generated and ROS-activated glial cells lead to cognition linked brain damage. Further, it was also evidenced that the antioxidant compounds reduce the detrimental effects of ROS, OS and the extent of glial activation (Hui et al., 2013).
Mechanisms of protein, lipid and DNA damage
The brain cells are composed of numerous lipidic entities, i.e. fatty acyls, prenol lipids, glycerophospholipids, sterol lipids, glycerolipids, saccharolipids, polyketides and sphingolipids. Their basic functions are (a) lipid bilayers formation providing structural integrity, (b) providing energy reservoir and (c) generating precursors of secondary messengers, such as 1,2-diacylglycerol (DAG), arachidonic acid (ArAc), ceramide, phosphatidic acid, docosahexaenoic acid (DHA) and lysophosphatidic acid (Adibhatla et al., 2006; Adibhatla and Hatcher, 2007). Mostly, the fatty acids are monocarboxylic and straight-chain with one or, two unsaturated bonds in cis (Z) configuration, and usually, they are of even carbons ranging from C12 to C26. The lipids from neuronal membranes contribute in lipid peroxidation, and it is a free radical chain reaction in which lipids hydrocarbon reacts with the free OH˙ radical formed via Fenton’s reaction to eliminate H from a fatty acid side-chain thereby forming a carbon radical. Furthermore, the oxidation of carbon radical further forms peroxyl radicals that damage the membrane proteins, and the fatty acid side-chains. Again, the peroxyl radical reacts with hydrocarbon to form the lipid hyperperoxide, and also a carbon radical; hence, it retains a free radical and forms continuous chain reaction (Figure 2). As reported, the unsaturation of fatty acid’s side chain is essential for fluidity, but also it is very susceptible to free radicals (Halliwell, 2015). Patients suffering from OS condition were found to be more prone to excrete aldehydes, like 4-hydroxynonenal, and malondialdehyde along with thiobarbituric acid-reactive substances in their urine which are the by-products of the lipid peroxidation.

Mechanism of lipid peroxidation via OH free radicals formed during oxidative stress.
The proteins also play an important role in strengthening and sensory communication among the neurons. The molecules embedded in the membrane, e.g. ion channels, and receptors are responsible for communication between inside and outside located neurons. The actin, spectrins, neurofilaments and microtubules forming the cytoskeleton assist the neurotransmitter release and another regulatory mechanism as part of their functions. The damage caused by the OS to proteins results in their irregular functions. The OH free radical directly attacks the α-carbon of the amino acid chains via extracting hydrogens to form water and leaving the carbon radical. These carbon radicals are responsible for cross-linking. The carbon radicals are easily oxidized into peroxides, and Fenton’s reaction, which changes Fe+2 to Fe+3 with the production of a proton from the conversion of hydrogen dioxide radical to oxygen, is an important initiation. Thus, the produced proton reacts with the peroxides forming the hydrogen peroxides. Further, the dehydroxylation from hydrogen peroxides of amino acid takes place via the oxidation of Fe+2 to Fe+3 resulting in the formation of amino acid oxides. These oxides are responsible for peptide bond cleavage. The hyperperoxides and carbonyls proteins are the major products of protein peroxidation and oxidation, which are accumulated in the brain regions of the oxidatively stress patients, and are excreted in their urine (Figure 3).

Mechanism of protein damage via OH free radicals formed during oxidative stress.
The DNA copying errors and the faulty RNA transcripts lead to protein damage. The proteins’ sequences and shapes are dependent upon the genes (sequence of DNA), which determines the sequence of amino acid in a given polypeptide chain. The ambiguous base pair mismatch results in change of amino acids leading to an ambiguous translation. The hydroxyl free radical assists the nitrogen bases to form hydroxyl compounds. The purines nitrogenous bases, i.e. guanine, and adenine mostly form the 8-hydroxy products, whereas the pyrimidine nitrogenous bases, i.e. the cytosine, and thymine form 5-hydroxy or 4,5-dihydroxy product, e.g. thymine glycol. Also, a very common somatic mutation is found among guanosine and thymidine, achieved by the hydroxylation of guanosine at the eighth position, which further goes to keto-enol tautomerism, mimicking the thymidine. During DNA replication, the 8-hydroxyguanosine complements the adenosine-containing template. The rate of this mutation depends upon the proportion of oxidized DNA damage resulting in the base pair mismatch. The mismatched pairing leads to translation of abnormal proteins (Figure 4). This whole mechanism also slows down the transcription and the translation (Cutler and Rodrigues, 2002; Maehara et al., 2008; Moreira et al., 2008). In the past researches, the elevated concentration of 8-hydroxy deoxyguanosine (8-OHdG) was established as a biomarker for DNA damage in animals kept under OS conditions. The same pattern of 8-hydroxy deoxyguanosine (8-OHdG) and damaged nitrogenous base products have been found in the urine of smokers and rheumatoid arthritis patients (Nunomura et al., 2007), and have also been debated as cancer risk (Halliwell 1998).

Mechanism of DNA damage via OH free radicals formed by the oxidative stress.
Generally, the nerve cells are incapable of division on maturity, which restricts them to a constant number of populations inside the fixed volume of the cranial capacity. The learned skills depend upon the interconnection of neurons and the loss of neurons or interruption in its function leads to several neurological disorders, prominent among them are amyotrophic lateral sclerosis, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, depression and epilepsy. The OS, directly or, indirectly, influences the pathogenesis of these diseases.
Amyotrophic lateral sclerosis
The amyotrophic lateral sclerosis (ALS) is characterized by dysfunction of motor neurons. It is designated as a chronic neurodegenerative disorder due to damage of the motor neurons in motor cortex, brain stem, corticospinal tract and spinal cord leading to muscle dimness (Neymotin et al., 2009). The factors responsible for the ALS pathogenesis are OS, cytoskeletal abnormalities, mitochondrial dysfunction and protein aggregation (Shaw, 2005). Around 20% of amyotrophic lateral sclerosis cases are found to be caused by the mutation of genes in the enzyme Cu-Zn superoxide dismutase (SOD1). Until date, a number of mutations have been reported, among them, H46R (characterize by the replacement of histidine to arginine at codon 46), A4V (replacement of alanine to valine at codon 4) and G93A (characterized by the replacement of glycine to alanine at codon 93) are well-known mutations (Vargas et al., 2011; Bhattacharya et al., 2012; Pan et al., 2012). The SOD1 imparts an active role in the endogenous antioxidant system by protecting the body from the damage caused by the highly reactive superoxide free radical produced by the mitochondrion during OS. Similar results of impairment of calcium transport, electron transport system and ROS production have been confirmed in a study on SOD1 mutated mice (Mattiazzi et al., 2002; Lin and Beal, 2006). The increased mutants SOD1 are responsible for the impairment of cellular activities by damaging the proteasomes, mitochondria and chaperones biomolecules (Boillée et al., 2006).
Alzheimer’s disease
Alzheimer’s disease (AD) is characterized by cognitive dysfunctions, memory loss with irreversible pathological condition. Unfortunately, it is followed by vascular dementia (Ashraf et al., 2016). In this disease, the neuronal cells gradually and over time degenerate and allow the disorder to take control due to extracellular amyloid beta (Aβ) depositions, intracellular neurofibrillary tangles (NFTs) retention and neuronal decline, which leads to neurotransmitter system derangement for the patient. The receptors acetylcholine (Bourin et al., 2003; Kihara and Shimohama, 2004; Zuchner et al., 2005; Oddo and LaFerla, 2006; Barrantes et al., 2010), N-methyl d-aspartate (NMDA), adenosine, histamine, insulin (Islam et al., 2017a,b), glutamate (Anggono et al., 2016), serotonin (Butzlaff and Ponimaskin, 2016) and others (Chu et al., 1987; Ramirez, 2013; Jin et al., 2015; Su et al., 2016; Zhao et al., 2017) are responsible. The affected subjects have also shown neuronal damage at the parietal lobe, temporal lobe, cingulate gyrus, frontal cortex and subcortical regions of the brain (Orth and Schapira, 2001; Wenk, 2006). The histopathological observations revealed the presence of masses of hyperphosphorylated tau protein (neurofibrillary tangles) and β-amyloid plaques (derived from the amyloid precursor protein) in the neuronal cytoplasm (LaFerla and Oddo, 2005; Anandatheerthavarada and Devi, 2007; Duyckaerts et al., 2009). The oxygen radical expedites the formation of glycated end products, lipid peroxidation, nitration, adduction products, carbonyl-modified neurofilament protein and free carbonyls (Smith et al., 1994; Montine et al., 1996; Smith et al., 1997). The glycated products are mainly linked to accumulation of β-amyloid and NFT deposits (Smith et al., 1994). The mitochondria are said to be the major source of oxidative radicals, and the major precursors are superoxide free radicals, i.e. O2˙− and H2O2. In an in situ hybridization study, the chimeric cDNA involvement and common deletion of 5 kb 3× folds increased the mitochondrion mutation as compared to control. The imbalances of trace elements and redox-active metals were seen in AD encountering among them the Cu (II), Zn (II) and Fe (II), which play an important role in the OS (Smith et al., 1994, 1995). A microplate X-ray emission study revealed higher concentrations of these metals in neuropil as well in the NFT and senile plaques of AD subjects (Lovell et al., 1998), thus confirming the earlier connotation concerning the role of iron, ferritin, transferrin and redox active iron in the mechanism of NFT and the senile plaques formation in AD patients (Perez et al., 1998). Certain proteins and sirtuins, secretase, lipoxygenases, glycogen synthase kinase 3 (GSK-3), acetylcholinesterase and caspases also play part towards pathogenesis of the disease (Islam and Tabrez, 2017; Islam et al., 2017a,b). The inflammation and insulin-based studies have lately provided the disease’s connection with the type 2 diabetes (Kamal et al., 2014), whereas the nitric oxide-dependent mitochondrial DNA overproliferation has been linked to the cancer (Aliev et al., 2013). Importantly, the inhibitors acting against the involved these enzymes and the targeting of the OS, auto-phagocytosis biomechanistics, as well as RNA interference can have primal role in developing suitable treatment and containment strategy for the disease (Jabir et al., 2014; Islam et al., 2017a,b). Recently, piperine based solid lipid nanoparticles were evaluated with successful results against experimentally OS induced Alzheimer’s disease by ibotenic acid (Yusuf et al., 2013).
Huntington’s disease
Huntington’s disease (HD) is a genetic neurodegenerative disorder inherited in an autosomal dominant pattern and usually has an adult onset of the disease in thirties or late thirties of the person. It is characterized by emotional problems, depression and loss of cognition, trouble in speaking, swallowing, uncontrolled and jerking movements termed as chorea with changes in personality. A juvenile form may start in early childhood and adolescence, and is exhibited by movement disorders, frequent falling, drooling, emotional changes and slurred talk (Walker, 2007). It severely affects a person’s cognitive, functional and psychiatric abilities (DiFiglia, 1990). The disease involves neuron degradation in striatum via autosomal dominant mutation of the Huntingtin (Htt) gene. This gene has more than 36 (up to 120, 40 being the threshold) repetitions of CAG (cytosine, adenine and guanine) trinucleotide repeat for glutamine amino acid (CAGCAG.....CAG), which codes for a mutant huntingtin protein. The methylation of DNA is also altered (Glajch and Sadri-Vakili, 2015). The huntingtin protein is involved in multiple proteins interactions and a larger set of biological functions. It is toxic to brain cells and with disease progression affects large part of the brain (Goehler et al., 2004). On a biomechanistics scale, a higher oxidative damage in HD leads to the DNA strands breaking down (Hersch et al., 2006), an increase in unsaturated fatty acid per-oxidations (Browne et al., 1999) with increased oxidation of proteins as well as lipids (Browne and Beal, 2006) and an impaired SOD activity (Santamaría et al., 2001; Sorolla et al., 2008). The mutant huntingtin (Htt) can effectively cause mitochondrial dysfunction via decreased mitochondrial enzyme activity. Other studies have proposed that mtDNA is the key target of mutant Htt-linked OS, probably leading to mitochondrial alteration, and the impairment of the apurinic/apyrimidinic (AP) endonuclease enzyme 1 (APE1) that is involved in the base excision repair (BER) pathway, which is one of the important targets in mitochondrial activity in HD patients (Oliveira and Lightowlers, 2010; Ayala-Pena, 2013). In other studies, the mutant Htt-expressing cells have showed reduced APE1 with reduced respiratory functions in comparison with control (Siddiqui et al., 2012). It has also increased the Ca2+-mediated ROS generation (Wang et al., 2013) with greater mtDNA damages. The implications of these biochemical processes provide an insight into the disease where no permanent cure is available but only symptomatic improving of conditions is possible.
Parkinson’s disease
Parkinson’s disease (PD) is a progressive degeneration of dopaminergic neurons in the substantia nigra and pars compacta (Braak et al., 2003; Hirsch et al., 2013). It is characterized by Lewy bodies’ inclusions and α-synuclein accumulation in the neurons. Several gene mutations are reported in leucine-rich repeat kinase 2 (LRRK2), α-synuclein, DJ-1, ATP13A2 and PINK1 (PTEN-induced putative kinase 1) (Biskup et al., 2008) that are involved. The oxidative damage induced mitochondrial alteration, mtDNA mutations and mitochondrial dysfunctions can aggravate the degeneration of dopaminergic neurons (Jin et al., 2014). The OS-mediated modification of α-synuclein by oxidation and nitration leads to oligomerization (Xiang et al., 2013; Shimoji et al., 2013). The antioxidant therapy could protect Parkinson’s disease by reducing OS as demonstrated in an in vitro study where ascorbic acid implementation decreased the death rate of dopaminergic neurons (Ballaz et al., 2013). The low ascorbic acid levels were found in the lymphocytes of PD (Ide et al., 2015). In other studies, it was concluded that dopamine self-metabolizes to produce ROS, which could aggravate the PD condition (Bindoff et al., 1989; Mizuno et al., 1989; Schapira et al., 1989; Henchcliffe and Beal, 2008). Many studies have proposed that the mitochondrial dysfunction is linked to PINK1. The disproportionate mitochondrial fission also obstructs the maintenance of oxidative phosphorylation (OP) mechanism with PINK1 losses, which results in defective OP complex assembly leading to reduced mitochondrial OP (Liu et al., 2011; Tufi et al., 2014). In another study, an increased mtDNA deletion was found associated with ETC (electron transport chain) paucity in substantia nigra neurons (Kraytsberg et al., 2006). Similar findings have been observed in PD-mito-Pst1 mouse, which expressed a restriction enzyme for damaging mtDNA (Pickrell et al., 2011). OS and tyrosine hydroxylase have been suggested as the mechanistic and molecular targets for developing the cure for the disease (Khan et al., 2012).
Epilepsy
Epilepsy is a chronic neurological disorder, characterized by recurrent and unprovoked seizures, which are proposed to be caused by an imbalance of oxidants, and antioxidants because of the excitotoxic OS (Shivakumar et al., 1991). A mathematical model have also been proposed (Rai and Khan, 2009; Khan and Rai, 2016). The human brain, rich in mitochondrion, generates highly reactive superoxide radicals during the respiratory cascade, which efficiently starts the pathological oxidative metabolism of bio-macromolecules leading to seizures (Turrens et al., 1982; Liang and Patel, 2006). Numerous findings concluded that an increase in mitochondrial O and NS (oxidative and nitrosative stress) followed by cell impairment causes persistent seizures (Cock, 2002; Liang and Patel, 2006; Waldbaum et al., 2010). It has also been reported that extended seizures can result in the production of superoxides by a series of mechanism initiated by extreme neuronal firing, ATP consumption, NMDA receptor activation, glutamate release, mitochondrial and cytosolic calcium influx (Patel, 2004). The mitochondrial dysfunction influences epilepsy in humans (Kunz and Oliw, 2001; Lee et al., 2008) and animal models (Cock et al., 2002; Kudin et al., 2002; Chuang et al., 2004; Liang and Patel, 2004) as a consequence of faulty oxidative phosphorylation complexes, which results into excessive superoxide production. In a previous study, reductive iron salts (Willmore et al., 1978) and toxins (Zuchora et al., 2001) enhanced the free radical load and seizure activity. On the other hand, an increased superoxide production was found in Mn-SOD mice via synaptic NMDA receptor activation, which also resulted in seizure activity (Melov et al., 1998). In status epilepticus (SE), a variation in redox potential with a drop of ATP was encountered, which resulted in a downfall of production and supply of energy to the brain (Wasterlain et al., 1993). The myoclonic epilepsy with ragged red fibers (MERRF) is a rare mutation syndrome caused by the transition mutation of A to G (A8344G mutation) in human mitochondrial DNA (Wallace et al., 1988), and it is said to be responsible for impaired biosynthesis of mitochondrial proteins that are linked with the oxidative phosphorylation (Shoffner et al., 1990). The excessive ROS generation, impaired expression of antioxidant enzymes and insufficient ATP generation are some of the consequences reported in MERRF (Wu et al., 2010). Other mitochondrial mutations involved are DNA polymerase γ (POLG1) (Zsurka et al., 2008) and tRNAPhe (phenylalanine-tRNA ligase) MT-TF (Mitochondrially Encoded TRNA Phenylalanine, RNA gene) (Zsurka et al., 2010) which are reported to be associated with generalized seizures, and affects the mitochondrial respiratory cascade along with ATP production (Zsurka and Kunz, 2010).
In animal models, kainic acid (KA), an epileptogenic, caused an increase in the ROS and NO syntheses, mitochondrial dysfunction and apoptosis of neurons when it was directly administered into CA3 area of the hippocampus (Liang et al., 2000; Chuang et al., 2007; Shin et al., 2011), whereas other KA-induced seizures reduced the activity of the nicotinamide adenine dinucleotide cytochrome c reductase (NCCR) in the hippocampus (Waldbaum and Patel, 2010). The mitochondrial OS can cause oxidative damage to DNA at different stages of seizures as elicited by pilocarpine or KA (Waldbaum et al., 2010). The pilocarpine-induced seizures have also been reported for enhancing the lipid peroxidation, ROS and nitrite production in the hippocampal, striatum and frontal cortex (Folbergrová and Kunz, 2012). The OS connection of epilepsy was substantiated by eliciting seizures via removal of the MnSOD enzyme in comparison with the MnSOD (SOD2) super-expressed animals, and it showed improved survival in comparison to KA-induced SE (Milder and Patel, 2012). The antioxidants, e.g. melatonin, and vitamin C-SOD mimic had shown prevention in experimentally induced seizures. A daily supplementation of coenzyme Q10 reduces the SE-induced RNA oxidation and protects individual from neuronal loss (Kong and Lin, 2010). Another antioxidant, β-carotene, had shown decent prevention of seizure as observed in the brain-targeted polysorbate-80-coated PLGA nanoparticles delivery in MES (maximal electroshock seizure), and PTZ (pentylenetetrazol) induced albino mice epileptics (Yusuf et al., 2012). In another study, higher concentrations of ceruloplasmin and lower concentrations of vitamins C, E and A were found epileptics in comparison to the controls (Sudha et al., 2001).
Major depressive disorder
Major depressive disorder (MDD) or clinical depression is the common mental disorder extending over 2 weeks in nearly all situations, resulting in behavioral changes, ideas of self-harm and suicide, low self-esteem and low energy, and often pain without any clear cause (Kessler et al., 2003; Mehlum et al., 2016). The role of oxidative disturbances is evidenced in the MDD, which was observed through oxidative marker studies and antioxidant effects of antidepressants. The oxidative stress biomarkers, such as increased serum levels of XO (xanthine oxidase) in thalamus (Herken et al., 2006; Leonard and Maes, 2012), the elevated levels of MAO (monoamine oxidase) (Sacher et al., 2015), SOD and catalase (Scapagnini et al., 2012; de Sousa et al., 2014) and the lowered activity of GPX (glutathione peroxidase) (Maes et al., 2010) were found in this disorder (Khanzode et al., 2003). The elevated ROS/RNS (reactive oxygen and nitrogen species) and suppressed antioxidant defenses also resulted in oxidative and nitrosative modifications of cellular biomolecules, i.e. fatty acids, proteins and DNA.
The telomerase with critical role-play instability of genomic sequence is susceptible to oxidative damage. The telomere shortening owing to oxidative damages to a critical length, as found in depression and oxidative stress, is presumed to be a prime cause. OS can induce DNA damages in telomeres in comparison to non-telomeric DNAs (Simon et al., 2006; Wolkowitz et al., 2011). The oxidative deamination of monoamine neurotransmitters by MAO produces hydrogen peroxide as the by-product which overloads the ROS, trails to mitochondrial dysfunction and neuron apoptosis (Scapagnini et al., 2012). The GPX protects DNA, and neuronal damages in MMD subjects where decreased activity of GPX with excessive ROS accumulation have been found (Kodydková et al., 2009; Gawryluk et al., 2011). A lower activity of paraoxonase 1 (PON1) was found in MDD patients; this enzyme protects lipid peroxidation of lipids (Aviram et al., 1998). The improved results were observed in MDD experimental models while using polyenes and polyphenol antioxidants in pure or, different delivery doses form like curcumin (Yusuf et al., 2016), resveratrol, chlorogenic acid and Ginkgo biloba EGb761 (Gomez-Pinilla and Nguyen, 2012; Ogle et al., 2013). The lower concentrations of omega-3 polyunsaturated fatty acids (n-3 PUFA) (Gharekhani et al., 2014), coenzyme Q10 (CoQ10) (Schmelzer et al., 2008) and N-acetyl cysteine (NAC) (Berk et al., 2014) were found in MDD patients as low levels of CoQ10 is linked with higher concentrations of ROS, RNS and tumor necrosis factor alpha with mitochondrion dysfunctions.
Conclusions
The oxygen consumption by cells for their energy and other physiological needs generates free radicals in the biochemical processes which in uncontrolled abundance damages the cells through various reactions and involvements in the physiology and biochemical functioning of the cells. The higher oxygen demands and the presence of susceptible substrates make the human brain more vulnerable than other organs and tissues in the body. The resulting redox imbalances through excess ROS and/or dysfunctional natural antioxidant systems of the body, slowly and over time, lead to pathogenesis of neurodegenerative disorders due to OS’s continual pressing. The roles of hereditary and non-hereditary factors are other concerns in this context. The biotherapies and better drug designing based on curtailing the roles and controls of the OS generators, biochemical steps propagating biomolecular entities, the free-radical conveyors in biochemical steps along with the vulnerable biological steps as well as the involved biomechanistics will shed more light on precise and focused causes, biochemical, enzymatic processing and the target entities to provide realistic goals for better drug design and development of drugs with elaborate and understood use of natural products, natural products modified by design, and synthetic agents in curtailing, controlling and curing the diseases.
Conflict of interest statement: The authors have no conflict of interests.
References
Adibhatla, R.M. and Hatcher, J.F. (2007). Role of lipids in brain injury and diseases. Future Lipidol. 2, 403–422.10.2217/17460875.2.4.403Suche in Google Scholar PubMed PubMed Central
Adibhatla, R.M., Hatcher, J.F., and Dempsey, R.J. (2006). Lipids and lipidomics in brain injury and diseases. AAPS J. 8, E314–E321.10.1007/BF02854902Suche in Google Scholar PubMed PubMed Central
Aliev, G., Obrenovich, M.E., Tabrez, S., Jabir, N.R., Reddy, V.P., Li, Y., Burnstock, G., Cacabelos, R., and Kamal, M.A. (2013). Link between cancer and Alzheimer disease via oxidative stress induced by nitric oxide-dependent mitochondrial DNA over-proliferation and deletion. Oxid. Med. Cell. Longev. 2013, 962984.10.1155/2013/962984Suche in Google Scholar PubMed PubMed Central
Allsop, D., Mayes, J., Moore, S., Masad, A., and Tabner, B.J. (2008). Metal-dependent generation of reactive oxygen species from amyloid proteins implicated in neurodegenerative disease. Biochem. Soc. Trans. 36, 1293–1298.10.1042/BST0361293Suche in Google Scholar PubMed
Anandatheerthavarada, H.K. and Devi, L. (2007). Amyloid precursor protein and mitochondrial dysfunction in Alzheimer’s disease. Neurosci. 13, 626–638.10.1177/1073858407303536Suche in Google Scholar PubMed
Anggono, V., Li-Huei T., and Götz, J. (2016). Glutamate receptors in Alzheimer’s disease: mechanisms and therapies. Neural Plast. 2016, 8256196.10.1155/2016/8256196Suche in Google Scholar PubMed PubMed Central
Aseervatham, G.S., Sivasudha, T., Jeyadevi, R., and Arul Ananth, D. (2013). Environmental factors and unhealthy lifestyle influence oxidative stress in humans – an overview. Environ. Sci. Poll. Res. Intern. 20, 4356–4369.10.1007/s11356-013-1748-0Suche in Google Scholar PubMed
Ashraf, G.M., Chibber, S., Mohammad, Zaidi, S.K., Tabrez, S., Ahmad, A., Shakil, S., Mushtaq, G., Baeesa, S.S., and Kamal, M.A. (2016). Recent updates on the association between alzheimer’s disease and vascular dementia. Med. Chem. 12, 226–237.10.2174/1573406411666151030111820Suche in Google Scholar PubMed
Auscher, C., Amory, N., van der Kemp, P., and Delbarre, F. (1979). Xanthine oxidase activity in human intestines. Histochemical and radiochemical study. Adv. Exp. Med. Biol. 122b, 197–201.10.1007/978-1-4684-8559-2_33Suche in Google Scholar PubMed
Aviram, M., Rosenblat, M., Bisgaier, C.L., Newton, R.S., Primo-Parmo, S.L. and La Du, B.N. (1998). Paraoxonase inhibits high-density lipoprotein oxidation and preserves its functions. A possible peroxidative role for paraoxonase. J. Clin. Invest. 101, 1581.10.1172/JCI1649Suche in Google Scholar PubMed PubMed Central
Ayala-Pena, S. (2013). Role of oxidative DNA damage in mitochondrial dysfunction and Huntington’s disease pathogenesis. Free Radic. Biol. Med. 62, 102–110.10.1016/j.freeradbiomed.2013.04.017Suche in Google Scholar
Ballaz, S., Morales, I., Rodríguez, M., and Obeso, J.A. (2013). Ascorbate prevents cell death from prolonged exposure to glutamate in an in vitro model of human dopaminergic neurons. J. Neurosci. Res. 91, 1609–1617.10.1002/jnr.23276Suche in Google Scholar
Barber, S.C., Mead, R.J., and Shaw, P.J. (2006). Oxidative stress in ALS: a mechanism of neurodegeneration and a therapeutic target. Biochim. Biophys. Acta Mol. Basis Dis. 1762, 1051–1067.10.1016/j.bbadis.2006.03.008Suche in Google Scholar
Barrantes, F.J., Borroni, V., and Vallés, S. (2010).Neuronal nicotinic acetylcholine receptor-cholesterol crosstalk in Alzheimer’s disease. FEBS Lett. 584, 856–863.10.1016/j.febslet.2009.11.036Suche in Google Scholar
Berk, M., Dean, O.M., Cotton, S.M., Jeavons, S., Tanious, M., Kohlmann, K., Hewitt, K., Moss, K., Allwang, C., and Schapkaitz, I. (2014). The efficacy of adjunctive N-acetylcysteine in major depressive disorder: a double-blind, randomized, placebo-controlled trial. J. Clin. Psych. 75, 628–636.10.4088/JCP.13m08454Suche in Google Scholar
Bhattacharya, A., Bokov, A., Muller, F.L., Jernigan, A.L., Maslin, K., Diaz, V., Richardson, A., and Van Remmen, H. (2012). Dietary restriction but not rapamycin extends disease onset and survival of the H46R/H48Q mouse model of ALS. Neurobiol. Aging 33, 1829–1832.10.1016/j.neurobiolaging.2011.06.002Suche in Google Scholar
Bindoff, L., Birch-Machin, M., Cartlidge, N., Parker, W., and Turnbull, D. (1989). Mitochondrial function in Parkinson‘s disease. Lancet 334, 49.10.1016/S0140-6736(89)90291-2Suche in Google Scholar
Biskup, S., Gerlach, M., Kupsch, A., Reichmann, H., Riederer, P., Vieregge, P., Wüllner, U., and Gasser, T. (2008). Genes associated with Parkinson syndrome. J. Neurol. 255, 8–17.10.1007/s00415-008-5005-2Suche in Google Scholar PubMed
Boillée, S., Vande Velde, C., and Cleveland, D.W. (2006). ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron. 52, 39–59.10.1016/j.neuron.2006.09.018Suche in Google Scholar PubMed
Bourin, M., Ripoll, N., and Dailly, E. (2003). Nicotinic receptors and Alzheimer’s disease. Curr. Med. Res. Opin. 19, 169–177.10.1185/030079903125001631Suche in Google Scholar PubMed
Braak, H., Del Tredici, K., Rüb, U., de Vos, R.A., Steur, E.N.J., and Braak, E. (2003). Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 24, 197–211.10.1016/S0197-4580(02)00065-9Suche in Google Scholar
Browne, S.E. and Beal, M.F. (2006). Oxidative damage in Huntington’s disease pathogenesis. Antioxd. Redox Signal. 8, 2061–2073.10.1089/ars.2006.8.2061Suche in Google Scholar
Browne, S.E., Ferrante, R.J., and Beal, M.F. (1999). Oxidative stress in Huntington’s disease. Brain Pathol. 9, 147–163.10.1111/j.1750-3639.1999.tb00216.xSuche in Google Scholar
Butzlaff, M. and Ponimaskin, E. (2016). The role of serotonin receptors in Alzheimer’s disease. Opera. Med. Physiol. 2, 77–86.Suche in Google Scholar
Carrard, G., Bulteau, A.L., Petropoulos, I., and Friguet, B. (2002). Impairment of proteasome structure and function in aging. Int. J. Biochem. Cell Bio. 34, 1461–1474.10.1016/S1357-2725(02)00085-7Suche in Google Scholar
Chu, D.C.M., Penney, J.B., and Young, A.B. (1987). Cortical GABA-B and GABA-A receptors in Alzheimer’s disease: a quantitative autoradiographic study. Neurology, 37, 1454.10.1212/WNL.37.9.1454Suche in Google Scholar
Chuang, Y.C., Chang, A.Y.W., Lin, J.W., Hsu, S.P., and Chan, S.H.H. (2004). Mitochondrial dysfunction and ultrastructural damage in the hippocampus during kainic acid-induced status epilepticus in the rat. Epilepsia 45, 1202–1209.10.1111/j.0013-9580.2004.18204.xSuche in Google Scholar
Chuang, Y., Chen, S., Lin, T., Liou, C., Chang, W., Chan, S., and Chang, A. (2007). Upregulation of nitric oxide synthase II contributes to apoptotic cell death in the hippocampal CA3 subfield via a cytochrome c/caspase-3 signaling cascade following induction of experimental temporal lobe status epilepticus in the rat. Neuropharmacology 52, 1263–1273.10.1016/j.neuropharm.2007.01.010Suche in Google Scholar
Clarke, D. and Sokoloff, L. (1999). Circulation and energy metabolism of the brain. (Philadelphia, USA: Lippincott-Raven).Suche in Google Scholar
Cock, H.R. (2002). The role of mitochondria and oxidative stress in neuronal damage after brief and prolonged seizures. Prog. Brain Res. 135, 187–196.10.1016/S0079-6123(02)35018-0Suche in Google Scholar
Cock, H.R., Tong, X., Hargreaves, I.P., Heales, S.J.R., Clark, J.B., Patsalos, P.N., Thom, M., Groves, M., Schapira, A.H.V., and Shorvon, S.D. (2002). Mitochondrial dysfunction associated with neuronal death following status epilepticus in rat. Epilepsy. Res. 48, 157–168.10.1016/S0920-1211(01)00334-5Suche in Google Scholar
Cutler, R.G. and Rodrigues, H. (Ed.). (2002). Critical Reviews of Oxidative Stress and Ageing: Advances in Basic Science, Diagnostics and Intervention. ISBN: 978-981-4490-94-8 (ebook), World Scientific Publishing Company. UK.10.1142/4714Suche in Google Scholar
Danial, N.N. and Korsmeyer, S.J. (2004). Cell death: critical control points. Cell 116, 205–219.10.1016/S0092-8674(04)00046-7Suche in Google Scholar
de Sousa, R.T., Zarate, C.A., Zanetti, M.V., Costa, A.C., Talib, L.L., Gattaz, W.F., and Machado-Vieira, R. (2014). Oxidative stress in early stage bipolar disorder and the association with response to lithium. J. Psych. Res. 50, 36–41.10.1016/j.jpsychires.2013.11.011Suche in Google Scholar
DiFiglia, M. (1990). Excitotoxic injury of the neostriatum: a model for Huntington’s disease. Trends Neurosci. 13, 286–289.10.1016/0166-2236(90)90111-MSuche in Google Scholar
Droge, W. (2002). Free radicals in the physiological control of cell function. Physiol. Rev. 82, 47–95.10.1152/physrev.00018.2001Suche in Google Scholar PubMed
Duyckaerts, C., Delatour, B., and Potier, M.-C. (2009). Classification and basic pathology of Alzheimer disease. Acta Neuropathol. 118, 5–36.10.1007/s00401-009-0532-1Suche in Google Scholar PubMed
Eghiaian, F., Grosclaude, J., Lesceu, S., Debey, P., Doublet, B., Tréguer, E., Rezaei, H., and Knossow, M. (2004). Insight into the PrP(C) → PrP(Sc) conversion from the structures of antibody-bound ovine prion scrapie-susceptibility variants. Proc. Natl. Acad. Sci. USA 101, 10254–10259.10.1073/pnas.0400014101Suche in Google Scholar PubMed PubMed Central
Emerit, J., Edeas, M., and Bricaire, F. (2004). Neurodegenerative diseases and oxidative stress. Biomed. Pharmacother. 58, 39–46.10.1016/j.biopha.2003.11.004Suche in Google Scholar PubMed
Engel, T. and Henshall, D.C. (2009). Apoptosis, Bcl-2 family proteins, and caspases: the ABCs of seizure-damage and epileptogenesis. Int. J. Physiol. Pathophysiol. Pharmacol. 1, 97–115.Suche in Google Scholar
Folbergrová, J. and Kunz, W.S. (2012). Mitochondrial dysfunction in epilepsy. Mitochondrion. 12, 35–40.10.1016/j.mito.2011.04.004Suche in Google Scholar PubMed
Gawryluk, J.W., Wang, J.-F., Andreazza, A.C., Shao, L., and Young, L.T. (2011). Decreased levels of glutathione, the major brain antioxidant, in post-mortem prefrontal cortex from patients with psychiatric disorders. Int. J. Neuropsychopharmacol. 14, 123–130.10.1017/S1461145710000805Suche in Google Scholar PubMed
Gharekhani, A., Khatami, M.-R., Dashti-Khavidaki, S., Razeghi, E., Noorbala, A.-A., Hashemi-Nazari, S.-S., and Mansournia, M.-A. (2014). The effect of ω-3 fatty acids on depressive symptoms and inflammatory markers in maintenance hemodialysis patients: a randomized, placebo-controlled clinical trial. Eur. J. Clin. Pharmacol. 70, 655–665.10.1007/s00228-014-1666-1Suche in Google Scholar PubMed
Gibson, A. and Lilley, E. (1997). Superoxide anions, free-radical scavengers, and nitrergic neurotransmission. General Pharmacol. 28, 489–493.10.1016/S0306-3623(96)00281-9Suche in Google Scholar
Gil-Mohapel, J., Brocardo, P.S., and Christie, B.R. (2014). The role of oxidative stress in Huntington’s disease: are antioxidants good therapeutic candidates? Curr. Drug Targ. 15, 454–468.10.2174/1389450115666140115113734Suche in Google Scholar
Girgin, S.F., Sozmen, E.Y., Ersoz, B., and Mentes, G. (2004). Link between monoamine oxidase and nitric oxide. NeuroToxicol. 25, 91–99.10.1016/S0161-813X(03)00089-5Suche in Google Scholar
Glajch, K.E. and Sadri-Vakili, G. (2015). Epigenetic mechanisms involved in Huntington’s disease pathogenesis. J. Huntington’s Dis. 4, 1–15.10.3233/JHD-140134Suche in Google Scholar
Goehler, H., Lalowski, M., Stelzl, U., Waelter, S., Stroedicke, M., Worm, U., Droege, A., Lindenberg, K.S., Knoblichm M., Haenig, C., et al. (2004). A protein interaction network links GIT1, an enhancer of Huntingtin aggregation, to Huntington’s disease. Mol. Cell 15, 853–865.10.1016/j.molcel.2004.09.016Suche in Google Scholar PubMed
Gomez-Pinilla, F. and Nguyen, T.T. (2012). Natural mood foods: the actions of polyphenols against psychiatric and cognitive disorders. Nutri. Neurosci. 15, 127–133.10.1179/1476830511Y.0000000035Suche in Google Scholar PubMed PubMed Central
Granger, D.N. and Kvietys, P.R. (2015). Reperfusion injury and reactive oxygen species: the evolution of a concept. Redox Biol. 6, 524–551.10.1016/j.redox.2015.08.020Suche in Google Scholar PubMed PubMed Central
Guo, C., Sun, L., Chen, X., and Zhang, D. (2013). Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen. Res. 8, 2003–2014.Suche in Google Scholar
Halliwell B. (1998). Can oxidative DNA damage be used as a biomarker of cancer risk in humans? Problems, resolutions, and preliminary results from nutritional supplementation studies. Free Radic. Res. 29, 469–486.10.1080/10715769800300531Suche in Google Scholar PubMed
Halliwell, B. (2015). Free Radicals and Other Reactive Species in Disease. (NY, USA: John Wiley & Sons), eLS. DOI: 10.1038/npg.els.0003913.10.1038/npg.els.0003913Suche in Google Scholar
Halliwell, B. and Gutteridge, J.M.C. (1999). Free Radicals in Biology and Medicine, Vol 135 (Oxford, UK: Oxford University Press).Suche in Google Scholar
Henchcliffe, C. and Beal, M.F. (2008). Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis. Nat. Clin. Practice Neurol. 4, 600–609.10.1038/ncpneuro0924Suche in Google Scholar PubMed
Hengartner, M.O. (2000). The biochemistry of apoptosis. Nature 407, 770–776.10.1038/35037710Suche in Google Scholar PubMed
Henshall, D.C. and Engel, T. (2013). Contribution of apoptosis-associated signaling pathways to epileptogenesis: lessons from Bcl-2 family knockouts. Neuronal mechanisms of epileptogenesis. Front Cell. Neurosci. 7, 110.10.3389/fncel.2013.00110Suche in Google Scholar PubMed PubMed Central
Herken, H., Akyol, O., Yilmaz, H.R., Tutkun, H., Savas, H.A., Ozen, M.E., Kalenderoglu, A., and Gulec, M. (2006). Nitric oxide, adenosine deaminase, xanthine oxidase and superoxide dismutase in patients with panic disorder: alterations by antidepressant treatment. Hum. Psychopharmacol. Clin. Exp. 21, 53–59.10.1002/hup.742Suche in Google Scholar PubMed
Hersch, S., Gevorkian, S., Marder, K., Moskowitz, C., Feigin, A., Cox, M., Como, P., Zimmerman, C., Lin, M., and Zhang, L. (2006). Creatine in Huntington disease is safe, tolerable, bioavailable in brain and reduces serum 8OH2′ dG. Neurology 66, 250–252.10.1212/01.wnl.0000194318.74946.b6Suche in Google Scholar PubMed
Hirsch, E.C., Jenner, P., and Przedborski, S. (2013). Pathogenesis of Parkinson’s disease. Move Disord. 28, 24–30.10.1002/mds.25032Suche in Google Scholar PubMed
Hui, J., Zhang, Z.J., Zhang, X., Shen, Y., and Gao, Y.J. (2013). Repetitive hyperbaric oxygen treatment attenuates complete Freund’s adjuvant-induced pain and reduces glia-mediated neuroinflammation in the spinal cord. J. Pain 14, 747–758.10.1016/j.jpain.2013.02.003Suche in Google Scholar PubMed
Hwang, O. (2013). Role of oxidative stress in Parkinson’s disease. Exp. Neurobiol. 22, 11–17.10.5607/en.2013.22.1.11Suche in Google Scholar PubMed PubMed Central
Ide, K., Yamada, H., Umegaki, K., Mizuno, K., Kawakami, N., Hagiwara, Y., Matsumoto, M., Yoshida, H., Kim, K., and Shiosaki, E. (2015). Lymphocyte vitamin C levels as potential biomarker for progression of Parkinson’s disease. Nutrition 31, 406–408.10.1016/j.nut.2014.08.001Suche in Google Scholar PubMed
Islam, B.U. and Tabrez, S. (2017). Management of Alzheimer’s disease-an insight of the enzymatic and other novel potential targets. Int. J. Biol. Macromol. 97, 700–709.10.1016/j.ijbiomac.2017.01.076Suche in Google Scholar PubMed
Islam, B.U., Khan, M.S., Jabir, N.R., Kamal, M.A., and Tabrez. S. (2017a). Elucidating treatment of Alzheimer’s disease via different receptors. Curr. Top Med. Chem. 17, 1400–1407.10.2174/1568026617666170103163715Suche in Google Scholar PubMed
Islam, B.U., Zaidi, S.K., Kamal, M.A., and Tabrez, S. (2017b). Exploration of various proteins for the treatment of Alzheimer’s disease. Curr. Drug Metab. 18, 808–813. doi: 10.2174/1389200218666170203110135.10.2174/1389200218666170203110135Suche in Google Scholar
Jabir, N.R., Kamal, M.A., Abuzenadah, A.M., Gan, S.H., Alama, M.N., Baeesa, S.S., and Tabrez, S. (2014). Alzheimer’s and type 2 diabetes treatment via common enzyme targeting. CNS Neurol. Disord. Drug Targets 13, 299–304.10.2174/18715273113126660145Suche in Google Scholar
Jabir, N.R., Firoz, C.K., Baeesa, S.S., Ashraf, G.M., Akhtar, S., Kamal, W., Kamal, M.A., and Tabrez, S. (2015). Synopsis on the linkage of Alzheimer’s and Parkinson’s disease with chronic diseases. CNS Neurosci. Ther. 21, 1–7.10.1111/cns.12344Suche in Google Scholar
Jin, H., Kanthasamy, A., Ghosh, A., Anantharam, V., Kalyanaraman, B., and Kanthasamy, A.G. (2014). Mitochondria-targeted antioxidants for treatment of Parkinson’s disease: preclinical and clinical outcomes. Biochim. Biophys. Acta 1842, 1282–1294.10.1016/j.bbadis.2013.09.007Suche in Google Scholar
Jin, J.L., Fang, M., Zhao, Y.-X., and Liu, X.-Y. (2015). Roles of sigma-1 receptors in Alzheimer’s disease. Int. J. Clin. Exp. Med. 8, 4808–4820.Suche in Google Scholar
Jomova, K., Vondrakova, D., Lawson, M., and Valko, M. (2010). Metals, oxidative stress and neurodegenerative disorders. Mol. Cell. Biochem. 345, 91–104.10.1007/s11010-010-0563-xSuche in Google Scholar
Jucker, M. and Walker, L.C. (2011). Pathogenic protein seeding in Alzheimer’s disease and other neurodegenerative disorders. Ann. Neurol. 70, 532–540.10.1002/ana.22615Suche in Google Scholar
Kajta, M.G. (2004). Apoptosis in the central nervous system: mechanisms and protective strategies. Pol. J. Pharmacol. 56, 689–700.Suche in Google Scholar
Kamal, M.A., Priyamvada, S., Anbazhagan, A.N., Jabir, N.R., Tabrez, S., and Greig, N.H. (2014). Linking Alzheimer’s disease and type 2 diabetes mellitus via aberrant insulin signaling and inflammation. CNS Neurol. Disord. Drug Targets 13, 338–346.10.2174/18715273113126660137Suche in Google Scholar
Keller, J.N., Huang, F.F., and Markesbery, W.R. (2000). Decreased levels of proteasome activity and proteasome expression in aging spinal cord. Neurosci. 98, 149–156.10.1016/S0306-4522(00)00067-1Suche in Google Scholar
Kennedy, T.P., Rao, N.V., Hopkins, C., Pennington, L., Tolley, E., and Hoidal, J.R. (1989). Role of reactive oxygen species in reperfusion injury of the rabbit lung. J. Clin. Invest. 83, 1326–1335.10.1172/JCI114019Suche in Google Scholar PubMed PubMed Central
Kessler, R.C., Berglund, P., Demler, O., Jin, R., Koretz, D., Merikangas, K.R., Rush, A.J., Walters, E.E., and Wang, P.S. (2003). The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R). J. Am. Med. Assoc. 289, 3095–3105.10.1001/jama.289.23.3095Suche in Google Scholar PubMed
Khan, R.A. and Rai, V. (2016). Towards Understanding Epileptic Seizures in the Human Brain: A Computational Approach to Origins. International Conference: Excitatory-Inhibitory Signaling Balance as Therapeutic Target in Epilepsy. August 26–27. Montreal, Canada.Suche in Google Scholar
Khan, M.S., Tabrez, S., Priyadarshini, M., Priyamvada, S., and Khan, M.M. (2012). Targeting Parkinson’s – tyrosine hydroxylase and oxidative stress as points of interventions. CNS Neurol. Disord. Drug Targets 11, 368–380.Suche in Google Scholar
Khanzode, S.D., Dakhale, G.N., Khanzode, S.S., Saoji, A., and Palasodkar, R. (2003). Oxidative damage and major depression: the potential antioxidant action of selective serotonin reuptake inhibitors. Redox Rep. 8, 365–370.10.1179/135100003225003393Suche in Google Scholar PubMed
Kihara, T. and Shimohama, S. (2004). Alzheimer’s disease and acetylcholine receptors. Acta Neurobiol. Exp. (Warsaw) 64, 99–105.Suche in Google Scholar
Kodydková, J., Vávrová, L., Zeman, M., Jirák, R., Macášek, J., Staňková, B., Tvrzická, E., and Žák, A. (2009). Antioxidative enzymes and increased oxidative stress in depressive women. Clin. Biochem. 42, 1368–1374.10.1016/j.clinbiochem.2009.06.006Suche in Google Scholar PubMed
Kong, Q. and Lin, C.-l.G. (2010). Oxidative damage to RNA: mechanisms, consequences, and diseases. Cell. Mol. Life Sci. 67, 1817–1829.10.1007/s00018-010-0277-ySuche in Google Scholar PubMed PubMed Central
Kraytsberg, Y., Kudryavtseva, E., McKee, A.C., Geula, C., Kowall, N.W., and Khrapko, K. (2006). Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat. Genet. 38, 518–520.10.1038/ng1778Suche in Google Scholar PubMed
Kudin, A.P., Kudina, T.A., Seyfried, J., Vielhaber, S., Beck, H., Elger, C.E., and Kunz, W.S. (2002). Seizure-dependent modulation of mitochondrial oxidative phosphorylation in rat hippocampus. Eur. J. Neurosci. 15, 1105–1114.10.1046/j.1460-9568.2002.01947.xSuche in Google Scholar PubMed
Kunz, T. and Oliw, E.H. (2001). Nimesulide aggravates kainic acid-induced seizures in the rat. Pharmacol. Toxicol. 88, 271–276.10.1111/j.1600-0773.2001.880509.xSuche in Google Scholar
LaFerla, F.M. and Oddo, S. (2005). Alzheimer’s disease: a beta, tau, and synaptic dysfunction. Trends Mol. Med. 11, 170–176.10.1016/j.molmed.2005.02.009Suche in Google Scholar PubMed
Lavrik, I.N., Golks, A., and Krammer, P.H. (2005). Caspases: pharmacological manipulation of cell death. J. Clin. Invest. 115, 2665–2672.10.1172/JCI26252Suche in Google Scholar PubMed PubMed Central
Lee, Y.M., Kang, H.C., Lee, J.S., Kim, S.H., Kim, E.Y., Lee, S.K., Slama, A., and Kim, H.D. (2008). Mitochondrial respiratory chain defects: underlying etiology in various epileptic conditions. Epilepsia 49, 685–690.10.1111/j.1528-1167.2007.01522.xSuche in Google Scholar PubMed
Leonard, B. and Maes, M. (2012). Mechanistic explanations how cell-mediated immune activation, inflammation and oxidative and nitrosative stress pathways and their sequels and concomitants play a role in the pathophysiology of unipolar depression. Neurosci. Biobehav. Rev. 36, 764–785.10.1016/j.neubiorev.2011.12.005Suche in Google Scholar PubMed
Liang, L.-P. and Patel, M. (2004). Mitochondrial oxidative stress and increased seizure susceptibility in SOD2 mice. Free Radic. Biol. Med. 36, 542–554.10.1016/j.freeradbiomed.2003.11.029Suche in Google Scholar
Liang, L.-P. and Patel, M. (2006). Seizure-induced changes in mitochondrial redox status. Free Radic. Biol. Med. 40, 316–322.10.1016/j.freeradbiomed.2005.08.026Suche in Google Scholar
Liang, L., Ho, Y., and Patel, M. (2000). Mitochondrial superoxide production in kainate-induced hippocampal damage. Neurosci. 101, 563–570.10.1016/S0306-4522(00)00397-3Suche in Google Scholar
Lin, M.T. and Beal, M.F. (2006). Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443, 787–795.10.1038/nature05292Suche in Google Scholar
Ling, J. and Söll, D. (2010). Severe oxidative stress induces protein mistranslation through impairment of an aminoacyl-tRNA synthetase editing site. Proc. Natl. Acad. Sci. USA 107, 4028–4033.10.1073/pnas.1000315107Suche in Google Scholar
Liu, W., Acin-Perez, R., Geghman, K.D., Manfredi, G., Lu, B., and Li, C. (2011). Pink1 regulates the oxidative phosphorylation machinery via mitochondrial fission. Proc. Natl. Acad. Sci. USA 108, 12920–12924.10.1073/pnas.1107332108Suche in Google Scholar
Lobo, V., Patil, A., Phatak, A., and Chandra, N. (2010). Free radicals, antioxidants, and functional foods: impact on human health. Pharmacog. Rev. 4, 118–126.10.4103/0973-7847.70902Suche in Google Scholar
Lovell, M.A., Robertson, J.D., Teesdale, W.J., Campbell, J.L., and Markesbery, W.R. (1998). Copper, iron, and zinc in Alzheimer’s disease senile plaques. J. Neurol. Sci. 158, 47–52.10.1016/S0022-510X(98)00092-6Suche in Google Scholar
Maehara, Y., Egashira, A., Oki, E., Kakeji, Y., and Tsuzuki, T. (2008). DNA repair dysfunction in gastrointestinal tract cancers. Cancer Sci. 99, 451–458.10.1111/j.1349-7006.2007.00671.xSuche in Google Scholar PubMed
Maes, M., Mihaylova, I., Kubera, M., Uytterhoeven, M., Vrydags, N., and Bosmans, E. (2010). Lower whole blood glutathione peroxidase (GPX) activity in depression, but not in myalgic encephalomyelitis/chronic fatigue syndrome: another pathway that may be associated with coronary artery disease and neuroprogression in depression. Neurol. Endocrinol. Lett. 32, 133–140.Suche in Google Scholar
Malonek, D. and Grinvald, A. (1996). Interactions between electrical activity and cortical microcirculation revealed by imaging spectroscopy: implications for functional brain mapping. Science 272, 551.10.1126/science.272.5261.551Suche in Google Scholar PubMed
Martindale, J.L. and Holbrook, N.J. (2002). Cellular response to oxidative stress: signaling for suicide and survival. J. Cell. Physiol 192, 1–15.10.1002/jcp.10119Suche in Google Scholar
Mattiazzi, M., D’Aurelio, M., Gajewski, C.D., Martushova, K., Kiaei, M., Beal, M.F., and Manfredi, G. (2002). Mutated human SOD1 causes dysfunction of oxidative phosphorylation in mitochondria of transgenic mice. J. Biol. Chem. 277, 29626–29633.10.1074/jbc.M203065200Suche in Google Scholar
Mecocci, P., MacGarvey, U., Kaufman, A.E., Koontz, D., Shoffner, J.M., Wallace, D.C., and Beal, M.F. (1993). Oxidative damage to mitochondrial DNA shows marked age-dependent increases in human brain. Ann. Neurol. 34, 609–616.10.1002/ana.410340416Suche in Google Scholar
Mehlum, L., Ramberg, M., Tørmoen, A.J., Haga, E., Diep, L.M., Stanley, B.H., Miller, A.L., Sund, A.M., and Grøholt, B. (2016). Dialectical behavior therapy compared with enhanced usual care for adolescents with repeated suicidal and self-harming behavior: outcomes over a one-year follow-up. J. Am. Acad. Child Adolesc. Psychiatry. 55, 295–300.10.1016/j.jaac.2016.01.005Suche in Google Scholar
Melov, S., Schneider, J.A., Day, B.J., Hinerfeld, D., Coskun, P., Mirra, S.S., Crapo, J.D., and Wallace, D.C. (1998). A novel neurological phenotype in mice lacking mitochondrial manganese superoxide dismutase. Nat. Genet. 18, 159–163.10.1038/ng0298-159Suche in Google Scholar
Merzbacher, E. (1998). Quantum Mechanics, 3rd Ed. (NY, USA: John Wiley Inc.), ISBN-10: 0471887021.Suche in Google Scholar
Michel, T.M., Pulschen, D., and Thome, J. (2012). The role of oxidative stress in depressive disorders. Curr. Pharm. Des. 18, 5890–5899.10.2174/138161212803523554Suche in Google Scholar
Milder, J. and Patel, M. (2012). Modulation of oxidative stress and mitochondrial function by the ketogenic diet. Epilepsy Res. 100, 295–303.10.1016/j.eplepsyres.2011.09.021Suche in Google Scholar
Mizuno, Y., Ohta, S., Tanaka, M., Takamiya, S., Suzuki, K., Sato, T., Oya, H., Ozawa, T., and Kagawa, Y. (1989). Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochem. Biophys. Res. Commun. 163, 1450–1455.10.1016/0006-291X(89)91141-8Suche in Google Scholar
Montine, T.J., Amarnath, V., Martin, M.E., Strittmatter, W.J., and Graham, D.G. (1996). E-4-hydroxy-2-nonenal is cytotoxic and cross-links cytoskeletal proteins in P19 neuroglial cultures. Am. J. Pathol. 148, 89.Suche in Google Scholar
Moreira, P.I., Nunomura, A., Nakamura, M., Takeda, A., Shenk, J.C., Aliev, G., Smith, M.A., and Perry, G. (2008). Nucleic acid oxidation in Alzheimer disease. Free Radic. Biol. Med. 44, 1493–1505.10.1016/j.freeradbiomed.2008.01.002Suche in Google Scholar PubMed
Neymotin, A., Petri, S., Calingasan, N.Y., Wille, E., Schafer, P., Stewart, C., Hensley, K., Beal, M.F., and Kiaei, M. (2009). Lenalidomide (Revlimid®) administration at symptom onset is neuroprotective in a mouse model of amyotrophic lateral sclerosis. Exp. Neurol. 220, 191–197.10.1016/j.expneurol.2009.08.028Suche in Google Scholar PubMed PubMed Central
Ng, C.F., Schafer, F.Q., Buettner, G.R., and Rodgers, V.G. (2007). The rate of cellular hydrogen peroxide removal shows dependency on GSH: mathematical insight into in vivo H2O2 and GPX concentrations. Free Radic. Res. 41, 1201–1211.10.1080/10715760701625075Suche in Google Scholar PubMed PubMed Central
Nunomura, A., Moreira, P.I., Takeda, A., Smith, M.A., and Perry, G. (2007). Oxidative RNA damage and neurodegeneration. Curr. Med. Chem. 14, 2968–2975.10.2174/092986707782794078Suche in Google Scholar PubMed
Oddo, S. and LaFerla, F.M. (2006). The role of nicotinic acetylcholine receptors in Alzheimer’s disease. J. Physiol. (Paris) 99, 172–179.10.1016/j.jphysparis.2005.12.080Suche in Google Scholar PubMed
Ogle, W.O., Speisman, R.B., and Ormerod, B.K. (2013). Potential of treating age-related depression and cognitive decline with nutraceutical approaches: a mini-review. Gerontology 59, 23–31.10.1159/000342208Suche in Google Scholar PubMed
Oliveira, J.M. and Lightowlers, R.N. (2010). Could successful (mitochondrial) networking help prevent Huntington’s disease? EMBO Mol. Med. 2, 487–489.10.1002/emmm.201000104Suche in Google Scholar PubMed PubMed Central
Orth, M. and Schapira, A. (2001). Mitochondria and degenerative disorders. Am. J. Med. Genet. 106, 27–36.10.1002/ajmg.1425Suche in Google Scholar PubMed
Pacher, P., Beckman, J.S., and Liaudet, L. (2007). Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 87, 315–424.10.1152/physrev.00029.2006Suche in Google Scholar PubMed PubMed Central
Pan, L., Yoshii, Y., Otomo, A., Ogawa, H., Iwasaki, Y., Shang, H.-F., and Hadano, S. (2012). Different human copper-zinc superoxide dismutase mutants, SOD1, and SOD1 exert distinct harmful effects on gross phenotype in mice. PLoS One 7, e33409.10.1371/journal.pone.0033409Suche in Google Scholar PubMed PubMed Central
Panickar, K.S. and Anderson, R.A. (2011). Effect of polyphenols on oxidative stress and mitochondrial dysfunction in neuronal death and brain edema in cerebral ischemia. Int. J. Mol. Sci. 12, 8181–8207.10.3390/ijms12118181Suche in Google Scholar PubMed PubMed Central
Patel, M. (2004). Mitochondrial dysfunction and oxidative stress: cause and consequence of epileptic seizures. Free Radic. Biol. Med. 37, 1951–1962.10.1016/j.freeradbiomed.2004.08.021Suche in Google Scholar PubMed
Perez, M., Valpuesta, J.M., de Garcini, E.M., Quintana, C., Arrasate, M., Carrascosa, J.L.L., Rabano, A., de Yebenes, J.G., and Avila, J. (1998). Ferritin is associated with the aberrant tau filaments present in progressive supranuclear palsy. Am. J. Pathol. 152, 1531–1539.Suche in Google Scholar
Pickrell, A.M., Pinto, M., Hida, A., and Moraes, C.T. (2011). Striatal dysfunctions associated with mitochondrial DNA damage in dopaminergic neurons in a mouse model of Parkinson’s disease. J Neurosci 31, 17649–17658.10.1523/JNEUROSCI.4871-11.2011Suche in Google Scholar
Rai, V. and Khan, R.A. (2009). Epileptic Seizures: A Dynamical Systems Approach to Understand the Origin. https://www.researchgate.net/publication/301698292_Epileptic_Seizures_A_Dynamical_Systems_Approach_to_Understand_the_Origin. DOI: 10.13140/RG.2.1.4620.8881.10.13140/RG.2.1.4620.8881Suche in Google Scholar
Ramirez, M.J. (2013). 5-HT6 receptors and Alzheimer’s disease. Alzheimer’s Res. Ther. 2013, 5:15.10.1186/alzrt169Suche in Google Scholar
Rosen, D.R., Siddique, T., Patterson, D., Figlewicz, D.A., Sapp, P., Hentati, A., Donaldson, D., Goto, J., O’Regan, J.P., Deng, H.X., et al. (1993). Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362, 59–62.10.1038/362059a0Suche in Google Scholar
Rosini, M., Simoni, E., Milelli, A., Minarini, A., and Melchiorre, C. (2014). Oxidative stress in Alzheimer’s disease: are we connecting the dots? J. Med. Chem. 57, 2821–2831.10.1021/jm400970mSuche in Google Scholar
Sacher, J., Rekkas, P.V., Wilson, A.A., Houle, S., Romano, L., Hamidi, J., Rusjan, P., Fan, I., Stewart, D.E., and Meyer, J.H. (2015). Relationship of monoamine oxidase-A distribution volume to postpartum depression and postpartum crying. Neuropsychopharmacol. 40, 429–435.10.1038/npp.2014.190Suche in Google Scholar
Santamaría, A., Pérez-Severiano, F., Rodríguez-Martínez, E., Maldonado, P.D., Pedraza-Chaverri, J., Ríos, C., and Segovia, J. (2001). Comparative analysis of superoxide dismutase activity between acute pharmacological models and a transgenic mouse model of Huntington’s disease. Neurochem. Res. 26, 419–424.10.1023/A:1010911417383Suche in Google Scholar
Scapagnini, G., Davinelli, S., Drago, F., De Lorenzo, A., and Oriani, G. (2012). Antioxidants as antidepressants. CNS Drugs 26, 477–490.10.2165/11633190-000000000-00000Suche in Google Scholar
Schapira, A., Cooper, J., Dexter, D., Jenner, P., Clark, J., and Marsden, C. (1989). Mitochondrial complex I deficiency in Parkinson’s disease. Lancet 333, 1269.10.1016/S0140-6736(89)92366-0Suche in Google Scholar
Schmelzer, C., Lindner, I., Rimbach, G., Niklowitz, P., Menke, T., and Döring, F. (2008). Functions of coenzyme Q10 in inflammation and gene expression. Biofactors 32, 179–183.10.1002/biof.5520320121Suche in Google Scholar PubMed
Shaw, P. (2005). Molecular and cellular pathways of neurodegeneration in motor neuron disease. J. Neurol. Neurosurg. Psychiatry 76, 1046–1057.10.1136/jnnp.2004.048652Suche in Google Scholar
Shimoji, M.J., Mhyre, T.R., and Maguire-Zeiss, K.A. (2013). Microglial activation and antioxidant responses induced by the Parkinson’s disease protein α-synuclein. J. Neuroimm. Pharmacol. 8, 94–117.10.1007/s11481-012-9401-0Suche in Google Scholar
Shin, E.J., Jeong, J.H., Chung, Y.H., Kim, W.K., Ko, K.H., Bach, J.H., Hong, J.S., Yoneda, Y., and Kim, H.C. (2011). Role of oxidative stress in epileptic seizures. Neurochem. Int. 59, 122–137.10.1016/j.neuint.2011.03.025Suche in Google Scholar
Shivakumar, B.R., Anandatheerthavarada, H.K., and Ravindranath, V. (1991). Free radical scavenging systems in developing rat brain. Int. J. Dev. Neurosci. 9, 181–185.10.1016/0736-5748(91)90010-JSuche in Google Scholar
Shoffner, J.M., Lott, M.T., Lezza, A.M., Seibel, P., Ballinger, S.W., and Wallace, D.C. (1990). Myoclonic epilepsy and ragged-red fiber disease (MERRF) is associated with a mitochondrial DNA tRNA Lys mutation. Cell 61, 931–937.10.1016/0092-8674(90)90059-NSuche in Google Scholar
Siddiqui, A., Rivera-Sanchez, S., Castro Mdel, R., Acevedo-Torres, K., Rane, A., Torres-Ramos, C.A., Nicholls, D.G., Andersen, J.K. and Ayala-Torres, S. (2012). Mitochondrial DNA damage is associated with reduced mitochondrial bioenergetics in Huntington’s disease. Free Radic. Biol. Med. 53, 1478–1488.10.1016/j.freeradbiomed.2012.06.008Suche in Google Scholar PubMed PubMed Central
Sies, H. (1997). Oxidative stress: oxidants and antioxidants. Exp. Physiol. 82, 291–295.10.1113/expphysiol.1997.sp004024Suche in Google Scholar PubMed
Simon, N.M., Smoller, J.W., McNamara, K.L., Maser, R.S., Zalta, A.K., Pollack, M.H., Nierenberg, A.A., Fava, M., and Wong, K.-K. (2006). Telomere shortening and mood disorders: preliminary support for a chronic stress model of accelerated aging. Biol. Psychiatry 60, 432–435.10.1016/j.biopsych.2006.02.004Suche in Google Scholar PubMed
Skaper, S.D., Facci, L., and Giusti, P. (2013). Glia and mast cells as targets for palmitoylethanolamide, an anti-inflammatory and neuroprotective lipid mediator. Mol. Neurobiol. 48, 340–352.10.1007/s12035-013-8487-6Suche in Google Scholar PubMed
Smith, M.A., Taneda, S., Richey, P.L., Miyata, S., Yan, S.-D., Stern, D., Sayre, L.M., Monnier, V.M., and Perry, G. (1994). Advanced Maillard reaction end products are associated with Alzheimer disease pathology. Proc. Nat. Acad. Sci. USA 91, 5710–5714.10.1073/pnas.91.12.5710Suche in Google Scholar PubMed PubMed Central
Smith, M.A., Sayre, L.M., Monnier, V.M., and Perry, G. (1995). Radical AGEing in Alzheimer’s disease. Trends Neurosci. 18, 172–176.10.1016/0166-2236(95)93897-7Suche in Google Scholar
Smith, M.A., Richey Harris, P.L., Sayre, L.M., Beckman, J.S., and Perry, G. (1997). Widespread peroxynitrite-mediated damage in Alzheimer’s disease. J. Neurosci. 17, 2653–2657.10.1523/JNEUROSCI.17-08-02653.1997Suche in Google Scholar
Sohal, R.S., Sohal, B.H., and Orr, W.C. (1995). Mitochondrial superoxide and hydrogen peroxide generation, protein oxidative damage, and longevity in different species of flies. Free Radic. Biol. Med. 19, 499–504.10.1016/0891-5849(95)00037-XSuche in Google Scholar
Sorolla, M.A., Reverter-Branchat, G., Tamarit, J., Ferrer, I., Ros, J., and Cabiscol, E. (2008). Proteomic and oxidative stress analysis in human brain samples of Huntington disease. Free Radic. Biol. Med. 45, 667–678.10.1016/j.freeradbiomed.2008.05.014Suche in Google Scholar
Stefanis, L. (2012). α-Synuclein in Parkinson’s disease. Cold Spring Harb. Perspect Med. 2, a009399.10.1101/cshperspect.a009399Suche in Google Scholar
Su, F., Bai, F., Zhou, H., and Zhang, Z. (2016). Microglial toll-like receptors and Alzheimer’s disease. Brain Behav. Immun. 52, 187–198.10.1016/j.bbi.2015.10.010Suche in Google Scholar
Sudha, K., Rao, A.V., and Rao, A. (2001). Oxidative stress and antioxidants in epilepsy. Clin. Chim. Acta. 303, 19–24.10.1016/S0009-8981(00)00337-5Suche in Google Scholar
Suraweera, A., Munch, C., Hanssum, A., and Bertolotti, A. (2012). Failure of amino acid homeostasis causes cell death following proteasome inhibition. Mol. Cell 48, 242–253.10.1016/j.molcel.2012.08.003Suche in Google Scholar PubMed PubMed Central
Tabner, B.J., El-Agnaf, O.M., German, M.J., Fullwood, N.J., and Allsop, D. (2005). Protein aggregation, metals and oxidative stress in neurodegenerative diseases. Biochem. Soc. Transac. 33, 1082–1086.10.1042/BST0331082Suche in Google Scholar
Thompson, J.K., Peterson, M.R., and Freeman, R.D. (2003). Single-neuron activity and tissue oxygenation in the cerebral cortex. Science 299, 1070–1072.10.1126/science.1079220Suche in Google Scholar PubMed
Trifunovic, A. and Larsson, N.G. (2008). Mitochondrial dysfunction as a cause of aging. J. Internal Med. 263, 167–178.10.1111/j.1365-2796.2007.01905.xSuche in Google Scholar
Tufi, R., Gandhi, S., de Castro, I.P., Lehmann, S., Angelova, P.R., Dinsdale, D., Deas, E., Plun-Favreau, H., Nicotera, P., Abramov, A.Y., et al. (2014). Enhancing nucleotide metabolism protects against mitochondrial dysfunction and neurodegeneration in a PINK1 model of Parkinson’s disease. Nat. Cell Biol. 16, 157–166.10.1038/ncb2901Suche in Google Scholar
Turner, C. and Schapira, A.H. (2001). Mitochondrial dysfunction in neurodegenerative disorders and aging. Adv. Exp. Med. Biol. 487, 229–251.10.1007/978-1-4615-1249-3_19Suche in Google Scholar
Turrens, J.F., Freeman, B.A., Levitt, J.G., and Crapo, J.D. (1982). The effect of hyperoxia on superoxide production by lung submitochondrial particles. Arch. Biochem. Biophys. 217, 401–410.10.1016/0003-9861(82)90518-5Suche in Google Scholar
Uttara, B., Singh, A.V., Zamboni, P., and Mahajan, R.T. (2009). Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr. Neuropharmacol. 7, 65–74.10.2174/157015909787602823Suche in Google Scholar
Vargas, M.R., Johnson, D.A., and Johnson, J.A. (2011). Decreased glutathione accelerates neurological deficit and mitochondrial pathology in familial ALS-linked hSOD1(G93A) mice model. Neurobiol. Dis. 43, 543–551.10.1016/j.nbd.2011.04.025Suche in Google Scholar
Waldbaum, S. and Patel, M. (2010). Mitochondrial dysfunction and oxidative stress: a contributing link to acquired epilepsy? J. Bioenerget. Biomembr. 42, 449–455.10.1007/s10863-010-9320-9Suche in Google Scholar
Waldbaum, S., Liang, L.P., and Patel, M. (2010). Persistent impairment of mitochondrial and tissue redox status during lithium-pilocarpine-induced epileptogenesis. J. Neurochem. 115, 1172–1182.10.1111/j.1471-4159.2010.07013.xSuche in Google Scholar
Walker, F.O. (2007). Huntington’s disease. Lancet 369, 218–228.10.1016/S0140-6736(07)60111-1Suche in Google Scholar
Walker, L.C. and LeVine, H., 3rd. (2000). The cerebral proteopathies. Neurobiol. Aging 21, 559–561.10.1016/S0197-4580(00)00160-3Suche in Google Scholar
Wallace, D.C., Zheng, X., Lott, M.T., Shoffner, J.M., Hodge, J.A., Kelley, R.I., Epstein, C.M., and Hopkins, L.C. (1988). Familial mitochondrial encephalomyopathy (MERRF): genetic, pathophysiological, and biochemical characterization of a mitochondrial DNA disease. Cell 55, 601–610.10.1016/0092-8674(88)90218-8Suche in Google Scholar
Wang, J.Q., Chen, Q., Wang, X., Wang, Q.C., Wang, Y., Cheng, H.P., Guo, C., Sun, Q., Chen, Q., and Tang, T.S. (2013). Dysregulation of mitochondrial calcium signaling and superoxide flashes cause mitochondrial genomic DNA damage in Huntington disease. J. Biol. Chem. 288, 3070–3084.10.1074/jbc.M112.407726Suche in Google Scholar PubMed PubMed Central
Wasterlain, C.G., Fujikawa, D.G., Penix, L., and Sankar, R. (1993). Pathophysiological mechanisms of brain damage from status epilepticus. Epilepsia 34, S37–S53.10.1111/j.1528-1157.1993.tb05905.xSuche in Google Scholar PubMed
Weinreb, O., Amit, T., Bar-Am, O., and Youdim, M.B. (2016). Neuroprotective effects of multifaceted hybrid agents targeting MAO, cholinesterase, iron, and β-amyloid in aging and Alzheimer’s disease. British J. Pharmacol. 173, 2080–2094.10.1111/bph.13318Suche in Google Scholar PubMed PubMed Central
Wenk, G.L. (2006). Neuropathologic changes in Alzheimer’s disease: potential targets for treatment. J. Clin. Psychiatry 67, 3.Suche in Google Scholar
Weydert, C.J. and Cullen, J.J. (2010). Measurement of superoxide dismutase, catalase, and glutathione peroxidase in cultured cells and tissue. Nat. Protoc. 5, 51–66.10.1038/nprot.2009.197Suche in Google Scholar PubMed PubMed Central
Willmore, L.J., Sypert, G.W., and Munson, J.B. (1978). Recurrent seizures induced by cortical iron injection: a model of posttraumatic epilepsy. Ann. Neurol. 4, 329–336.10.1002/ana.410040408Suche in Google Scholar PubMed
Wolkowitz, O.M., Mellon, S.H., Epel, E.S., Lin, J., Dhabhar, F.S., Su, Y., Reus, V.I., Rosser, R., Burke, H.M., and Kupferman, E. (2011). Leukocyte telomere length in major depression: correlations with chronicity, inflammation and oxidative stress-preliminary findings. PLoS One 6, e17837.10.1371/journal.pone.0017837Suche in Google Scholar PubMed PubMed Central
Wu, S.-B., Ma, Y.-S., Wu, Y.-T., Chen, Y.-C., and Wei, Y.-H. (2010). Mitochondrial DNA mutation-elicited oxidative stress, oxidative damage, and altered gene expression in cultured cells of patients with MERRF syndrome. Mol. Neurobiol. 41, 256–266.10.1007/s12035-010-8123-7Suche in Google Scholar PubMed
Xiang, W., Schlachetzki, J.C., Helling, S., Bussmann, J.C., Berlinghof, M., Schäffer, T.E., Marcus, K., Winkler, J., Klucken, J., and Becker, C.-M. (2013). Oxidative stress-induced posttranslational modifications of alpha-synuclein: specific modification of α-synuclein by 4-hydroxy-2-nonenal increases dopaminergic toxicity. Mol. Cell. Neurosci. 54, 71–83.10.1016/j.mcn.2013.01.004Suche in Google Scholar PubMed
Yu, B.P. (1994). Cellular defenses against damage from reactive oxygen species. Physiol. Rev. 74, 139–162.10.1152/physrev.1994.74.1.139Suche in Google Scholar PubMed
Yusuf, M., Khan, R.A., and Ahmed, B. (2008). Syntheses and anti-depressant activity of 5-amino-1, 3, 4-thiadiazole-2-thiol imines and thiobenzyl derivatives. Bioorg Med Chem 16, 8029–8034.10.1016/j.bmc.2008.07.056Suche in Google Scholar PubMed
Yusuf, M., Khan, R.A., Khan, M., and Ahmed, B. (2012). Plausible antioxidant biomechanics and anticonvulsant pharmacological activity of brain-targeted β-carotene nanoparticles. Int. J. Nanomed. 7, 4311–4322.10.2147/IJN.S34588Suche in Google Scholar PubMed PubMed Central
Yusuf, M., Khan, M., Khan, R.A., and Ahmed, B. (2013). Preparation, characterization, in vivo and biochemical evaluation of brain-targeted piperine solid lipid nanoparticles in an experimentally induced Alzheimer’s disease model. J. Drug Target. 21, 300–311.10.3109/1061186X.2012.747529Suche in Google Scholar PubMed
Yusuf, M., Khan, M., Khan, R.A., Maghrabi, I.A., and Ahmed, B. (2016). Polysorbate-80-coated, polymeric curcumin nanoparticles for in vivo anti-depressant activity across BBB and envisaged biomolecular mechanism of action through a proposed pharmacophore model. J. Microencap. 33, 646–655.10.1080/02652048.2016.1242666Suche in Google Scholar PubMed
Zabel, M.K. and Kirsch, W.M. (2013). From development to dysfunction: Microglia and the complement cascade in CNS homeostasis. Ageing Res. Rev. 12, 749–756.10.1016/j.arr.2013.02.001Suche in Google Scholar PubMed PubMed Central
Zhao, N., Liu, C.-C., Qiao, W., and Bu, G. (2017). Apolipoprotein E, receptors, and modulation of Alzheimer’s disease. Biol. Psychiatry 83, 347–357.10.1016/j.biopsych.2017.03.003Suche in Google Scholar PubMed PubMed Central
Zsurka, G. and Kunz, W.S. (2010). Mitochondrial dysfunction in neurological disorders with epileptic phenotypes. J. Bioenerg. Biomembr. 42, 443–448.10.1007/s10863-010-9314-7Suche in Google Scholar PubMed
Zsurka, G., Baron, M., Stewart, J.D., Kornblum, C., Bös, M., Sassen, R., Taylor, R.W., Elger, C.E., Chinnery, P.F., and Kunz, W.S. (2008). Clonally expanded mitochondrial DNA mutations in epileptic individuals with mutated DNA polymerase γ. J. Neuropathol. Exp. Neurol. 67, 857–866.10.1097/NEN.0b013e3181839b2dSuche in Google Scholar PubMed
Zsurka, G., Hampel, K., Nelson, I., Jardel, C., Mirandola, S., Sassen, R., Kornblum, C., Marcorelles, P., Lavoué, S., and Lombès, A. (2010). Severe epilepsy as the major symptom of new mutations in the mitochondrial tRNAPhe gene. Neurology 74, 507–512.10.1212/WNL.0b013e3181cef7abSuche in Google Scholar PubMed
Zuchner, T., Schliebs, R., and Perez-Polo, J.R. (2005). Down-regulation of muscarinic acetylcholine receptor M2 adversely affects the expression of Alzheimer’s disease-relevant genes and proteins. J. Neurochem. 95, 20–32.10.1111/j.1471-4159.2005.03335.xSuche in Google Scholar PubMed
Zuchora, B., Turski, W.A., Wielosz, M., and Urbaska, E.M. (2001). Protective effect of adenosine receptor agonists in a new model of epilepsy seizures evoked by mitochondrial toxin, 3-nitropropionic acid, in mice. Neurosci. Lett. 305, 91–94.10.1016/S0304-3940(01)01816-XSuche in Google Scholar
©2018 Walter de Gruyter GmbH, Berlin/Boston
Artikel in diesem Heft
- Frontmatter
- Reviews
- Biomechanistic insights into the roles of oxidative stress in generating complex neurological disorders
- Metformin-induced anticancer activities: recent insights
- Research Articles/Short Communications
- Protein Structure and Function
- Interaction of the middle domains stabilizes Hsp90α dimer in a closed conformation with high affinity for p23
- How human serum albumin recognizes DNA and RNA
- In vitro reconstitution and biochemical characterization of human phospholipid scramblase 3: phospholipid specificity and metal ion binding studies
- Cell Biology and Signaling
- LncRNA KCNQ1OT1 ameliorates particle-induced osteolysis through inducing macrophage polarization by inhibiting miR-21a-5p
- LncRNA PART1 modulates toll-like receptor pathways to influence cell proliferation and apoptosis in prostate cancer cells
- High-content hydrogen water-induced downregulation of miR-136 alleviates non-alcoholic fatty liver disease by regulating Nrf2 via targeting MEG3
Artikel in diesem Heft
- Frontmatter
- Reviews
- Biomechanistic insights into the roles of oxidative stress in generating complex neurological disorders
- Metformin-induced anticancer activities: recent insights
- Research Articles/Short Communications
- Protein Structure and Function
- Interaction of the middle domains stabilizes Hsp90α dimer in a closed conformation with high affinity for p23
- How human serum albumin recognizes DNA and RNA
- In vitro reconstitution and biochemical characterization of human phospholipid scramblase 3: phospholipid specificity and metal ion binding studies
- Cell Biology and Signaling
- LncRNA KCNQ1OT1 ameliorates particle-induced osteolysis through inducing macrophage polarization by inhibiting miR-21a-5p
- LncRNA PART1 modulates toll-like receptor pathways to influence cell proliferation and apoptosis in prostate cancer cells
- High-content hydrogen water-induced downregulation of miR-136 alleviates non-alcoholic fatty liver disease by regulating Nrf2 via targeting MEG3