Open access peer-reviewed chapter

Reactive Oxygen Species and Selenium in Epilepsy and in Other Neurological Disorders

Written By

Erkki Antila, Tuomas Westermarck, Arno Latvus and Faik Atroshi

Reviewed: 05 March 2020 Published: 14 October 2020

DOI: 10.5772/intechopen.92003

From the Monograph

Personalized Medicine, in Relation to Redox State, Diet and Lifestyle

Edited by Faik Atroshi

Chapter metrics overview

648 Chapter Downloads

View Full Metrics

Abstract

Oxidative stress has been implicated in epilepsy and various neurodegenerative disorders. In this review, we elaborate oxidative stress-mediated neuronal loss and assess the role of selenium in some neurological disorders including epilepsy. Selenium as an essential trace element has attracted the attention of many researchers because of its potentialities in human health. It has an important role in the brain, immune response, defense against tissue damage, and thyroid function. Selenium forms part of the active site of the peroxide-destroying enzyme glutathione peroxidase (GSHPx), and it also has other functions, for example in biotransformation and detoxification. Functional and clinical consequences of selenium deficiency states in neurological diseases have been described, and the selenium requirement, which is influenced by various processes, has been discussed. Wide variations have been found in selenium status in different parts of the world, and populations or groups of patients exposed to marginal deficiency are more numerous than was previously thought. Chronic diseases, such as neurological disorders, heart disease, diabetes, cancer, aging, and others, are reported to associate with markers of oxidative damage. It is, therefore, not unreasonable to suggest that antioxidants would alleviate the oxidative damage, resulting in health improvements. In recent years, accumulated evidence in nutrigenomics, laboratory experiments, clinical trials, and epidemiological data have established the role of selenium in a number of conditions. Most of these effects are related to the function of selenium in the antioxidant enzyme systems. Current research activities in the field of human medicine and nutrition are devoted to the possibilities of using selenium as an adjuvant for the treatment of degenerative or free radical diseases such as neurological disorders, inflammatory diseases, and cancer.

Keywords

  • selenium
  • antioxidants
  • oxidative stress
  • pathogenesis of neurological disorders

1. Introduction

Selenium is a trace mineral essential to human health, which has an important role in the immune response, defense against tissue damage, and thyroid function. Improving selenium status could help protect against overwhelming tissue damage and infection in critically ill adults [1, 2, 3, 4]. Selenium is incorporated into proteins to make selenoproteins, which are important antioxidant enzymes. The antioxidant properties of selenoproteins help prevent cellular damage from free radicals. Free radicals are natural by-products of oxygen metabolism that may contribute to the development of chronic diseases such as cancer and heart disease [5, 6]. There is evidence that selenium deficiency may contribute to development of a form of hypothyroidism and a weakened immune system. Specific diseases have been associated with selenium deficiency such as Keshan and Kashin-Beck disease, which results in osteoarthropathy and myxedematous endemic cretinism, which results in mental retardation [7, 8].

In recent years, considerable evidence has emerged implicating a role for oxygen free radicals in the initiation of cellular injury which can lead to the development of several neurological disorders. The neonatal brain, with its high concentrations of unsaturated fatty acids (lipid content), high rate of oxygen consumption, and low concentrations of antioxidants, is particularly vulnerable to oxidative damage. Thus, increased oxidative stress has been implicated in various neurological disorders such as seizures, ischemia-reperfusion injury, and neurodegenerative diseases [9, 10] such as Alzheimer’s, Parkinson’s, and Lou Gehrig’s disease. Free radical damage has been implicated in the initiation and propagation of seizure activity as well as the accompanying seizure-induced neuronal damage [11]. Therefore, antioxidants could play an important role in modulating susceptibility to seizure activity and seizure-induced neuronal injury.

The use of selenium as a supplement in neurological disorders has been reported. The rationale for selenium supplementation comes from the nutrient’s role as an antioxidant [12], working primarily as a component of glutathione peroxidase, an important cellular protector against free radical damage. Furthermore, selenium deficiency is known to result in neuromuscular disease. Attempts have been taken to relate selenium to different neurological disorders as epilepsy, phenylketonuria and maple syrup urine disease, Parkinson’s disease, amyotrophic lateral sclerosis, neuronal ceroid lipofuscinoses, myotonic dystrophy, multiple sclerosis, Down syndrome (DS), and Alzheimer’s disease [13, 14]. The relevant connection between selenium and the majority of these disorders rests on clinical observations during selenium supplementation alone or in combination with other antioxidants.

Advertisement

2. Selenium distribution in humans

Due to the uneven geographic distribution of selenium in soil, the amount of whole-body selenium in adult humans was reported to differ in different countries [15, 16, 17, 18]. At normal dietary levels, the highest selenium concentration was detected in reindeer liver and kidney, followed by the spleen, pancreas, heart, brain, lung, bone, and skeletal muscle. Selenium concentration in the human body was also found to vary with age. For instance, selenium concentration in fetal brain decreased with age but increased with age postnatally. Blood selenium levels were negatively correlated with age in healthy adults, and the same was documented for 40 patients with dementia of the Alzheimer type (DAT). Furthermore, Ejima et al. [19] reported that selenium concentrations varied in different adult human brain regions.

Advertisement

3. Selenium in the brain

Neurochemical aspects of selenium have been widely reported. In this approach to the etiopathogenetic role of selenium in CNS diseases, teleological ideas are explicitly correlated to the paradigm of oxygen toxicity. The brain differs from many other tissues, being a highly aerobic and totally oxygen-dependent tissue. Oxygen reduction produces reactive radical intermediates, i.e., singlet oxygen, a superoxide radical which is thought to be a major agent of oxygen toxicity. Hydrogen peroxide, H2O2, is formed through dismutation of a singlet oxygen catalyzed by Cu-Zn and Mn forms of superoxide dismutase, both found in CNS tissues. Other hydrogen peroxide-generating enzymes are associated with d- and l-amino acid oxidase, monoamine oxidase, a-hydroxyacid oxidase, xanthine oxidase, and cytochrome P-450 system.

Unlike charged oxygen radicals being a rather unreactive and stable, H2O2 rapidly crosses cell membranes. Cellular damage is accomplished when H2O2 decomposes to the highly reactive hydroxyl radical in iron(II) or copper(I) catalyzed reactions. Scavenging of H2O2 and contemporaneous prevention of hydroxyl radical formation occurs predominantly at two cellular sites, in the peroxisomes and in the cytoplasm by catalase and GSHPx (GSHPx, glutathione/hydrogen peroxide oxidoreductase, EC 1.11.1.9), respectively. If this is not done, the hydroxyl radical may attack the fatty acid side chains and start a chain reaction of lipid peroxidation. Lipid peroxidation causes gradual loss of membrane fluidity and membrane potential and increases membrane permeability to ions. Radical attack may also destroy membrane-bound enzymes and receptors, e.g., the binding of serotonin is decreased. Oxidative degradation and polymerization of lipids leads to the accumulation of lipofuscin, the age pigment. The presence of catalytic iron and copper complexes in human CSF, and the high iron content of brain, suggests that they are very sensitive to oxygen radical generation.

The crucial role of selenium as a trace element in the nervous tissue has been associated with a selenoenzyme glutathione peroxidase. Selenium is thought to be present at the active site of GSHPx in its selenolate form as selenocysteine [20]. The fairly homogenous distribution of selenium in the human brain corresponds well with the regions of the highest and lowest GSHPx activity found in the rat brain. However, estimates of the amounts of selenium in rat brain have indicated that GSHPx may account for only 1/5 of the total Se found in the brain [21]. Most of the selenium is bound to proteins and not to amino acids or nucleic acids [22]. Selenoproteins, other than GSHPx, found in the brain and the reproductive and the endocrine organs seem to serve as a priority pathway of the element during inadequate selenium intake. The function of selenium in proteins has been explained in terms of semiconduction [23]. It is possible although not yet proven that selenium may have this or some other special functions outside of GSHPx too.

Observations suggest that free radical intermediates may be involved in the coupling between depolarization of the plasma membrane, Ca2+ fluxes, and neurotransmitter release [24]. In general, cellular redox adjustments regulate functional sulfhydryl groups of proteins. Therefore, cellular prooxidant states may be involved in the generation of physiological responses. This means that the adjustment of redox equilibrium in CNS is a far more delicate phenomenon than just a tendency to a normal balance.

The regulation of GSH level (GSH/GSSG) through pentose phosphate pathway producing NADPH, GSH-reductase, and GSHPx contributes to the overall redox state of cells in the brain [25]. The brain tends to need radical reactions as well as to possess specific or high endogenous levels of free radical scavengers such as dopamine, norepinephrine, and catechol estrogens, taurine and carnosine [26], in neurons. Carnosine is involved with GSBA activity in the brain, and a study by Takahashi [27] demonstrated that homocarnosine levels were high in patients who responded to antiepileptic drugs. The functional balance between various free radical scavenger systems in the brain seems reasonable. Significant positive correlations between catalase and SOD levels have been reported in tissues of normal subjects excluding erythrocytes. Factors concomitantly influencing the variation of the activities of SOD, catalase, and GHSPx have been reported. Enzymes frequently called a protective should rather be envisaged as being regulatory, controlling the levels of different states of oxygen reduction.

Advertisement

4. Brain antioxidant homeostasy in relation to selenium and GSHPx

As a trace element in nature, the availability of selenium may be limited. GSHPx activity has been shown to reflect selenium status in deficient and adequate states [6]. On the other hand, protection against toxicity is likely to involve the alterations in GSH metabolism that occur in nutritional Se deficiency. High concentrations of erythrocyte glutathione in patients with neurological disorders have been reported [28]. However, regulatory mechanisms apparently exist which ensure that during periods of insufficient selenium intake, the content of the element is kept up above all in the brain and the reproductive and endocrine organs.

Yoshida et al. [29] reported a comprehensive method for identifying the selenium-binding proteins using PenSSeSPen as a model of the selenium metabolite, selenotrisulfide (RSSeSR, STS), which was applied to a complex cell lysate generated from the rat brain. The authors stated that a thiol-containing protein at m/z 15155 in the brain cell lysate was identified as the cystatin-12 precursor (CST12) from a rat protein database search and a tryptic fragmentation experiment. CST12 belongs to the cysteine proteinase inhibitors of the cystatin superfamily that are of interest in mechanisms regulating the protein turnover and polypeptide production in the central nervous system and other tissues. Consequently, CST12 is suggested to be one of the cytosolic proteins responsible for the selenium metabolism in the brain [29].

Selenium seems to be somehow involved in the regulation of oxygen metabolism through its influence on a variety of enzymes. In concentrations of 6 × 10−7 to × 10−6 M, selenite induces a 30-fold increase of GSHPx activity in neuroblast cells in vitro. Other studies with the rat liver have suggested that Se status regulates the level of GSHPx mRNA as well as regulates GSHPx protein concentration and GSHPx activity [30]. In concentrations of 0.7–2 × 10−5 M, Se in rat liver increases the activities of g-glutamylcysteine synthetase, the first rate-limiting enzyme in GSH biosynthesis, and GSSG-reductase, which catalyzes the reduction of GSSG to GSH [31]. In some species the induction of GSH-S-transferase has been shown to occur as a result of Se deficiency. H2O2 as the most stable and diffusible of the oxygen reduction intermediates may exert an influence on the expression of SOD, catalase, and GSHPx activities. GSHPx, which exists in several forms that differ in their primary structure and localization, catalyzes the reduction of hydrogen peroxide and organic hydroperoxide by glutathione and functions in the protection of cells against oxidative damage [32]. The homeostasy in the oxidative metabolism and oxygen reduction may be distorted by different means, either inherent or acquired. Depending on the spatial and temporal occurrence of the distortion, various neurological states are expressed.

The developing brain is particularly susceptible to oxidative stress, more so than the mature brain [33]. H2O2 accumulation has also been associated with increased injury in superoxide dismutase-overexpressing neonatal murine brain, and greater cell death is seen when immature neurons are exposed to H2O2 than mature neurons. Increased H2O2 accumulation may be the result of relative insufficiency of the endogenous enzyme GSHPx.

Under physiologic circumstances, the brain has efficient antioxidant defense mechanisms, including GSHPx, which converts potentially harmful H2O2 to oxygen and water at the expense of reduced GSH. Under oxidative stress, in the immature brain, endogenous levels of GSHPx may be inadequate for converting excess H2O2. Transgenic mice that overexpress GSHPx (hGPx-tg), when subjected to hypoxic-ischemic (HI), have less histologic brain injury than their Wt littermates [25]. In addition, the cortex exhibits increased GSHPx enzyme activity at 24 h, whereas GSHPx activity remains unaltered in the Wt brain. In addition, neurons cultured from GSHPx-tg brain are resistant to injury from exogenously applied H2O2 [34]. Neurons cultured from the hippocampus and cortex that are transfected (transfection describes the introduction of foreign material into eukaryotic cells) with genes for catalase and GSHPx also show protection from neurotoxic insults and a corresponding decrease in H2O2 accumulation [35]. These findings indicate that adequate GSHPx activity can ameliorate injury to the immature brain from oxidative stress due to H2O2.

It is well established that previous stress to the brain can induce tolerance to subsequent injury, a phenomenon called personality change (PC). In neonatal rodents, protection against HI brain injury has been induced by PC with a period of hypoxia before the induction of HI [36]. The mechanisms of this protection have yet to be fully determined, but it has been established that a large number of genes are induced in response to hypoxia [37]. Several of these genes are regulated by the transcription factor hypoxia-inducible factor-1α (HIF-1α) and perhaps most importantly vascular endothelial growth factor (VEGF) and erythropoietin (EPO). VEGF is upregulated after focal ischemic injury in the neonatal rat, in parallel with induction of HIF-1α [38, 39].

Advertisement

5. Aging, dementia, and Alzheimer’s disease

Major interest in CNS selenium is related to aspects of oxidative stress and aging. The decrease in cerebral blood flow, glucose utilization, and oxygen consumption common to many dementias results from abnormalities of brain structure with a high oxidative capacity. During mental activity, regional cerebral oxidative metabolism and regional cerebral blood flow increase in several areas of the brain. In dementia of the Alzheimer type, brain blood flow and oxidative metabolism are reduced. This situation may lead to loss of balance between prooxidants and antioxidants [40]. The role of H2O2 in the etiology of Alzheimer’s disease has been reported [41, 42]. Furthermore, the activities of catalase and GSHPx decrease with aging in intact animals. Some reports suggest that the SOD activity is significantly greater in Alzheimer cell fibroblasts. Both GSHPx and SOD activities in the erythrocytes of AD have been reported to be normal, while other studies show significantly higher erythrocyte SOD level in AD [43]. It remains to be seen whether oxidative damage will still be related to the accumulation of aluminum silicates in the brain as well as to that of the senile plaques and tangles. Experiments have indicated that aluminum salts may not only accelerate Fe(II)-induced peroxidation of membrane lipids but do this especially in the brain [44, 45].

In order to evaluate the peroxidative stress in dementias, autopsy brain samples should be studied for GSHPx, SOD, catalase, and selenium. A direct causal relationship between brain antioxidant defenses and dementia in aging and Alzheimer’s disease is hard to demonstrate because of the extremely slow process. Interestingly a high proportion of Down syndrome patients develop the neuropathological and clinical changes of AD, suggesting a close pathogenetic relationship between these disorders. Thus, the correction of antioxidant balance in AD by Se supplementation should be demonstrated by other means so as to direct it preventatively to those with a high risk of developing AD.

In Alzheimer’s disease the H2O2 molecule should be considered a therapeutic target for treatment of the oxidative stress associated with the disease. The actions of H2O2 include modification of DNA, proteins, and lipids, all of which are effects seen in an Alzheimer’s disease brain, possibly contributing to the loss of synaptic function characteristic of the disease. Future research and development of agents that specifically target the H2O2 molecule or enzymes involved in its metabolism may provide the future route to Alzheimer’s disease therapy [42].

Advertisement

6. Down syndrome

Trisomy 21 (Down syndrome) is the most common genetic cause of learning disability in humans [46], occurring in about 1 per 1000 babies born each year [47]. Postmortem studies have reported neuronal depletion and structural abnormalities of the brain during late gestation and early postnatal life [48]. Down syndrome was found to have increased activity of superoxide dismutase without a compensatory increase in glutathione peroxidase activity [49]. However, there is no evidence to support the use of antioxidant or folinic acid supplements in children with Down syndrome [50].

Increased primary gene products which may contribute to the pathology of DS include cytoplasmic CuZn-superoxide dismutase (SOD). Consistent with the gene dosage effect, SOD activity is increased by 50%, leading to noxious concentrations of H2O2, while brain GSHPx remains normal. The overall redox state in other tissues is corrected by an adaptive increase of GSHPx activity. This means that the brain is especially susceptible to oxygen free radical stress. Our primary survey of specific antioxidant therapy with selenium [51] rests on this theory.

The whole-body retention of 5–8 kBq 75Se-sodium selenite with 0.4 g Se as carrier/kg body weight in DS patients has been earlier estimated to be 53.3 + 21.1%. Stable Se supplementation increased 75Se elimination, indicating a saturated Se pool in the body. Twenty-four patients aged 1–41 years were either given selenium supplements of 0.025 mg Se/kg/d in the form of sodium selenite or given placebo or no preparation. The serum levels of selenium indicated no real deficiency as compared to the normal healthy population. However, the mean compensatory increase of erythrocyte GSHPx activity before supplementation was lower than expected. Because of difficulties in obtaining brain biopsies, variables found in the plasma and erythrocyte samples were used as indicators of antioxidant balance. Sinet et al. [52] have reported a high positive correlation between erythrocyte GSHPx values and the intelligence quotient in DS patients. Because of this and the difficulties in testing changes of IQ which is one of the most decisive clinical goals of therapy, we found it reasonable to follow changes of E-GSHPX. Selenium supplementation increased E-GSHPx activity by 28% (59.9% above normal). The correction is sensitive to adequate Se supplementation as indicated by SOD/GSHPx index which decreased by 23.9% (P < 0.01) [51]. Interestingly the primarily high serum and blood mononuclear cell levels of cupric and ferrous ions decreased, and that of zinc ions increased during supplementation. In conclusion we believe that the patients have benefited from the selenium supplementation through optimization of their antioxidant protection by GSHPx.

Advertisement

7. Selenium and epilepsy

An epilepsy syndrome is a complex of signs and symptoms defining a unique condition [53]. Oxidative stress and generation of reactive oxygen species are strongly implicated in a number of neuronal and neuromuscular disorders, including epilepsy. The functions of selenium as an antioxidant trace element are believed to be carried out by selenoproteins that possess antioxidant activities and the ability to promote neuronal cell survival. Selenoproteins are important for normal brain function, and decreased function of selenoproteins may lead to impaired cognitive function and neurological disorders [54].

Free radicals and lipoperoxidation reactions seem to be involved in epileptic seizure developing after brain hemorrhage of different kinds. There is an association between hemosiderin deposition and post-traumatic epilepsy. An extravasation of blood and hemolysis of erythrocytes result in the decompartmentalization of free iron and accelerate the rates of lipoperoxidation and superoxide-dependent formation of OH radicals, which are propagated by reperfusion and reoxygenation in postischemic tissue injury. Simultaneously the activity of GSHPx in the ischemic tissue is decreasing. Selenium and other antioxidants have been observed to prevent synergistically the lipoperoxidation in animals and in man. Pretreatment of rats with vitamin E and selenium prior to iron injections has been shown to prevent the development of seizures to a high degree in a large percentage of experimental rats [55]. There are also reports of the normalization of the EEG of patients with the juvenile type of neuronal ceroid lipofuscinosis (JNCL) after vitamin E and sodium selenite supplementation. In addition, the onset of epilepsy is significantly earlier among JNCL patients not given this antioxidant therapy (11.1 year) than patients receiving antioxidant therapy (13.6 year) [56].

Numerous evidences suggest that selenium may ameliorate some of the adverse metabolic consequences of valproic acid. Valproic acid therapy has been shown to deplete plasma selenium levels, a cofactor required for glutathione peroxidase activity. Selenium supplement may help lower ammonia level in patients with valproate-induced hyperammonemia over long-term treatment. Selenium deficiency may lead to the loss of seizure control, even when the patient is remained on the same dose of valproic acid [57, 58]. Furthermore, Ashrafi et al. [59] concluded that the measurement of serum selenium in patients with intractable epilepsy should be considered.

Advertisement

8. Juvenile type of neuronal ceroid lipofuscinosis

The neuronal ceroid lipofuscinoses (NCL) are a group of recessively inherited neurodegenerative-lysosomal storage diseases of infancy, with an estimated occurrence of 1 in 12,500 live births [60, 61, 62]. Characteristics of the diseases are deposits of ceroid and lipofuscin pigments in the tissues, particularly in the neural tissue, visual failure, and progressive mental retardation. Depending on the age of onset and clinical, electrophysiological, and neuropathological features, the NCLs can be subdivided into the infantile, the late infantile, the juvenile, and the adult type of NCL. The pathogenesis of NCL is unknown. The polyenic acid level with low levels of linoleic acid and an inverse relationship between GSHPx activity and the level of eicosatrienoic acid has been observed in JNCL [63].

The occurrence of the fluorescent pigments suggested the peroxidation of lipids in the etiology of NCL. It is likely that the diseased tissues peroxidize more rapidly than normal tissues and cytotoxic end products of lipoperoxidation cause secondary damage. On a weight basis, ceroid seen in JNCL patients binds five times more iron than the lipofuscin seen in normal elderly individuals. The increased levels of aluminum salts greatly enhance iron-dependent damage to membranes. Heiskala et al. [64] have confirmed the presence of complexable iron and copper in the CSF of patients with NCL and other neurological disorders, and when the pH value of the assay for iron was lowered, the NCL group had substantially more complexable iron in their CSFs. Interestingly aluminum has been observed in CSF and in ceroid lipofuscin pigments of the brain of NCL patients [65]. It is well established that damaged tissue releases metals from protein-bound sites and these metals stimulate peroxidative damage to lipids and other biomolecules.

One of the most essential enzymes counteracting lipoperoxidation is the selenium-containing GSHPx. Two independent reports have demonstrated that erythrocyte GSHPx activity is decreased in JNCL patients [66, 67]. This low GSHPx activity was reversed to normal level by selenium supplementation.

The evaluation of sodium selenite absorption and losses before supplementation of JNCL patients has been studied by using total body counting for 75Se detection. These studies showed that in three JNCL patients, about 55% of the administered 75Se was eliminated during the first 11 days in the feces and about 10% in the urine [68]. Compared to healthy controls (n = 2, percentages 42% and 7%, respectively), findings indicate a reduced absorption of selenium in JNCL patients contrary to a previous report. The low GSHPx activity in NCL patients may indeed reflect a low selenium intake, most probably due to a disturbed absorption of selenium and secondary phenomena due to an inborn error of metabolism. Apart from the low selenium status, also very low vitamin E levels are found in the serum of advanced and hospitalized NCL patients. This can be explained by the recent finding of a pronounced reduction of apoprotein B as well as the whole fraction of very low density lipoprotein (VLDL) in JNCL patients.

JNCL patients (genetically subgroups) have been given daily supplementation of sodium selenite (0.05–0.1 mg/Se/kg of b.w.), vitamin E (α-tocopherol acetate 0.014–0.05 g/kg b.w.), vitamin B2 (0.025–0.05 mg/kg b.w.), and vitamin B6 (0.63–0.8 mg/kg b.w.). The benefits of the therapy are corroborated by the significant negative correlation of GSHPx activity with neurological dysfunction of motor performance, balance, coordination, and speech [69]. The mean age at death has been extended by 4 years as compared to that at the beginning of the century. As the best responders to antioxidant therapy show no neurological dysfunction at the age of over 20 years, there is no doubt that the life expectancy of JNCL patients receiving antioxidants, including selenium, will be significantly prolonged in the future [70]. Complications of the antioxidant therapy have been few and not severe. Six patients have experienced vomiting and nausea when the serum concentration of selenium reached the level of 4.5–5 M. Serum levels up to 4.0 M were usually well tolerated as well as when the sodium selenite was changed to EbselenR (2-Phenyl-1,2-benzoselenazol-3-one).

Advertisement

9. Multiple sclerosis (MS)

Multiple sclerosis is a severe neurodegenerative disease of polygenic etiology affecting the central nervous system. Low levels of polyenic acids are involved in the pathogenesis of both MS and JNCL [71, 72]. In 1972 Thompson et al. found decreased levels of serum linoleate as well as unsaturated fatty acids of brain phospholipids in MS patients. It has also been shown that supplementation with essential fatty acids may improve the clinical status of young MS patients diagnosed early. As in NCL, the selenium may by activating GSHPx (scavenger of organic peroxides) regulate the metabolic transformation of essential fatty acids and biotransformation of these to prostaglandins, thromboxanes, and leukotrienes. Curiously decreased GSHPx activities in erythrocytes have been found in female but not in male MS patients [73].

Blood selenium levels have been reported to be lower in MS patients than in healthy controls [74, 75, 76]. However, selenium concentration has been shown to be normal in plasma and erythrocytes but lowered in platelets of MS patients. Impaired Se status has been found in MS largely in the connection of severe protein-calorie malnutrition. Treatment of MS with Se supplementation does not seem warranted in the absence of demonstrated deficiency. Thus, in the reported selenium-containing antioxidant treatments, the clinical benefit to the course of MS has remained open to speculation.

Advertisement

10. Neurotoxicity

10.1 Mercury intoxication

Mercury is well known for its severe toxicity especially by inhalation [77]. People exposure to Hg is mainly due to environmental pollution and the consumption of fish or other aquatic product [78]. Chronic mercury poisoning is characterized by neurological and psychological symptoms, such as tremor, restlessness, personality changes, anxiety, sleep disturbance, and depression. Symptoms are reversible after cessation of exposure. Because of the blood-brain barrier, there is no central nervous involvement related to inorganic mercury exposure [79]. Selenium interacts in the body with a wide range of toxic metals such as arsenic, cadmium, mercury, copper, silver, and lead. It has been shown to be highly effective in animals in preventing brain damage of organic and inorganic mercury. In postmortem brain samples from persons exposed to mercury vapors, mercury and selenium were found at a molar ratio of approximately 1. This indicates that the brain is the target organ in human exposure to mercury vapors. Mercury and selenium react in various ways. The role of brain selenium in inorganic heavy metal toxicity is thought to be minimal [80, 81]. Mercuric ion bound to selenium is proposed to form a biologically inert complex, leading to increased body burden of both elements. This reaction seems to take place only when a threshold of mercury exposure is exceeded. Selenium influences the oxidation rate of elemental mercury in cases of low GSHPx activity; decreased mercury oxidation may lead to increased brain uptake. Selenium may also together with vitamin E counteract mercury-induced lipid peroxidation.

11. Other CNS diseases related to selenium

11.1 Parkinson’s disease

Parkinson’s disease (PD) is a chronic and progressive movement disorder, meaning that symptoms continue and worsen over time. Oxidative stress is also thought to have a pathogenic role in Parkinson’s disease [82, 83]. Selenium protects cellular elements from oxidative damage and may participate in redox-type reactions. Low plasma selenium concentrations are associated with subtle neurological impairments reflected in soft neurological signs [84, 85]. Plasma Se was the only statistically significant difference of up to 16 elements identified for PD patients [86] relative to Alzheimer's disease patients. Redox-active role is evidenced by an increased lipid peroxidation and reduced glutathione levels [87] and high concentration of iron and free radical generation via autocatalytic mechanisms within neuromelanin-containing catecholaminergic neurons in the substantia nigra. In addition, the observation that exogenous administration of cysteine, N-acetyl cysteine, or glutathione decreased the neurotoxic effects of 6-hydroxydopamine in vitro and in vivo reinforces this hypothesis [88].

11.2 Tardive dyskinesia

Tardive dystonia (TD), a rarer side effect after longer exposure to antipsychotics, is characterized by local or general, sustained, involuntary contraction of a muscle or muscle group, with twisting movements, generally slow, which may affect the limbs, trunk, neck, or face [89, 90]. This condition is characterized by involuntary movements. These abnormal movements most often occur around the mouth. The disorder may range from mild to severe. For some people, it cannot be reversed, while others recover partially or completely. Tardive dyskinesia is seen most often after long-term treatment with antipsychotic medications. Other names for this specific disorder are linguofacial dyskinesia, oral-facial dyskinesia, tardive dystonia, tardive oral dyskinesia, and TD. Many preclinical and clinical studies have investigated the possible role of selenium and other antioxidants. These studies suggest that free radicals are probably involved in the pathogenesis of TD and that vitamin E and selenium could be efficacious in its treatment.

11.3 Duchenne muscular dystrophy

Muscular dystrophy (MD) is a group of genetic diseases involving progressive weakness and degeneration of the muscles that control movement. In some forms of MD, the heart muscles and other involuntary muscles, as well as other organs, are also affected. There are nine distinct types of MD, with myotonic the most common form among adults and Duchenne the most common form among children, primarily affecting males. MD is an incurable, often fatal disease. It is usually obvious by the age of 5 and evolves progressively until it causes disablement and death, around the age of 20. Death commonly results from involvement of the respiratory muscles. It is recessively inherited and linked to sex, and the gene determining DMD has been mapped in the Xp-21 locus. It has an incidence of 1/3000–1/3500 male births, and one third of the cases come from new mutation. Some affected individuals may develop intellectual disturbance due to unknown mechanism, so far. The sister of an affected individual has a 50% chance of carrying the defective gene. The result of the dystrophic locus on the gene is the absence of dystrophin, a rod-shaped protein that is part of the muscle cytoskeleton.

The genetic alteration produces abnormality in the membrane of the muscular fibers that consists of a disturbance in the calcium transport (Ca2+), inside the muscular fibers, which is the base mechanism of cellular degeneration and necrosis. There is fiber necrosis and replacement of fibers by fat. A nucleotide degradation, and decreased muscle ATP and ADP content, has been reported. The ATP is necessary to drive the Na+/K+ pump, which maintains ionic gradients across the sarcolemma; re-sequester the Ca2+ into the cisternae; and have power contraction. The production of ATP can be the result of anaerobic respiration, which breaks glucose down into ATP and lactic acid, or aerobic respiration when ATP, carbon dioxide, and water are formed. A second immediate reserve of energy exists in the form of creatine phosphate, which can donate phosphate to ADP to form ATP, becoming itself creatine. In the resting muscle, glucose is stored as glycogen, and in such a muscle aerobic respiration synthesizes ATP from glucose or fatty acids.

Therapy of DMD has been an elusive goal. Studies with isolated myocytes have shown that lipid peroxidation with an enhanced free radical production can be activated by increasing Ca concentration. Low oxygen saturation in the muscle tissue may stimulate the Il-6 production, a cytokine, which is produced by contracting muscles and released into the blood. The blood circulation of the older Duchenne patients is particularly disturbed. Pedersen et al. [91] have demonstrated that Il-6 affects the metabolic genes, induction of lipolysis, inhibition of insulin resistance, and stimulation of cortisol production. In addition, carbohydrate supplementation during exercise was shown to inhibit the release of Il-6 from contracting muscle. Thus carnitine supplementation is indicated to the Duchenne patients, to make sure that the energy supply will be good.

Johansson et al. [92] hypothesized that increased production of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) may be important underlying mechanisms in myotonic dystrophy. Patients with high body fat mass had significantly increased insulin levels and decreased morning levels of cortisol, ACTH, and testosterone. IL-6 and TNF-alpha levels are increased, and adrenocortical hormone regulation is disturbed in MD. Adiposity may contribute to these disturbances, which may be of importance for decreased adrenal androgen hormone production and metabolic, muscular, and neuropsychiatric dysfunction in MD [92]. Henríquez-Olguín et al. [93] reported that IL-6 is a key metabolic modulator that is released by the skeletal muscle to coordinate a multisystemic response (liver, muscle, and adipocytes) during physical exercise; the alteration of this response in dystrophic muscles may contribute to an abnormal response to contraction and exercise.

Thus, several kinds of antioxidants have been proposed as a treatment since increased levels of thiobarbituric acid (TBA) reactive material have been found in the muscles and blood of DMD boys [94]. Increased amounts of pentane are expelled by the DMD patients [95]. We have previously reported that the biological half-life of 75Se in DMD patients was significantly shorter than in healthy controls [96]. We also reported that patients with myotonic muscular dystrophy, the most common form of muscular dystrophy in adults, show improvement in muscular force and function when treated with selenium and vitamin E [97].

Shimomura et al. [98] observed a group of trained animals, part of which were coenzyme Q10 treated and had to exercise for 30 min on treadmill, in downhill position. CoQ10-treated animals had higher level of CoQ10 In their muscles, and the early rise in creatine kinase and lactic dehydrogenase plasma levels, due to the exercise, was evident at a remarkably significant lower extent, in the treated ones. Similar observations were also made in humans [99]. Therefore we have been treating the Duchenne patients with CoQ10. We have been given two siblings of whom the elder one got practically no antioxidants and the younger one whose antioxidant treatment started at the age of 6 years. Nutrient supplement protocol for DMD patients included sodium selenite 0.05-0.1 mg Se kg−1 b.w. day−1 ; alpha-tocopherol, 10-20 mg kg−1 b.w. day−1; vitamin B2, 0.2 mg kg−1 b.w. day−1; vitamin B6, 5 mg kg−1 b.w. day−1; L-carnitine 10–20 mg kg−1 b.w. day−1; ubiquinone-10 (coenzyme Q10) 3 mg kg−1 b.w. day−1 [100]. In the future the therapy may be by producing functional amounts of dystrophin by skipping the mutated exon like what has been done in the mdx dystrophic mouse [101].

12. Personalized gene therapy

The analytical power of modern methods for DNA analysis has outstripped our capability to interpret and understand the data generated. It is vital that we understand the mechanisms through which mutations affect biochemical pathways and physiological systems [102]: major bcr-abl mRNA nucleic acid amplification assay, genetic analysis of progressive muscular dystrophy, genetic analysis of rearranged immunoglobulin gene, and genetic analysis of malignant tumor. The promise of personalized medicines is enormous, particularly for rare disease [103, 104]. The genetic diversity of Emery-Dreifuss muscular dystrophy (EDMD) predicts that a cure will ultimately depend upon the individual’s defect at the gene level, making this an ideal candidate for a precision medicine approach [105]. Ataluren known as PTC124 is a drug for the treatment of Duchenne muscular dystrophy caused by a nonsense mutation (nmDMD) and cystic fibrosis caused by a nonsense mutation (nmCF). PTC124 can lead to restoration of some dystrophin expression in human Duchenne muscular dystrophy muscles with mutations resulting in premature stops [106]. Eteplirsen, a phosphoramidite morpholino sequence complementary to a portion of exon 51, is designed to force the exclusion of exon 51 from the mature DMD mRNA. Similar drugs targeting other DMD exons are under development and could theoretically restore reading frame in majority of patients. The fact that such drugs rely on specific sequence information and target the proximate cause of the disease makes these one of the first examples of precision genetic medicine [104, 107].

A common denominator to the spectrum of neurological disorders and selenium seems to be oxygen toxicity. Difficulties exist in giving proper weight to the interaction of the components of a complex system like the brain’s antioxidant defense. The presence of multiple and contemporaneous control mechanisms means that a dysregulated system is impaired not only in one but more regulatory or homeostatic mechanism. Supplementation by a single factor like selenium or together with other antioxidants may to a limited extent sustain these mechanisms. However, much more basic research should be done before these complexities can be better understood. The neurological diseases reviewed above have provided the theoretical framework for the continued investigation of the efficacy of the pharmacological manipulation of glutathione concentration and synthesis in treatment of these diseases [108, 109, 110].

Acknowledgments

In memoriam of Faik Atroshi (1949–2019)

Our dear friend, collaborator in research and present coauthor Dr Faik Atroshi, PhD, born on September 22, 1949 in Mosul passed unexpectedly away on February 25, 2019. He was until his retirement docent in University of Helsinki, Senior Researcher in Pharmacology and Toxicology, Adjunct Professor in Clinical Genetics and Clinical Nutrition, and Visiting Professor at different international universities. He will be remembered as an exceptionally dedicated and respectable scientist as well as an inspirational mentor and collaborator. We will miss his enthusiasm, creativity, and desire to continuously learn and to integrate knowledge from various fields of biomedicine.

Abbreviations

DATdementia of the Alzheimer type
LPOlipid peroxides
SODsuperoxide dismutase
CATcatalase
GSHglutathione
GSHPxglutathione peroxidase
Znzinc
Cucopper
Cu-Zn SODcopper zinc superoxide dismutase
ROSreactive oxygen species
CNScentral nervous system
DHPenzyme dehydropeptidase
TNFtumor necrosis factor
GRglutathione reductase
GSHglutathione
ADCarginine decarboxylase
ODCornithine decarboxylase
NADPHnicotinamide adenine dinucleotide phosphate
GSTglutathione-S-transferases
GSSGoxidized glutathione
ADAlzheimer’s disease
·OHhydroxyl radical
E-GSHPxerythrocyte glutathione peroxidase
EEGelectroencephalogram activation: EEG is an essential component in the evaluation of epilepsy
CSFcerebrospinal fluid
JNCLjuvenile neuronal ceroid lipofuscinosis
MSmultiple sclerosis

References

  1. 1. Rayman MP. The importance of selenium to human health. Lancet. 2000;356(9225):233-241
  2. 2. Weiss G, Carver PL. Role of divalent metals in infectious disease susceptibility and outcome. Clinical Microbiology and Infection. Jan 2018;24(1):16-23
  3. 3. Winkel LH, Johnson CA, Lenz M, Grundl T, Leupin OX, Amini M, et al. Environmental selenium research: From microscopic processes to global understanding. Environmental Science & Technology. 2012;46(2):571-579
  4. 4. Fairweather-Tait SJ, Bao Y, Broadley MR, Collings R, Ford D, Hesketh JE, et al. Selenium in human health and disease. Antioxidants & Redox Signaling. 2011;14:1337-1383
  5. 5. Prabhu KS, Lei XG. Selenium. Advances in Nutrition. 2016;7(2):415-417
  6. 6. Thomson CD. Assessment of requirements for selenium and adequacy of selenium status: A review. European Journal of Clinical Nutrition. 2004;58:391-402
  7. 7. Ellis DR, Salt DE. Plants, selenium and human health. Current Opinion in Plant Biology. 2003;6:273-279
  8. 8. Iwasa K, Kanzaki N, Fujishiro T, Hayashi S, Hashimoto S, Kuroda R, et al. Arthroscopic ankle arthrodesis for treating osteoarthritis in a patient with kashin-beck disease. Case Reports in Medicine. 2014;2014:931278
  9. 9. Bains JS, Shaw CA. Neurodegenerative disorders in humans: The role of glutathione in oxidative stress-mediated neuronal death. Brain Research. Brain Research Reviews. 1997;25:335-358
  10. 10. Paglia G, Miedico O, Cristofano A, Vitale M, Angiolillo A, Chiaravalle AE, et al. Distinctive pattern of serum elements during the progression of Alzheimer's disease. Scientific Reports. 2016;6:22769
  11. 11. Halliwell B. Role of free radicals in the neurodegenerative diseases: Therapeutic implications for antioxidant treatment. Drugs & Aging. 2001;18(9):685-716
  12. 12. Cameron A, Rosenfeld J. Nutritional issues and supplements in amyotrophic lateral sclerosis and other neurodegenerative disorders. Current Opinion in Clinical Nutrition and Metabolic Care. 2002;5(6):631-643
  13. 13. Olson D, Westermarck T, Ekvall SW. Seizures and epilepsy. In: Ekvall SW, Ekvall VK, editors. Pediatric Nutrition in Chronic Diseases and Developmental Didsorders. 2nd ed. New York: Oxford University Press; 2005. pp. 93-96 (Chapter 10)
  14. 14. Sinha I, Karagoz K, Fogle RL, Hollenbeak CS, Zea AH, Arga KY, et al. "Omics" of selenium biology: A prospective study of plasma proteome network before and after selenized-yeast supplementation in healthy men. OMICS. 2016;20(4):202-213
  15. 15. Lemire M, Fillion M, Barbosa F Jr, Guimarães JR, Mergler D. Elevated levels of selenium in the typical diet of Amazonian riverside populations. Science of the Total Environment. 2010;408(19):4076-4084
  16. 16. Schiavon M, Pilon-Smits EA. The fascinating facets of plant selenium accumulation—Biochemistry, physiology, evolution and ecology. The New Phytologist. 2017;213(4):1582-1596
  17. 17. Xun P, Bujnowski D, Liu K, Morris JS, Guo Z, He K. Distribution of toenail selenium levels in young adult Caucasians and African Americans in the United States: The CARDIA Trace Element Study. Environmental Research. 2011;111(4):514-519
  18. 18. Zachara BA, Pawluk H, Bloch-Boguslawska E, Sliwka KM, Korenkiewicz J, Skok Z, et al. Tissue level, distribution, and total body selenium content in healthy and diseased humans in Poland. Archives of Environmental Health. 2001;56:461-466
  19. 19. Ejima A, Watanabe C, Koyama H, Matsuno K, Satoh H. Determination of selenium in the human brain by graphite furnace atomic absorption spectrometry. Biological Trace Element Research. 1996;54:9-21
  20. 20. Wajner M, Latini A, Wyse ATS, Dutra-Filho CS. The role of oxidative damage in the neuropathology of organic acidurias: Insights from animal studies. Journal of Inherited Metabolic Disease. 2004;27(4):427-448
  21. 21. Larsen PR, Berry MJ. Nutritional and hormonal regulation of thyroid hormone deiodinases. Annual Review of Nutrition. 1995;15:323-352
  22. 22. Wrobel JK, Power R, Toborek M. Biological activity of selenium: Revisited. IUBMB Life. 2016;68(2):97-105
  23. 23. Parantainen J, Sankari S, Atroshi F. Biological functions of silicon, selenium and glutathione peroxidase (GSH Px) explained in terms of semiconduction. In: Hurly LS, Keen CL, Bo L, Rucker RB, editors. Trace Elements in Man and Animals 6. New York & London: Plenum Press; 1988. pp. 359-360
  24. 24. Hall ED, Yonkers PA. Attenuation of motor nerve terminal repetitive discharge by the 21-aminosteroid tirilazad: Evidence of a neural calcium antagonist action. Brain Research. 1998;779(1-2):346-349
  25. 25. Sheldon R, Jiang X, Francisco C, Christen S, Vexler ZS, Tauber MG, et al. Manipulation of antioxidant pathways in neonatal murine brain. Pediatric Research. 2004;56(4):656-662
  26. 26. Nakano M, Sugioka K, Naito I, Takekoshi S, Niki E. Novel and potent antioxidants on membrane phospholipid peroxidation: 2-hydroxy estrone and 2-hydroxy estradiol. Biochemical and Biophysical Research Communications. 1987;142:919-924
  27. 27. Takahashi H. Studies on homocarnosine in cerebrospinal fluid in infancy and childhood. Part II. Homocarnosine levels in cerebrospinal fluid from children with epilepsy, febrile convulsion or meningitis. Brain and Development. 1981;3:263-270
  28. 28. Atroshi F, Westermarck T. High concentrations of erythrocyte glutathione in patients with neurological disorders: Possible clinical implications. In: Rice-Evans C, editor. Free Radical Oxidant Stress & Drug Action. London: Richelieu Press; 1987. pp. 419-424
  29. 29. Yoshida S, Hori E, Ura S, Haratake M, Fuchigami T, Nakayama M. A comprehensive analysis of selenium-binding proteins in the brain using its reactive metabolite. Chemical & Pharmaceutical Bulletin (Tokyo). 2016;64(1):52-58
  30. 30. Saedi MS, Smith CG, Frampton J, Chambers I, Harrison PR, Sunde RA. Effect of selenium status on MRNA levels for glutathione peroxidase in rat liver. Biochemical and Biophysical Research Communications. 1988;153:855-861
  31. 31. Chung A-S, Maines MD. Effect of selenium on glutathione metabolism. Induction of gamma-glutamylcysteine synthetase and glutathione reductase in the rat liver. Biochemical Pharmacology. 1981;30:3217-3223
  32. 32. Takebe G, Yarimizu J, Saito Y, Hayashi T, Nakamura H, Yodoi J, et al. A comparative study on the hydroperoxide and thiol specificity of the glutathione peroxidase family and selenoprotein P. The Journal of Biological Chemistry. 2002;277:41254-41258
  33. 33. LaFemina MJ, Sheldon RA, Ferriero DM. Acute hypoxia-ischemia results in hydrogen peroxide accumulation in neonatal but not adult mouse brain. Pediatric Research. 2006;59:680-683
  34. 34. McLean CW, Mirochnitchenko O, Claus CP, Noble-Haeusslein LJ, Ferriero DM. Overexpression of glutathione peroxidase protects immature murine neurons from oxidative stress. Developmental Neuroscience. 2005;27:169-175
  35. 35. Wang H, Cheng E, Brooke S, Chang P, Sapolsky R. Over-expression of antioxidant enzymes protects cultured hippocampal and cortical neurons from necrotic insults. Journal of Neurochemistry. 2003;87:1527-1534
  36. 36. Miller BA, Perez RS, Shah AR, Gonzales ER, Park TS, Gidday JM. Cerebral protection by hypoxic preconditioning in a murine model of focal ischemiareperfusion. NeuroReport. 2001;12:1663-1669
  37. 37. Bernaudin M, Tang Y, Reilly M, Petit E, Sharp FR. Brain genomic response following hypoxia and re-oxygenation in the neonatal rat. Identification of genes that might contribute to hypoxia-induced ischemic tolerance. Journal of Biological Chemistry. 2002;277:39728-39738
  38. 38. Mu D, Jiang X, Sheldon RA, Fox CK, Hamrick SE, Vexler ZS, et al. Regulation of hypoxia-inducible factor 1 alpha and induction of vascular endothelial growth factor in a rat neonatal stroke model. Neurobiology of Disease. 2003;14:524-534
  39. 39. Song H, Ren X, Liu P. Distribution and inhibition effect of seleno-L-methionine on 4T1 mouse mammary carcinoma. International Journal of Physiology, Pathophysiology and Pharmacology. 2015;7(2):76-86
  40. 40. Berman K, Brodaty H. Tocopherol (Vitamin E) in Alzheimer's disease and other neurodegenerative disorders. CNS Drugs. 2004;18(12):807-825
  41. 41. Huebbe P, Jofre-Monseny L, Boesch-Saadatmandi C, Minihane AM, Rimbach G. Effect of apoE genotype and vitamin E on biomarkers of oxidative stress in cultured neuronal cells and the brain of targeted replacement mice. Journal of Physiology and Pharmacology. 2007;58(4):683-698
  42. 42. Milton NGN. Role of hydrogen peroxide in the aetiology of Alzheimer's disease: Implications for treatment. Drugs and Aging. 2004;21(2):81-100
  43. 43. Chang YT, Chang WN, Tsai NW, Huang CC, Kung CT, Su YJ, et al. The roles of biomarkers of oxidative stress and antioxidant in Alzheimer’s disease: A systematic review. BioMed Research International. 2014;2014: Article ID 182303, 14 p. DOI: 10.1155/2014/182303
  44. 44. Kuroda Y, Kobayashi K, Ichikawa M, Kawahara M, Muramoto K. Application of long-term cultured neurons in aging and neurological research: Aluminum neurotoxicity, synaptic degeneration and Alzheimer's disease. Gerontology. 1995;41(Suppl 1):2-6
  45. 45. Sumathi T, Shobana C, Kumari BR, Nandhini DN. Protective role of Cynodon dactylon in ameliorating the aluminium-induced neurotoxicity in rat brain regions. Biological Trace Element Research. 2011;144(1-3):843-853
  46. 46. Malt EA, Dahl RC, Haugsand TM, Ulvestad IH, Emilsen NM, Hansen B, et al. Health and disease in adults with Down syndrome. Tidsskrift for den Norske laegeforening: Tidsskrift for praktisk medicin, ny raekke. 2013;133(3):290-294
  47. 47. Weijerman ME, de Winter JP. Clinical practice. The care of children with Down syndrome. European Journal of Paediatrics. 2010;169(12):1445-1452
  48. 48. Becker L, Mito T, Takashima S, Onodera K. Growth and development of the brain in Down syndrome. Progress in Clinical and Biological Research. 1991;373:133-152
  49. 49. Brooksbank BW, Balazs R. Superoxide dismutase, glutathione peroxidase and lipoperoxidation in Down’s syndrome fetal brain. Brain Research. 1984;318:37-44
  50. 50. Ellis JM, Tan HK, Gilbert RE, Muller DP, Henley W, Moy R, et al. Supplementation with antioxidants and folinic acid for children with Down's syndrome: Randomised controlled trial. BMJ. 2008;336(7644):594-597
  51. 51. Antila E, Nordberg UR, Syvaoja EL, Westermarck T. Selenium therapy in Down syndrome (DS): A theory and a clinical trial. Advances in Experimental Medicine and Biology. 1990;264:183-186
  52. 52. Sinet PM, Lejeune J, Jerome H. Trisomy 21 (Down's syndrome), glutathione peroxidase, hexose monophosphate shunt and IQ. Life Sciences. 1979;24:29-34
  53. 53. Guerrini R. Valproate as a mainstay of therapy for pediatric epilepsy. Pediatric Drugs. 2006;8(2):113-129
  54. 54. Pillai R, Uyehara-Lock JH, Bellinger FP. Selenium and selenoprotein function in brain disorders. IUBMB Life. 2014;66(4):229-239
  55. 55. Willmore LJ. Post-traumatic epilepsy: Cellular mechanisms and implications for treatment. Epilepsia. 1990;31(Suppl 3):S67-S73
  56. 56. Santavuori P, Westermarck T, Rapola J, Pohja P, Moren R, Lappi M, et al. Antioxidant treatment in Spielmeyer-Sjögren disease. Acta Neurologica Scandinavica. 1985;71:136-145
  57. 57. Humphreys S, Murti G, Holmes MD. Selenium deficiency and valproate-induced hyperammonemia: 1.320. Epilepsia. 2004;45(Suppl 7):122-123
  58. 58. Płonka-Półtorak E, Zagrodzki P, Nicol F, Kryczyk J, Bartoń H, Westermarck T, et al. Antioxidant agents and physiological responses in adult epileptic patients treated with lamotrigine. Pharmacological Reports. 2013;65(1):99-106
  59. 59. Ashrafi MR, Shabanian R, Abbaskhanian A, Nasirian A, Ghofrani M, Mohammadi M, et al. Selenium and intractable epilepsy: Is there any correlation? Pediatric Neurology. 2007;36(1):25-29
  60. 60. Herrmann P, Druckrey-Fiskaaen C, Kouznetsova E, Heinitz K, Bigl M, Cotman SL, et al. Developmental impairments of select neurotransmitter systems in brains of Cln3(Deltaex7/8) knock-in mice, an animal model of juvenile neuronal ceroid lipofuscinosis. Journal of Neuroscience Research. 2008;86(8):1857-1870
  61. 61. Kyttala A, Lahtinen U, Braulke T, Hofmann SL. Functional biology of the neuronal ceroid lipofuscinoses (NCL) proteins. Biochimica et Biophysica Acta. 2006;1762:920-933
  62. 62. Mole SE. Batten disease: Four genes and still counting. Neurobiology of Disease. 1998;1998(5):287-303
  63. 63. Banerjee P, Dasgupta A, Siakotos AN, Dawson G. Evidence for lipase abnormality: High levels of free and triacylglycerol forms of unsaturated fatty acids in neuronal ceroid-lipofuscinosis tissue. American Journal of Medical Genetics. 1992;42(4):549-554
  64. 64. Heiskala H, Gutteridge JMC, Westermark T, Alanen T, Santavuori P. Bleomycin-detectable iron and phenanthroline-detectable copper in the cerebrospinal fluid of patients with neuronal ceroid-lipofuscinoses. American Journal of Medical Genetics. Supplement. 1988;5:193-202
  65. 65. Johansson E, Lindh U, Westermarck T, Heiskala H, Santavuori P. Altered elemental profiles in neuronal ceroid lipofuscinosis. Journal of Trace Elements and Electrolytes in Health and Disease. 1990;4(3):139-142
  66. 66. Benedict JW, Sommers CA, Pearce DA. Progressive oxidative damage in the central nervous system of a murine model for juvenile Batten disease. Journal of Neuroscience Research. 2007;85(13):2882-2891
  67. 67. Hall NA, Lake BD, Patrick AD. Recent biochemical and genetic advances in our understanding of Batten's disease (ceroid-lipofuscinosis). Developmental Neuroscience. 1991;13(4-5):339-344
  68. 68. Westermarck T, Erkki A, Faik A. Vitamin E therapy in neurological diseases. In: Packer L, Fuchs J, editors. Vitamin E in Health and Disease. New York, Basel, Hong Kong: Marcel Dekker; 1993. pp. 799-806
  69. 69. Piattella L, Cardinali C, Zamponi N. Papa O Spielmeyer-Vogt disease: Clinical and neurophysiological aspects. Child's Nervous System. 1991;7(4):226-230
  70. 70. Santavuori P, Heiskala H, Westermark T, Sainio K, Moren R. Experience over 17 years with antioxidant treatment in Spielmeyer-Sjögren disease. American Journal of Medical Genetics. Supplement. 1988;5:265-274
  71. 71. Gourraud PA, Harbo HF, Hauser SL, Baranzini SE. The genetics of multiple sclerosis: An up-to-date review. Immunological Reviews. 2012;248(1):87-103
  72. 72. Lvovs D, Favorova OO, Favorov AV. A polygenic approach to the study of polygenic diseases. Acta Naturae. 2012;4(3):59-71
  73. 73. Clausen J, Jensen GE, Nielsen SA. Selenium in chronic neurologic diseases. In: Schrauzer GN, editor. Biological Trace Element Research. Clifton, New Jersey: The Humana Press; 1988. pp. 179-203
  74. 74. Komatsu F, Kagawa Y, Kawabata T, Kaneko Y, Kudoh H, Purvee B, et al. Influence of essential trace minerals and micronutrient insufficiencies on harmful metal overload in a Mongolian patient with multiple sclerosis. Current Aging Science. 2012;5(2):112-125
  75. 75. Mai J, Sorensen PS, Hansen JC. High dose antioxidant supplementation to MS patients. Effects on glutathione peroxidase, clinical safety, and absorption of selenium. Biological Trace Element Research. 1990;24(2):109-117
  76. 76. Socha K, Kochanowicz J, Karpińska E, Soroczyńska J, Jakoniuk M, Mariak Z, et al. Dietary habits and selenium, glutathione peroxidase and total antioxidant status in the serum of patients with relapsing-remitting multiple sclerosis. Nutrition Journal. 2014;13:62
  77. 77. Kosnett M. Mercury. In: Olson K, editor. Poisoning & Drug Over-dose. NY, USA: The McGraw-Hill Companies, Inc; 2012. pp. 271-276
  78. 78. McNutt M. Mercury and health. Science. 2013;341:1430-1430
  79. 79. Jarup L. Heavy metals in the environment. Paediatric and Perinatal Epidemiology. 2003;17(2):221-222
  80. 80. Nehru B, Dua R. The effect of dietary selenium on lead neurotoxicity. Journal of Environmental Pathology, Toxicology and Oncology. 1997;16(1):47-50
  81. 81. Ruszkiewicz J, Albrecht J. Changes in the mitochondrial antioxidant systems in neurodegenerative diseases and acute brain disorders. Neurochemistry International. 2015;88:66-72
  82. 82. Dézsi L, Vécsei L. Monoamine oxidase B inhibitors in Parkinson's disease. CNS & Neurological Disorders—Drug Targets. 2017. [Epub ahead of print]
  83. 83. Lacher SE, Slattery M. Gene regulatory effects of disease-associated variation in the NRF2 network. Current Opinion in Toxicology. 2016;1:71-79
  84. 84. Dominiak A, Wilkaniec A, Jęśko H, Czapski GA, Lenkiewicz AM, Kurek E, et al. Selol, an organic selenium donor, prevents lipopolysaccharide-induced oxidative stress and inflammatory reaction in the rat brain. Neurochemistry International. 2017;108:66-77
  85. 85. Shahar A, Patel KV, Semba RD, Bandinelli S, Shahar DR, Ferrucci L, et al. Plasma selenium is positively related to performance in neurological tasks assessing coordination and motor speed. Movement Disorders. 2010;25(12):1909-1915
  86. 86. McIntosh KG, Cusack MJ, Vershinin A, Chen ZW, Zimmerman EA, Molho ES, et al. Evaluation of a prototype point-of-care instrument based on monochromatic X-ray fluorescence spectrometry: Potential for monitoring trace element status of subjects with neurodegenerative disease. Journal of Toxicology and Environmental Health. Part A. 2012;75(21):1253-1268
  87. 87. Delanty N, Dichter M. Antioxidant therapy in neurologic disease. Archives of Neurology. 2000;57(9):1265-1270
  88. 88. Soto-Otero R, Mendez-Alvarez E, Hermida-Ameijeiras A, et al. Autoxidation and neurotoxicity of 6-hydroxydopamine in the presence of some antioxidants: Potential implication in relation to the pathogenesis of Parkinson's disease. Journal of Neurochemistry. 2000;74:1605-1612
  89. 89. Burke RE, Fahn S, Jankovic J, Marsden CD, Lang AE, Gollomp S, et al. Tardive dystonia: Late-onset and persistent dystonia caused by antipsychotic drugs. Neurology. 1982;32(12):1335-1346
  90. 90. Rakesh G, Muzyk A, Szabo ST, Gupta S, Pae CU, Masand P. Tardive dyskinesia: 21st century may bring new treatments to a forgotten disorder. Annals of Clinical Psychiatry. 2017;29(1):e9-e20
  91. 91. Pedersen BK, Steensberg A, Fischer C, Keller C, Keller P, Plomgaard P, et al. Searching for the exercise factor: Is IL-6 a candidate? Journal of Muscle Research and Cell Motility. 2003;24(2-3):113-119
  92. 92. Johansson A, Carlström K, Ahrén B, Cederquist K, Krylborg E, Forsberg H, et al. Abnormal cytokine and adrenocortical hormone regulation in myotonic dystrophy. The Journal of Clinical Endocrinology and Metabolism. 2000;85(9):3169-3176
  93. 93. Henríquez-Olguín C, Altamirano F, Valladares D, López JR, Allen PD, Jaimovich E. Altered ROS production, NF-κB activation and interleukin-6 gene expression induced by electrical stimulation in dystrophic mdx skeletal muscle cells. Biochimica et Biophysica Acta. 2015;1852(7):1410-1419
  94. 94. Jackson MJ, Edwards RHT. Free radicals and trials of antioxidant therapy in muscle diseases. Advances in Experimental Medicine and Biology. 1990;1990(264):485-491
  95. 95. Grinio LP, Orlov ON, Prilipko LL, Kagan VE. Lipid peroxidation in children with Duchenne’s hereditary myopathy. Bulletin of Experimental Biology and Medicine. 1984;98:423-425
  96. 96. Westermarck T, Rahola T, Kallio A-K, Suomela M. Long term Turnover of selenite-Se in children with motor disorders. Klinische Pädiatrie. 1982;194:301-302
  97. 97. Westermarck T, Aberg L, Santavuori P, Antila E, Edlund P, Atroshi F. Evaluation of the possible role of coenzyme Q10 and vitamin E in juvenile neuronal ceroid-lipofuscinosis (JNCL). Molecular Aspects of Medicine. 1997;18(Suppl):S259-S262
  98. 98. Shimomura Y, Suzuki M, Sugiyama S, et al. Protective effect of coenzyme Q10 on exercise induced muscular injury. Biochemical and Biophysical Research Communications. 1991;176:349-355
  99. 99. Littarru GP, Tiano L, et al. Clinical aspects of coenzyme Q 10: An update. Nutrition. 2010;26:250-254
  100. 100. Westermarck T, Antila E, Kaksonen S, Laakso J, Härkönen M, Atroshi F. Long-term follow-up of two Duchenne muscle dystrophy patients treated with antioxidants. In: Hiramatsu M, Yoshikawa T, Inoue M, editors. Food and Free Radicals. New York, London: Plenum Press; 1997. pp. 161-163
  101. 101. Lu QL, Mann CJ, Lou F, Bou-Gharios G, Morris GE, et al. Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse. Nature Medicine. 2003;9(8):1009-1014
  102. 102. Bonthron DT, Foulkes WD. Genetics meets pathology—An increasingly important relationship. The Journal of Pathology. 2017;241(2):119-122
  103. 103. Lim LE, Rando TA. Technology insight: Therapy for Duchenne muscular dystrophy-an opportunity for personalized medicine? Nature Clinical Practice. Neurology. 2008;4(3):149-158
  104. 104. Miceli MC, Nelson SF. The case for eteplirsen: Paving the way for precision medicine. Molecular Genetics and Metabolism. 2016;118(2):70-71
  105. 105. Pillers DA, Von Bergen NH. Emery-Dreifuss muscular dystrophy: A test case for precision medicine. The Application of Clinical Genetics. 2016;9:27-32
  106. 106. Nelson SF, Crosbie RH, Miceli MC, Spencer MJ. Emerging genetic therapies to treat Duchenne muscular dystrophy. Current Opinion in Neurology. 2009;22(5):532-538
  107. 107. Mendell JR, Goemans N, Lowes LP, Alfano LN, Berry K, Shao J, et al. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Annals of Neurology. 2016;79(2):257-271
  108. 108. Atroshi F, Antila E, Westermarck T. The role of selenium in epilepsy and other neurological disorders. Epileptologia. 2007;15:211-224
  109. 109. Dworkin RH. Linoleic acid and multiple sclerosis. Lancet. 1981;1(8230):1153-1154
  110. 110. Tiano L, Padella L, Santoro L, Carnevali P, Principi F, Brugè F, et al. Prolonged coenzyme Q10 treatment in Down syndrome patients: Effect on DNA oxidation. Neurobiology of Aging. 2012;33(3):626.e1-626.e8

Written By

Erkki Antila, Tuomas Westermarck, Arno Latvus and Faik Atroshi

Reviewed: 05 March 2020 Published: 14 October 2020