Next Article in Journal
Bidirectional Resonant Converter for DC Microgrid Applications
Previous Article in Journal
Application of Green Surfactants in the Remediation of Soils Contaminated by Hydrocarbons
Previous Article in Special Issue
The Role of Oxidative Stress and the Importance of miRNAs as Potential Biomarkers in the Development of Age-Related Macular Degeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomolecular Modifications Linked to Oxidative Stress in Amyotrophic Lateral Sclerosis: Determining Promising Biomarkers Related to Oxidative Stress

1
Department of Neurology, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
2
Department of Internal Medicine, Ibaraki Western Medical Center, Tsukuba University Hospital Kensei Area Medical Education Center, Chikusei 308-0813, Ibaraki, Japan
3
Department of Internal Medicine, Ibaraki Western Medical Center, Chikusei 308-0813, Ibaraki, Japan
*
Author to whom correspondence should be addressed.
Processes 2021, 9(9), 1667; https://doi.org/10.3390/pr9091667
Submission received: 18 August 2021 / Revised: 12 September 2021 / Accepted: 13 September 2021 / Published: 15 September 2021
(This article belongs to the Special Issue Advances of Redox Status in Disease)

Abstract

:
Reduction–oxidation reactions are essential to cellular homeostasis. Oxidative stress transcends physiological antioxidative system damage to biomolecules, including nucleic acids and proteins, and modifies their structures. Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease. The cells present in the central nervous system, including motor neurons, are vulnerable to oxidative stress. Neurodegeneration has been demonstrated to be caused by oxidative biomolecular modifications. Oxidative stress has been suggested to be involved in the pathogenesis of ALS. Recent progress in research on the underlying mechanisms of oxidative stress in ALS has led to the development of disease-modifying therapies, including edaravone. However, the clinical effects of edaravone remain limited, and ALS is a heretofore incurable disease. The reason for the lack of reliable biomarkers and the precise underlying mechanisms between oxidative stress and ALS remain unclear. As extracellular proteins and RNAs present in body fluids and represent intracellular pathological neurodegenerative processes, extracellular proteins and/or RNAs are predicted to promise diagnosis, prediction of disease course, and therapeutic biomarkers for ALS. Therefore, we aimed to elucidate the underlying mechanisms between oxidative stress and ALS, and promising biomarkers indicating the mechanism to determine whether therapy targeting oxidative stress can be fundamental for ALS.

1. Introduction

Among the various biochemical responses supporting our life, reduction–oxidation (redox) reactions play important roles in energy production by mitochondria, intracellular protein quality control, and intracellular signal transduction cascade. Several types of reactive species, including reactive oxygen species (ROS), reactive nitrogen species, and reactive sulfur species, are essential factors in redox signaling in biology [1,2]. These reactive species are produced by cellular homeostatic metabolism and exogenous pro-oxidant factors, including environmental and atmospheric pollution, heavy metals, or tobacco [3]. Among these reactive species, two ROS species, hydrogen peroxide (H2O2) and superoxide anion radical (O2•−), are key redox signaling agents generated under the tight control of growth factors and cytokines by over 40 enzymes [4]. The physiological concentrations of ROS regulate numerous cellular functions, including cellular signaling pathways, phosphorylation of proteins, signaling transduction via cell membrane receptors, and activation of ionic channels and transcription factors; the physiological intracellular concentration of H2O2 is maintained at approximately 1–100 nM and that of O2•− is 10−11 M, much lower than that of H2O2 [5]. Supraphysiological concentrations of H2O2 and O2•− lead to nonspecific oxidation and irreversible damage to biomolecules, resulting in growth arrest and cell death, as demonstrated in various diseases, including neurodegenerative diseases [1,2,6]. Conversely, to prevent biomolecules from nonspecific and irreversible damage, the cells have evolved several antioxidant defenses, including activation of various antioxidant molecules to counteract the injurious effects of ROS. As such, a number of antioxidant enzymes, including superoxide dismutase (SOD), catalase, glutathione peroxidase (GPX), and glutathione reductase (GSHRd), exist in cells [7].
The cellular redox state maintains an equilibrium between oxidation and reduction reactions. Oxidative stress, which is the consequence of oxidative damage to cells and organs by increased generation of ROS and/or decreased effects of antioxidant defenses, is responsible for damage to various biomolecules, including nucleic acids and proteins, and contributes to stress-induced aging [8]. Neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), are incurable and characterized by progressive neuronal cell loss in the central nervous system (CNS). Since the ability of cells to maintain their normal redox state is reduced during the normal aging process, aging is the most important risk factor for neurodegenerative diseases [9]. Furthermore, the CNS is enriched in polyunsaturated fatty acids that consume a large amount of oxygen and redox-active metals that actively participate in ROS generation, whereas the CNS has low levels of antioxidants. The brain is more vulnerable to oxidative stress than other parts of the body [10,11]. Therefore, the evidence of a significant connection between oxidative stress and neurodegenerative disease is increasing, and therapies aimed at reducing cellular ROS levels are expected to be neuroprotective therapies for multiple neurodegenerative diseases [11].
ALS is the most common adult-onset lethal motor neuron disease and is characterized by the selective degeneration of both upper and lower motor neurons. These degenerations cause progressive muscle weakness and atrophy, leading to respiratory failure and death within a few years following disease onset [12,13]. The ALS-associated gene, superoxide dismutase 1 (SOD-1), an enzyme that converts superoxide radicals into hydrogen peroxide, was identified in 1993 [14]. Since then, although a large number of pathogenic hypotheses have been proposed, oxidative stress has been considered one of the pathogenic hypotheses in ALS [15]. In addition, the evidence of perturbed cellular redox regulatory mechanisms and associated proteins in both sporadic and familial ALS is increasing [16]. Edaravone, a free radical scavenger that eliminates lipid peroxides and hydroxyl radicals, was recently approved for ALS patients, suggesting that modifying the redox regulation could be a therapeutic strategy for ALS [17]. However, as the precise understanding of the association between disturbed redox regulation and motor neuron death is unclear, the administration of edaravone is not particularly effective and is not a fundamental therapy. To develop more successful therapies, the establishment of diagnostic biomarkers for early phase ALS and predictive and pharmacodynamic biomarkers for initiation of therapy is important. However, the vast majority of ALS cases are sporadic; thus, the diagnosis of ALS is not determined by genetic screening [12]. A diagnosis of ALS is currently based on a combination of clinical assessment and electrophysiological examination using El Escorial criteria and Awaji criteria, and no reliable diagnostic biomarkers have been established [18,19].
As a definite diagnosis of neurodegenerative diseases comes from histological observation of the spinal cords and brains, accurate antemortem diagnosis of these diseases is currently impossible. A biomarker is characterized as an indicator of normal biological processes or pathogenic processes that must be objectively measured and evaluated [20]. Therefore, noninvasive or minimally invasive biomarkers, including easily accessible body fluids or tissues that reflect disease-specific pathological events in the CNS, are needed. Liquid biopsy targeting the patient’s body fluids, including serum and cerebrospinal fluid (CSF), is less invasive than conventional tissue biopsy and can be an alternative diagnostic tool [21]. Extracellular proteins and RNAs representing homeostatic intracellular environments are secreted from several cell types in the CNS tissue, and neuronal interactions via extracellular proteins and RNAs exert key functions in neurite growth, synaptic function, and neuronal regeneration [22,23]. Therefore, extracellular proteins and RNAs have been considered promising biomarker candidates for various types of neurodegenerative diseases [24].
In this review, we summarize the oxidative biomolecular alterations involved in the pathogenesis of ALS and promising treatments related to oxidative stress in these diseases. Moreover, we discuss promising biomarker candidates linked to oxidative stress in ALS.

2. Oxidative Biomolecular Modification Leading to Neurodegeneration

As described in the introduction section, aging leads to increased oxidative stress and the brain is highly susceptible to ROS damage, whereas the function of antioxidant systems declines with age [25,26]. Therefore, oxidative biomolecular modifications, including proteins and/or nucleic acids, increase with age, especially in the brain, leading to neurodegeneration induced by oxidative stress [26]. Abnormal protein aggregation, including amyloid in Alzheimer’s disease (AD), tau in Parkinson’s disease (PD), and transactive response DNA/RNA binding protein of 43 kDa (TDP-43) in ALS, are pathological hallmarks of neurodegenerative diseases [27,28,29]. Therefore, elimination of these abnormal protein aggregates is a therapeutic strategy for fundamental therapy of neurodegenerative diseases, and changes in the expression levels of protein and RNA in body fluids related to these abnormal protein aggregates must be a biomarker. In addition, accumulation of DNA and RNA damage and impairment of DNA repair pathways have been found primarily in neuronal tissues, and changes in expression levels of noncoding RNA due to oxidative stress have also been recognized in neuronal tissues, suggesting that oxidative damage to DNA and RNA causes neurodegenerative diseases. In this section, we review oxidative biomolecular modifications due to redox dysregulation (Figure 1).

2.1. Oxidative Modifications of Proteins Linked to Protein Aggregation

Since the rate of the oxidant reaction depends on the concentration of the target and proteins are major components of most biological systems, proteins are major targets for oxidative stress; therefore, protein oxidative modifications occur easily [30,31].
Although physiological oxidative stress is useful for physiological homeostasis, excessive oxidative stress can lead to conformational changes, protein misfolding, and accumulation of insoluble aggregates, resulting in neurodegeneration [32]. ROS can generate various post-translational modifications, such as the alteration of amino acids [30,33,34]. Among the amino acids, cysteine residues, which are critical for protein folding, function, and stability, are crucial for the maintenance of cellular redox homeostasis, and internal disulfide bond formation between Cys32 and Cys41 by oxidation have been shown to cause protein aggregation [35,36]. Additionally, oxidative stress promotes protein aggregation by modulating chaperone protein activity, such as preventing the interaction between chaperone proteins and heat shock proteins and perturbing the translational process [37]. The endoplasmic reticulum (ER), which has a higher ratio of oxidation similar to extracellular space, is the main component of protein folding in eukaryotic cells, and ER-associated degradation plays a crucial role in preventing protein accumulation and aggregation [38]. Oxidative stress and dysregulation of the ER redox balance can inhibit ER-associated degradation in response to unfolded proteins. These changes lead to protein misfolding, ER stress, and protein aggregation, resulting in neurodegeneration [1]. Moreover, oxidative stress also inhibits classical nuclear import via induction of relocation of Nup153 and importin-β and impairs autophagy, leading to protein aggregation [39,40]. Furthermore, protein aggregations tend to accumulate in foci, and oxidative stress acts as a sensor that triggers protein recruitment into foci [41].

2.2. Oxidative DNA Damage Linked to Apoptosis

A number of exogenous and endogenous agents, including oxidative stress and even normal cellular processes, can induce DNA damage, suggesting that oxidative DNA damage is an inevitable consequence of cellular metabolism. Several DNA repair pathways, including DNA base excision repair (BER) and the nucleotide excision repair pathway, function as correction mechanisms for the daily amounts of DNA damage [42,43]. This said, ROS levels are increased with age, in contrast to the decrease in antioxidant system ability, and neurons are highly susceptible to DNA damage [44,45]. Moreover, higher levels of oxidative DNA damage and impairment of DNA repair pathways have been shown to lead to increased p53 activity, ultimately resulting in apoptosis. As such, the accumulation of oxidative DNA damage is thought to contribute to neurodegeneration [46].
Mitochondria, which are the heart of redox reactions and generate ROS species, play essential roles in the survival of various cells, including motor neurons [47]. Mitochondrial DNA is more exposed to ROS than nuclear DNA because mitochondrial DNA exists in the proximity of the inner mitochondrial membrane in which ROS is formed, lacks protective histones, and has less efficient repair [48]. Age-associated increase in oxidative mitochondrial DNA damage reduces mitochondrial axonal transport. Moreover, oxidative damage of mitochondrial DNA genes encoding subunits of the mitochondrial respiratory chain due to ROS impairs adenosine triphosphate production [49,50]. These alterations lead to mitochondrial dysfunction and, ultimately, apoptosis. Furthermore, oxidative mitochondrial DNA damage accelerates oxidative stress [51]. Additionally, the accumulation of mutations in both nuclear and mitochondrial DNA due to oxidative damage also leads to the production of altered proteins, resulting in alteration of protein constructs and abnormal aggregation [52,53].

2.3. Oxidative RNA Damage

RNA is essential for maintaining cellular homeostasis and plays an important role in protein synthesis. As RNA accounts for approximately 80% of the total cellular nucleic acids, RNA can also be a major target of oxidative stress [54]. In addition, RNA may be more vulnerable to oxidative damage than DNA because of its characteristic single-stranded structure and lack of protection by hydrogen bonding and specific proteins [55,56]. Direct RNA strand breaks, translation errors, and protein synthesis disorders are induced by oxidative damage [57,58,59]. Moreover, since the spatial positioning of messenger RNA (mRNA) exists close to the mitochondria and temporal dynamics of RNA oxidation is shorter than that of RNA translation, a large amount of mRNA oxidation occurs [60]. RNA oxidation is not random, but instead highly selective, and abnormal processing of proteins and a decrease in protein expression leading to protein misfolding are caused by oxidative modification of mRNA [61,62]. Therefore, oxidative RNA modification, which occurs not only in protein-coding RNAs but also in noncoding RNAs, also contributes to aging and the underlying mechanisms of neurodegeneration. Noncoding RNAs, including microRNAs (miRNAs) and circular RNAs (circRNAs), are regulators of gene expression, crucial neuronal differentiation, survival, and activity. Several miRNAs can be oxidized themselves and regulate the expression levels of many genes involved in antioxidant defense pathways [63]; for example, oxidative modification of miR-184 reduced expression levels of Bcl-xL and Bcl-w due to misrecognitions, leading to apoptosis [64], and miR-27a influences redox homeostasis due to reduced expression levels of nuclear factor E2-related factor 2 (NRF2), which is a major regulator of antioxidant elements [65]. In addition, several circRNAs can be related to neurodegeneration via modulation of oxidative stress; for example, circHIPK3 can regulate oxidative stress due to work through a signaling network that consists of circHIPK3 and miR-29a [66].

3. Biomolecular Modifications Associated with Oxidative Stress in ALS

Aging and cellular senescence are risk factors for various neurodegenerative diseases. Aberrant protein aggregation is considered a pathological hallmark of neurodegenerative diseases such as β-amyloid in AD, α-synuclein in PD, and TDP-43 in ALS. Age-associated increase in oxidative stress contributes to protein misfolding and aggregation, and these protein aggregations cause upregulation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity, oxidant generation, and overactivated microglia, resulting in altered fluidity, permeability, transport, and metabolic processes [10,67]. Moreover, postmortem brain tissues from patients with neurodegenerative diseases have been reported to increase the expression of markers of ROS damage and nucleic acid damage, including glutathione (GSH) and 8-hydroxy-2′-deoxyguanosine (8-OHdG) [26,68,69,70]. Therefore, oxidative stress has been studied in various neurodegenerative diseases in relation to the degeneration of neurons, microglia, astrocytes, and the blood–brain barrier [71].
ALS is the most common adult-onset lethal motor neuron disease, characterized by progressive muscle weakness and atrophy, leading to respiratory failure and death within a few years following disease onset [12,13]. The analysis of postmortem neuronal tissue, CSF, serum, and urine from patients with sporadic or familial ALS harboring SOD-1 mutations showed redox system upregulation, suggesting that oxidative stress contributes to the pathogenesis of both sporadic and familial ALS [72,73,74,75]. In addition, several previously published studies have demonstrated that markers of oxidative damage to proteins, DNA, and RNA are increased in both sporadic and familial ALS patients [68,69,70]. Nonetheless, because the expression levels of various antioxidant enzymes and parameters of mitochondrial function differ between sporadic ALS and familial ALS, it is suggested that distinct mechanisms of oxidative stress are likely to act between sporadic ALS and familial ALS [76]. As the disease progresses, oxidative stress might be increased by nutritional deficiency, cachexia, psychological stress, and impending respiratory failure [10]. In addition, a correlation between oxidative stress measured by positron emission tomography of the brain with 62Cu diacetyl-bis (N4-methylthiosemicarbazone) and clinical severity in ALS was recently reported [77]. Oxidative stress might be both a cause and consequence of the disease, and is associated with pathogenesis as well as disease progression.

3.1. Abnormal Protein Aggregations and Oxidative Stress in ALS

Aberrant protein aggregation, including SOD-1 and TDP-43, is the main pathological hallmark of familial and sporadic ALS. Aberrant protein aggregation is induced by oxidative stress, and conversely, oxidative stress is induced by aberrant protein aggregation. Oxidative modifications to proteins have been considered as one of the mechanisms of motor neuron death in both sporadic and familial ALS.
SOD-1 is localized in the cytosol, nucleus, peroxisomes, and mitochondria. As SOD-1 plays an important role in the antioxidant defense of the cell by switching from O2•− to O2 and H2O2 [78], the absence of SOD-1 causes an increase in oxidative stress. In ALS patients, more than 170 mutations have been identified, and aberrant aggregation of SOD-1 exists in spinal motor neurons from patients with sporadic and familial ALS [79,80]. Moreover, the misfolded form of SOD-1 exists in the CSF of patients with sporadic ALS [81]. The deleterious effects of a toxic gain of function due to mutant SOD-1 and aberrant aggregation of SOD-1 have been considered as one of the pathogeneses of motor neuron death in ALS [82]. SOD-1 could become a target for oxidative stress, leading to aberrant aggregation [83], and an aberrant aggregation of SOD-1 contributes to ubiquitin/proteasome system dysfunction and interferes with mitophagy processes, which are the clearance of damaged mitochondria, through optineurin sequestration [84,85]. In addition, mutated SOD-1 could lead to oxidative damage by promoting reverse O2•− production through the dissociation of zinc from SOD-1, hyperactivation of NOX-dependent O2•− production, and decrease in the expression levels of NRF2 [86,87,88,89]. In addition, the presence of mutated SOD-1 could lead to impairment of the respiratory chain and mitochondrial redox homeostasis [90]. Studies of the antioxidant enzyme activity of SOD-1 have reported inconsistent results; one study showed that the activities in pyramidal cells of the cerebral cortex and in spinal motor neurons from sporadic ALS patients were reduced [91], whereas another study showed that the activity in spinal motor neurons from ALS patients did not change [92].
The aberrant aggregation of TDP-43, DNA- and RNA-binding protein encoded by TARDBP is the main pathological hallmark of many forms of familial and sporadic ALS and is used to determine the severity and stage of disease progression [93,94]. The intrinsic aggregation prone to TDP-43 has been attributed to the C-terminal glycine-rich domain, and activated caspases cleave TDP-43 to generate C-terminal fragments, which are particularly prone to aggregate formation [95,96]. Oxidative stress resulting in mitochondrial dysfunction due to activated caspases has been shown to induce TDP-43 aggregation [97]. In addition, oxidative stress causes TDP-43 mislocalization and increases its tendency to aggregate by promoting the acetylation of TDP-43 [98]. TDP-43 aggregation sequesters some miRNAs and proteins, resulting in changes in gene expression, which causes a global mitochondrial imbalance that increases oxidative stress [99]. Stress granules (SGs), which represent membrane-less organelles composed of RNAs and proteins, are transiently formed under stress conditions, including oxidative stress, to reprogram RNA translation [100]. Oxidized TDP-43 recruited into SGs has been observed in ALS [101,102], and ALS-linked mutant TDP-43 is incorporated into SGs earlier than wild-type TDP-43, and these mutants form significantly larger stress granules [103]. Moreover, the ALS-linked mutant TDP-43 influenced the NRF2 antioxidative pathway, which is a major regulator of antioxidant elements, through interactions with the family of heterogeneous nuclear ribonucleoproteins (hnRNPs) [104].

3.2. Oxidative DNA Damage and Impairment of DNA Repair in ALS

Oxidative DNA damage accumulates in the nuclear DNA and mitochondrial DNA of aging neurons and has been suspected to play an important role in the pathogenesis of both sporadic ALS and familial ALS [69]. Since guanine has a lower electron reduction potential than the other DNA bases, 8-OHdG is the most abundant oxidative alteration in DNA among more than 20 oxidation products. Additionally, 8-OHdG has been employed as a marker of oxidative damage to DNA [105,106]. The levels of 8-OHdG in both nuclear DNA and mitochondrial DNA of brain tissue increased with aging [107], and increased levels of 8-OHdG have been identified in the nuclear DNA of the motor cortex derived from sporadic ALS patients and the spinal cord derived from both sporadic ALS and familial ALS harboring SOD-1 mutations [68,108,109]. In addition, increased levels of 8-OHdG have been observed in nuclear DNA from the frontal cortex and spinal motor neurons and in mitochondrial DNA from spinal motor neurons of transgenic mice harboring SOD-1 mutations, but not from the cerebellum [110,111]. In addition, in spinal motor neurons of sporadic ALS patients, colocalization of 8-OHdG and activated p53 is recognized [109], and in motor neurons of the SOD-1 mutant mouse, which has been proposed as a model mouse of familial ALS harboring SOD-1 mutation, somal and mitochondrial swelling, formation of DNA strand breaks, and activation of p53, which triggers apoptosis as a result of the accumulation of DNA damage, was observed [112]. Similarly, higher levels of oxidative DNA damage and increased levels of apoptosis and p53 activity were observed in SH-SY5Y cells overexpressing the mutant SOD-1 protein, fused in sarcoma (FUS) knockdown SH-SY5Y cells, induced pluripotent stem cells (iPSCs) derived from C9ORF72-ALS patients [113,114,115]. Therefore, oxidative DNA damage is thought to be involved in motor neuron death in both sporadic and familial ALS.
The accumulation of abnormal DNA due to impairment of DNA damage repair was hypothesized approximately 40 years ago [116]. Apurinic/apyrimidinic endonuclease 1 (APE1) is a transcription factor involved in redox regulation and an enzyme involved in DNA repair, especially in BER [117], and negatively regulates NRF2 [118]. The levels of APE1 in the motor cortex and spinal cords derived from sporadic ALS were increased and APE1 localization was altered compared with those in control patients [119]. Furthermore, cytosolic mislocalization of APE1 is suggested to trigger oxidative DNA damage in spinal motor neurons of SOD-1 mutant mice [120]. Moreover, epigenetic mechanisms involved in gene expression are impaired by mislocalization of TDP-43, and the hypomethylation of the promoter of APE1 has been recognized in C9ORF72-ALS patients [109,121]. In contrast, other studies demonstrated that the activity of APE1 was lower in patients with sporadic ALS, and the loss of immunoreactivity for APE1 was seen in spinal motor neurons of SOD-1 mutant mice [122,123]. The expression levels and activities of another DNA repair element, 8-oxoguanine glycosylase (OGG1), which removes oxidized guanine from DNA and poly (ADP-robose) polymerase-1 (PARP-1), which is activated by oxidative DNA damage, have been investigated. The impairment of mitochondrial OGG1 activity in spinal motor neurons of sporadic ALS [124], increased expression levels of nuclear Ogg1 in the spinal motor neurons of mutant SOD-1 mice [123], and increased expression levels of PARP-1 in astrocytes and the motor cortex—but not in spinal motor neurons of sporadic ALS—have been recognized [125]. Moreover, impairment of DNA damage repair is recognized in spinal motor neurons of C9ORF72-ALS patients and is suspected to be associated with impairment of interaction between FUS and histone deacetylase 1 (HDAC1) and the loss of nuclear TDP-43 and SOD-1 [53,126,127].
These lines of evidence suggest that oxidative DNA damage and impairment of DNA repair are involved in the pathogenesis of sporadic ALS and familial ALS.

3.3. RNA Modifications Associated with Oxidative Stress in ALS

Oxidative stress and RNA metabolism are considered to be the pathogenesis of ALS, and there is increasing evidence of the relationship between these two aspects. Oxidative stress causes abnormalities in RNA metabolism, and conversely, abnormalities in RNA metabolism cause oxidative stress [128]. Indeed, increased oxidative RNA modification and alteration of splicing and expression of mRNA are recognized in the spinal motor neurons of SOD-1 mutant mice [129,130]. Moreover, oxidative RNA modification is an early event in the course of the disease and precedes the death of motor neurons [62,129].
Among the ALS-related genes, RNA-binding proteins encoded by TARDBP, FUS, and SOD-1 are implicated in miRNA processing [131], and some miRNAs regulate the expression levels of genes related to oxidative stress [65]. In ALS patients, miRNAs are involved in oxidative stress, which is involved in the pathogenesis of ALS.
MiR-27a, which plays a role in muscle proliferation, and miR-142-5p, which functions in inflammation and cell apoptosis, regulates oxidative stress via inhibition of NRF2 expression directly [132,133], and the expression levels of miR-27a were increased in the skeletal muscle of sporadic ALS [134]. MiR-338-3p, a brain-specific miRNA, is involved in mitochondrial function by regulating the expression of multiple nuclear-encoded mitochondrial mRNAs [135]. Indeed, the expression levels of miR-338-5p were increased in neuromuscular junctions of sporadic ALS and in the spinal cord and motor cortex of SOD-1 mutant mice [136,137]. Moreover, miR-338-3p modulates apoptosis-associated tyrosine kinase mRNA levels in neurons, resulting in the apoptosis of mature neurons and neurodegeneration of oligodendrocytes. In addition, miR-338-3p suppresses the expression levels of SLC1A2 mRNA, suggesting a role in glutamate clearance, which is suspected to be one of the pathogenesis of ALS [24,138]. MiR-34a, which is involved in cell cycle regulation, also regulates oxidative stress via direct inhibition of NRF2 expression [139]. Moreover, miR-34a is involved in oxidative stress-induced apoptosis due to the regulation of tumor protein 53 (TP53) and sirtuin 1 (SIRT1) expression, and the expression levels of miR-34a are decreased in iPSCs derived from sporadic ALS patients and in the spinal cord and brain stem of SOD1 mutant mice [140,141]. MiR-155 is involved in inflammatory response and mitochondrial function. The expression levels of miR-155 were increased in skeletal muscles of ALS patients and in the spinal cord of SOD-1 mutant mice and both sporadic and familial ALS patients [142,143,144]. These changes were recognized during disease progression and in early-stage and end-stage in SOD-1 mutant mice and ALS patients [142,145,146].
Intriguingly, enoxacin, a fluoroquinolone antibiotic, rescued the expression of miR-34a, and inhibition of miR-155 in SOD-1 mutant mice has been shown to prolong survival [141,143,144]. Therefore, a treatment strategy for miRNA dysregulation may be useful for ALS.
Although circRNAs have been demonstrated to modulate redox homeostasis, circRNAs related to both oxidative stress and the pathogenesis of ALS have not been identified.

3.4. Treatment Linked to Oxidative Stress in ALS

The evidence that oxidative biomolecular modification is involved in the pathogenesis of ALS is increasing; therefore, it is necessary to recognize redox dysregulation as a therapeutic target.
Edaravone, a free radical scavenger that eliminates lipid peroxides and hydroxyl radicals, is the only approved antioxidant drug for ALS patients [17]. Oxidative stress has been implicated in the pathogenesis of ALS, and edaravone has been shown to inhibit motor neuron death and abnormal SOD1 aggregation in G93A mutant SOD1 transgenic mice, suggesting that edaravone might ameliorate disease progression [147]. Moreover, edaravone, but not other antioxidants, has been demonstrated to have other neuroprotective functions, including inhibition of Bcl-2 apoptotic pathways, augmentation of nitric oxide release from vascular cells and platelets, and suppression of ER stress, leading edaravone to be the most promising therapeutic agent for targeting redox dysregulation in patients with [148,149,150]. In a phase three clinical study and meta-analysis, edaravone showed a significantly smaller decline in the revised ALS functional rating scale (ALSFRS-R) score compared with a placebo, without significant adverse effects in the early stage of ALS patients [17,151]. Furthermore, long-term (18 months) administration of edaravone improved the ALSFRS-R score and survival rate, and therapeutic effects in patients outside Japan have been reported [152,153]. Additionally, reduced concentrations of the oxidative damage marker, 3-nitrotyrosine (3-NT), and improved antioxidative activity after intravenous administration of edaravone have been demonstrated [154,155]. Therefore, although some controversial studies on the therapeutic effects of edaravone in ALS patients have been reported [156,157], edaravone hardly has harmful adverse effects and may inhibit disease progression in the early stages of ALS.
In addition, other multiple antioxidants and combinations of apoptotic inhibitors, anticancer agents, and antioxidants have been identified as potential therapeutic agents. Vitamins, including vitamin E, vitamin C, and coenzyme Q10 (CoQ10) have been suspected to be therapeutic agents because of their ability to regulate antioxidant systems, and clinical trials using these compounds have been conducted [158,159,160]. Additionally, to gain further effects by adding apoptotic inhibitors or anticancer agents, clinical studies using a combination of vitamins, CoQ10, anti-inflammatory agents, antiapoptotic agents, or anticancer agents have been conducted [161,162,163]. EH301, a combination of two antioxidants, pterostilbene and nicotinamide riboside, has been shown to suppress disease progression and improve ALSFRS-R and forced vital capacity in a small number of ALS patients [163]. However, other clinical trials have failed to show improvement in symptoms and/or suppression of disease progression, suggesting that these therapeutic strategies using monotherapy and/or combination therapy of antioxidants are not effective for ALS patients.
Although redox dysregulation has been recognized as a pivotal therapeutic target, the clinical effects of edaravone in ALS patients are limited, and other antioxidant therapies have mostly been unsuccessful. Therefore, the development of more successful therapies is expected [70]. As preclinical studies of multiple therapeutic targets have used mutant SOD-1 mice, which do not exhibit TDP-43 pathology, which is a pathological hallmark of ALS, the results of the preclinical studies may not enhance survival and improve motor function in ALS patients [16]. Therefore, to develop more successful therapies, using cells and animals representing molecular abnormalities that occur in ALS patients in a preclinical study is important.

4. Promising Biomarker Candidates Associated with Oxidative Stress in ALS

Oxidative stress has been considered to be involved in the onset and disease progression of ALS, and the evidence of oxidative biomolecular modifications and changes in molecular expression levels in neuronal tissues of ALS patients have been increasing, suggesting that the detection of these alterations would be a promising biomarker. Therefore, oxidative stress markers derived from body fluids have been investigated in many studies [164]. In this section, we review promising biomarker candidates linked to oxidative stress in proteins and RNAs (Table 1).

4.1. Protein

Oxidative stress results from the increased generation of oxidants and/or decrease in antioxidants. Therefore, changes in the expression levels of oxidants and antioxidants have been investigated.
With regard to oxidants, the expression levels of oxidative stress products such as 8-OHdG and malone dialdehyde (MDA) and antioxidant system products such as SOD-1 and GPX have been reported to be altered in body fluids in ALS patients compared with those in controls; therefore, this change has been suspected to be a promising biomarker candidate for ALS. As the levels of 8-OHdG in plasma and CSF increased with aging, 8-OHdG has been well studied to determine whether it will come to be an oxidative stress biomarker [69]. Several studies have shown that 8-OHdG levels in CSF, plasma, and urine were elevated in both sporadic and familial ALS patients compared with disease-free control patients and patients with other neurological diseases [69,74,165,166]. Moreover, 8-OHdG levels in urine were negatively correlated with both the rate of change of the ALSFRS-R and forced vital capacity (FVC) [69]. Lipid peroxidation is one of several outcomes of oxidative stress. MDA is a potential oxidative stress biomarker that can be used to evaluate lipid peroxidation. The concentration of MDA in plasma was significantly increased in sporadic ALS patients compared with that in control patients [166,167,168]. In addition, 4-hydroxynonenal (HNE) is one of the most toxic products of lipid peroxidation, and its levels are significantly increased in plasma and CSF from sporadic ALS patients. Moreover, the levels of HNE in plasma positively correlate with the extent of disease [75]. 3-nitrotyrosine (3NT), formed by the nitration of protein-bound and free tyrosine residues, is also a promising biomarker of oxidative stress. Indeed, the levels of 3NT were increased in the spinal cord of familial and sporadic ALS patients [72]. Moreover, the levels of 3NT have been shown to be increased in the CSF of patients with sporadic ALS [169]. However, it has been reported that the levels of 3NT in CSF did not change significantly between patients with sporadic ALS and controls [170].
Levels of antioxidants and antioxidant enzymes have also been considered as biomarker candidates for ALS. However, these results are inconsistent.
Regarding the antioxidant enzyme activity of SOD, several studies have shown that SOD activity is elevated in red blood cells (RBC) and plasma from sporadic ALS patients [167,171,172], whereas it has been shown that the activity is not changed significantly in RBC and plasma from sporadic ALS patients and reduced in RBC and CSF from sporadic and familial ALS patients [165,166,168,173,174,175,176,177]. Intriguingly, it has been reported that the antioxidant enzyme activity of SOD in bulbar onset sporadic ALS was significantly higher than in spinal onset sporadic ALS [167]. Deficiency of the antioxidant activity of GPX indicates cytotoxicity due to the increase in hydrogen peroxide and membrane lipid peroxidation [172]. This said, the antioxidant enzyme activity of GPX has also been shown to be inconsistent. Some studies have shown that the activity is reduced in whole blood, RBCs, and plasma from sporadic ALS patients [167,173,178], whereas other studies have shown that the activity is not changed in plasma from ALS patients [166,168,172]. Several studies have shown that the concentration of GSH in whole blood and plasma was further reduced in sporadic ALS patients than in controls [166,179], whereas some studies have shown that the concentration in plasma was not significantly changed in ALS patients [168,180]. Moreover, in patients with sporadic ALS, no significant temporal change during a 6-month period was observed [179]. In addition, the concentrations of glutathione disulfide (GSSG) in whole blood were significantly increased in patients with sporadic ALS compared with controls [166]. CoQ10 is an antioxidant and an enzyme cofactor in the mitochondrial electron transport chain. Levels of CoQ10 were not significantly increased in plasma and serum in sporadic ALS patients compared with those in control patients, and these levels were not influenced by clinical form, age at onset, or duration of disease [181,182,183]. However, the proportion of oxidized forms of CoQ10 was significantly higher in CSF and plasma derived from ALS patients compared with control patients and correlated with disease duration [184,185]. Uric acid (UA) is an important natural antioxidant that reduces oxidative damage to cellular components by scavenging free radicals [186]. The levels of UA and creatinine (Cr) in serum have been shown to be significantly decreased in patients with ALS than in controls [182,186,187,188,189,190,191]. Intriguingly, the levels of UA in bulbar-onset sporadic ALS patients were significantly decreased compared with those in spinal onset sporadic ALS patients, and those in female sporadic ALS patients were decreased compared with those in male sporadic ALS patients [189,191]. Moreover, the baseline levels of Cr and UA in serum were negatively correlated with the annual decline of ALSFRS-R, and the annual decline level of Cr in serum was positively correlated with the annual decline of ALSFRS-R [186,187,188,190]. However, some studies have reported that the levels of UA in plasma were not significantly changed, and no significant correlation between the levels of UA and ALSFRS-R was seen in sporadic ALS patients [185,189].

4.2. RNA

RNAs, including noncoding RNA and miRNA, have been shown to be redox sensitive and contribute to redox signaling, indicating that RNAs could be biomarker candidates related to oxidative stress [70,192]. Some miRNAs involved in oxidative stress have been suspected to be promising biomarkers of ALS [193]. The expression levels of miR-27a, which regulates NRF2 expression, were decreased in serum and serum exosomes, and downregulation of miR-27a was correlated with the degree of muscle atrophy [194,195]. In addition, the expression levels of miR-142-5p, which regulate NRF2 expression, are decreased in the CSF of patients with sporadic ALS, and the expression levels of miR-155, which is implicated in mitochondrial dysfunction and neuroinflammation, are increased in peripheral monocytes from sporadic ALS [145,196]. Moreover, the expression levels of miR-338-3p, which regulates expression of subunits of mitochondrial oxidative phosphorylation complexes, were increased in the serum, PBL, and CSF of patients with sporadic ALS patients [138,197].

5. Conclusions

In this review, we summarize currently established knowledge regarding oxidative biomolecular modifications and promising biomarker candidates linked to oxidative stress in ALS. Therapies targeting the elimination of oxidative stress, including edaravone administration, are developing and extensive evidence of an association between redox dysfunction and ALS is increasing, suggesting that oxidative stress is a pivotal therapeutic and biomarker target. However, the precise mechanisms underlying the relationship between oxidative stress and ALS has not been elucidated. If the present situation is to persist, it would be difficult to establish a therapy and biomarker linked to oxidative biomolecular modifications, and further investigations are required. If the underlying mechanisms between oxidative stress and ALS are clarified, oxidative biomolecular modifications would be not only be available as therapeutic targets, but also as diagnostic, predictive, and/or pharmacodynamic biomarkers.

Author Contributions

H.T. and A.T. supervised the project. All authors wrote the main text, produced the figures, discussed the results and reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by JSPS KAKENHI Grant Number 19K23957 and The Uehara Memorial Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Harumi Tobita at the University of Tsukuba and clinical laboratory at the Ibaraki Western Medical Center for technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  2. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  3. Carrera-Julia, S.; Moreno, M.L.; Barrios, C.; de la Rubia Orti, J.E.; Drehmer, E. Antioxidant Alternatives in the Treatment of Amyotrophic Lateral Sclerosis: A Comprehensive Review. Front. Physiol. 2020, 11, 63. [Google Scholar] [CrossRef] [Green Version]
  4. Sies, H. Role of metabolic H2O2 generation: Redox signaling and oxidative stress. J. Biol Chem. 2014, 289, 8735–8741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef] [Green Version]
  6. Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef]
  7. Tan, B.L.; Norhaizan, M.E.; Liew, W.P.; Sulaiman Rahman, H. Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Sies, H.; Cadenas, E. Oxidative stress: Damage to intact cells and organs. Philos. Trans. R. Soc. B Biol. Sci. 1985, 311, 617–631. [Google Scholar] [CrossRef]
  9. Castelli, V.; Benedetti, E.; Antonosante, A.; Catanesi, M.; Pitari, G.; Ippoliti, R.; Cimini, A.; d’Angelo, M. Neuronal Cells Rearrangement During Aging and Neurodegenerative Disease: Metabolism, Oxidative Stress and Organelles Dynamic. Front. Mol. Neurosci. 2019, 12, 132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. D’Amico, E.; Factor-Litvak, P.; Santella, R.M.; Mitsumoto, H. Clinical perspective on oxidative stress in sporadic amyotrophic lateral sclerosis. Free Radic. Biol. Med. 2013, 65, 509–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [Green Version]
  12. Brown, R.H.; Al-Chalabi, A. Amyotrophic Lateral Sclerosis. N. Engl. J. Med. 2017, 377, 162–172. [Google Scholar] [CrossRef] [Green Version]
  13. Chio, A.; Logroscino, G.; Hardiman, O.; Swingler, R.; Mitchell, D.; Beghi, E.; Traynor, B.G.; Eurals, C. Prognostic factors in ALS: A critical review. Amyotroph. Lateral Scler. 2009, 10, 310–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. 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. Mutations in Cu/Zn Superoxide Dismutase Gene Are Associated With Familial Amyotrophic Lateral Sclerosis. Nature 1993, 362, 59–62. [Google Scholar] [CrossRef]
  15. Taylor, J.P.; Brown, R.H., Jr.; Cleveland, D.W. Decoding ALS: From genes to mechanism. Nature 2016, 539, 197–206. [Google Scholar] [CrossRef] [Green Version]
  16. Jagaraj, C.J.; Parakh, S.; Atkin, J.D. Emerging Evidence Highlighting the Importance of Redox Dysregulation in the Pathogenesis of Amyotrophic Lateral Sclerosis (ALS). Front. Cell. Neurosci. 2020, 14, 581950. [Google Scholar] [CrossRef]
  17. Abe, K.; Aoki, M.; Tsuji, S.; Itoyama, Y.; Sobue, G.; Togo, M.; Hamada, C.; Tanaka, M.; Akimoto, M.; Nakamura, K.; et al. Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: A randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2017, 16, 505–512. [Google Scholar] [CrossRef]
  18. Brooks, B.R.; Miller, R.G.; Swash, M.; Munsat, T.L. El Escorial revisited: Revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Mot. Neuron Disord. 2000, 1, 293–299. [Google Scholar] [CrossRef] [PubMed]
  19. de Carvalho, M.; Dengler, R.; Eisen, A.; England, J.D.; Kaji, R.; Kimura, J.; Mills, K.; Mitsumoto, H.; Nodera, H.; Shefner, J.; et al. Electrodiagnostic criteria for diagnosis of ALS. Clin. Neurophysiol. 2008, 119, 497–503. [Google Scholar] [CrossRef] [PubMed]
  20. Biomarkers Definitions Working, G. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001, 69, 89–95. [Google Scholar] [CrossRef]
  21. Rappa, G.; Puglisi, C.; Santos, M.F.; Forte, S.; Memeo, L.; Lorico, A. Extracellular Vesicles from Thyroid Carcinoma: The New Frontier of Liquid Biopsy. Int. J. Mol. Sci. 2019, 20, 1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Candelario, K.M.; Steindler, D.A. The role of extracellular vesicles in the progression of neurodegenerative disease and cancer. Trends Mol. Med. 2014, 20, 368–374. [Google Scholar] [CrossRef] [Green Version]
  23. Caruso Bavisotto, C.; Scalia, F.; Marino Gammazza, A.; Carlisi, D.; Bucchieri, F.; Conway de Macario, E.; Macario, A.J.L.; Cappello, F.; Campanella, C. Extracellular Vesicle-Mediated Cell-Cell Communication in the Nervous System: Focus on Neurological Diseases. Int. J. Mol. Sci. 2019, 20, 434. [Google Scholar] [CrossRef] [Green Version]
  24. Hosaka, T.; Yamashita, T.; Tamaoka, A.; Kwak, S. Extracellular RNAs as Biomarkers of Sporadic Amyotrophic Lateral Sclerosis and other Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 3148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tarafdar, A.; Pula, G. The Role of NADPH Oxidases and Oxidative Stress in Neurodegenerative Disorders. Int J. Mol. Sci 2018, 19, 3814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Andersen, J.K. Oxidative stress in neurodegeneration: Cause or consequence? Nat. Med. 2004, 10, S18–S25. [Google Scholar] [CrossRef]
  27. Quinlan, S.; Kenny, A.; Medina, M.; Engel, T.; Jimenez-Mateos, E.M. MicroRNAs in Neurodegenerative Diseases. Int. Rev. Cell Mol. Biol. 2017, 334, 309–343. [Google Scholar] [CrossRef] [PubMed]
  28. Maniati, M.S.; Maniati, M.; Yousefi, T.; Ahmadi-Ahangar, A.; Tehrani, S.S. New insights into the role of microRNAs and long noncoding RNAs in most common neurodegenerative diseases. J. Cell. Biochem 2019. [Google Scholar] [CrossRef] [PubMed]
  29. Quek, C.; Hill, A.F. The role of extracellular vesicles in neurodegenerative diseases. Biochem. Biophys. Res. Commun 2017, 483, 1178–1186. [Google Scholar] [CrossRef]
  30. Davies, M.J. Protein oxidation and peroxidation. Biochem. J. 2016, 473, 805–825. [Google Scholar] [CrossRef] [Green Version]
  31. Hawkins, C.L.; Davies, M.J. Detection, identification, and quantification of oxidative protein modifications. J. Biol. Chem. 2019, 294, 19683–19708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sitia, R.; Molteni, S.N. Stress, protein (mis)folding, and signaling: The redox connection. Sci. STKE 2004, 2004, pe27. [Google Scholar] [CrossRef] [PubMed]
  33. Gianazza, E.; Crawford, J.; Miller, I. Detecting oxidative post-translational modifications in proteins. Amino Acids 2007, 33, 51–56. [Google Scholar] [CrossRef] [PubMed]
  34. Weids, A.J.; Ibstedt, S.; Tamas, M.J.; Grant, C.M. Distinct stress conditions result in aggregation of proteins with similar properties. Sci. Rep. 2016, 6, 24554. [Google Scholar] [CrossRef] [Green Version]
  35. Serebryany, E.; Woodard, J.C.; Adkar, B.V.; Shabab, M.; King, J.A.; Shakhnovich, E.I. An Internal Disulfide Locks a Misfolded Aggregation-prone Intermediate in Cataract-linked Mutants of Human γD-Crystallin. J. Biol. Chem. 2016, 291, 19172–19183. [Google Scholar] [CrossRef] [Green Version]
  36. Oka, O.B.; Bulleid, N.J. Forming disulfides in the endoplasmic reticulum. Biochim. Biophys. Acta 2013, 1833, 2425–2429. [Google Scholar] [CrossRef] [Green Version]
  37. Karri, S.; Singh, S.; Paripati, A.K.; Marada, A.; Krishnamoorthy, T.; Guruprasad, L.; Balasubramanian, D.; Sepuri, N.B.V. Adaptation of Mge1 to oxidative stress by local unfolding and altered Interaction with mitochondrial Hsp70 and Mxr2. Mitochondrion 2019, 46, 140–148. [Google Scholar] [CrossRef]
  38. Hwang, C.; Sinskey, A.J.; Lodish, H.F. Oxidized redox state of glutathione in the endoplasmic reticulum. Science 1992, 257, 1496–1502. [Google Scholar] [CrossRef]
  39. Kodiha, M.; Chu, A.; Matusiewicz, N.; Stochaj, U. Multiple mechanisms promote the inhibition of classical nuclear import upon exposure to severe oxidative stress. Cell Death Differ. 2004, 11, 862–874. [Google Scholar] [CrossRef]
  40. Yun, H.R.; Jo, Y.H.; Kim, J.; Shin, Y.; Kim, S.S.; Choi, T.G. Roles of Autophagy in Oxidative Stress. Int. J. Mol. Sci. 2020, 21, 3289. [Google Scholar] [CrossRef]
  41. Sanchez de Groot, N.; Gomes, R.A.; Villar-Pique, A.; Babu, M.M.; Coelho, A.V.; Ventura, S. Proteome response at the edge of protein aggregation. Open Biol. 2015, 5, 140221. [Google Scholar] [CrossRef] [Green Version]
  42. Lindahl, T.; Barnes, D.E. Repair of endogenous DNA damage. Cold Spring Harb. Symp. Quant. Biol. 2000, 65, 127–133. [Google Scholar] [CrossRef]
  43. Walker, C.; El-Khamisy, S.F. Perturbed autophagy and DNA repair converge to promote neurodegeneration in amyotrophic lateral sclerosis and dementia. Brain 2018, 141, 1247–1262. [Google Scholar] [CrossRef]
  44. Cooke, M.S.; Evans, M.D.; Dizdaroglu, M.; Lunec, J. Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J. 2003, 17, 1195–1214. [Google Scholar] [CrossRef] [Green Version]
  45. Radak, Z.; Zhao, Z.; Goto, S.; Koltai, E. Age-associated neurodegeneration and oxidative damage to lipids, proteins and DNA. Mol. Asp. Med. 2011, 32, 305–315. [Google Scholar] [CrossRef]
  46. Martin, L.J. DNA damage and repair: Relevance to mechanisms of neurodegeneration. J. Neuropathol. Exp. Neurol. 2008, 67, 377–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Obrador, E.; Salvador-Palmer, R.; Lopez-Blanch, R.; Jihad-Jebbar, A.; Valles, S.L.; Estrela, J.M. The Link between Oxidative Stress, Redox Status, Bioenergetics and Mitochondria in the Pathophysiology of ALS. Int. J. Mol. Sci. 2021, 22, 6352. [Google Scholar] [CrossRef] [PubMed]
  48. Borthwick, G.M.; Johnson, M.A.; Ince, P.G.; Shaw, P.J.; Turnbull, D.M. Mitochondrial enzyme activity in amyotrophic lateral sclerosis: Implications for the role of mitochondria in neuronal cell death. Ann. Neurol. 1999, 46, 787–790. [Google Scholar] [CrossRef]
  49. Coppede, F. An overview of DNA repair in amyotrophic lateral sclerosis. Sci. World J. 2011, 11, 1679–1691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Gredilla, R.; Weissman, L.; Yang, J.L.; Bohr, V.A.; Stevnsner, T. Mitochondrial base excision repair in mouse synaptosomes during normal aging and in a model of Alzheimer’s disease. Neurobiol. Aging 2012, 33, 694–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Lin, M.T.; Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 2006, 443, 787–795. [Google Scholar] [CrossRef]
  52. Wilson, D.M., III; Bohr, V.A. The mechanics of base excision repair, and its relationship to aging and disease. DNA Repair 2007, 6, 544–559. [Google Scholar] [CrossRef]
  53. Mitra, J.; Guerrero, E.N.; Hegde, P.M.; Liachko, N.F.; Wang, H.; Vasquez, V.; Gao, J.; Pandey, A.; Taylor, J.P.; Kraemer, B.C.; et al. Motor neuron disease-associated loss of nuclear TDP-43 is linked to DNA double-strand break repair defects. Proc. Natl. Acad. Sci. USA 2019, 116, 4696–4705. [Google Scholar] [CrossRef] [Green Version]
  54. Liu, Z.; Chen, X.; Li, Z.; Ye, W.; Ding, H.; Li, P.; Aung, L.H.H. Role of RNA Oxidation in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 5022. [Google Scholar] [CrossRef]
  55. Nunomura, A.; Hofer, T.; Moreira, P.I.; Castellani, R.J.; Smith, M.A.; Perry, G. RNA oxidation in Alzheimer disease and related neurodegenerative disorders. Acta Neuropathol. 2009, 118, 151–166. [Google Scholar] [CrossRef]
  56. Simms, C.L.; Zaher, H.S. Quality control of chemically damaged RNA. Cell. Mol. Life Sci. 2016, 73, 3639–3653. [Google Scholar] [CrossRef] [Green Version]
  57. Jacobs, A.C.; Resendiz, M.J.; Greenberg, M.M. Direct strand scission from a nucleobase radical in RNA. J. Am. Chem. Soc. 2010, 132, 3668–3669. [Google Scholar] [CrossRef] [Green Version]
  58. Tanaka, M.; Chock, P.B.; Stadtman, E.R. Oxidized messenger RNA induces translation errors. Proc. Natl. Acad. Sci. USA 2007, 104, 66–71. [Google Scholar] [CrossRef] [Green Version]
  59. Ding, Q.; Dimayuga, E.; Keller, J.N. Oxidative stress alters neuronal RNA- and protein-synthesis: Implications for neural viability. Free Radic. Res. 2007, 41, 903–910. [Google Scholar] [CrossRef] [PubMed]
  60. Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef]
  61. Shan, X.; Tashiro, H.; Lin, C.L. The identification and characterization of oxidized RNAs in Alzheimer’s disease. J. Neurosci. 2003, 23, 4913–4921. [Google Scholar] [CrossRef] [PubMed]
  62. Shan, X.; Chang, Y.; Lin, C.L. Messenger RNA oxidation is an early event preceding cell death and causes reduced protein expression. FASEB J. 2007, 21, 2753–2764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Engedal, N.; Zerovnik, E.; Rudov, A.; Galli, F.; Olivieri, F.; Procopio, A.D.; Rippo, M.R.; Monsurro, V.; Betti, M.; Albertini, M.C. From Oxidative Stress Damage to Pathways, Networks, and Autophagy via MicroRNAs. Oxid. Med. Cell. Longev. 2018, 2018, 4968321. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, J.X.; Gao, J.; Ding, S.L.; Wang, K.; Jiao, J.Q.; Wang, Y.; Sun, T.; Zhou, L.Y.; Long, B.; Zhang, X.J.; et al. Oxidative Modification of miR-184 Enables It to Target Bcl-xL and Bcl-w. Mol. Cell 2015, 59, 50–61. [Google Scholar] [CrossRef] [Green Version]
  65. Paladino, S.; Conte, A.; Caggiano, R.; Pierantoni, G.M.; Faraonio, R. Nrf2 Pathway in Age-Related Neurological Disorders: Insights into MicroRNAs. Cell. Physiol. Biochem. 2018, 47, 1951–1976. [Google Scholar] [CrossRef]
  66. Li, L.; Ni, Z.; Si, X.; Jiang, L.; Sang, H.; Xia, W.; Chen, Z.; Huang, J.; Jin, J.; Shao, A.; et al. Emerging Clues of Regulatory Roles of Circular RNAs through Modulating Oxidative Stress: Focus on Neurological and Vascular Diseases. Oxid. Med. Cell. Longev. 2021, 2021, 6659908. [Google Scholar] [CrossRef]
  67. Nayernia, Z.; Jaquet, V.; Krause, K.H. New insights on NOX enzymes in the central nervous system. Antioxid. Redox Signal. 2014, 20, 2815–2837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Ferrante, R.J.; Browne, S.E.; Shinobu, L.A.; Bowling, A.C.; Baik, M.J.; MacGarvey, U.; Kowall, N.W.; Brown, R.H., Jr.; Beal, M.F. Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J. Neurochem. 1997, 69, 2064–2074. [Google Scholar] [CrossRef]
  69. Bogdanov, M.; Brown, R.H.; Matson, W.; Smart, R.; Hayden, D.; O’Donnell, H.; Flint Beal, M.; Cudkowicz, M. Increased oxidative damage to DNA in ALS patients. Free Radic. Biol. Med. 2000, 29, 652–658. [Google Scholar] [CrossRef]
  70. Konovalova, J.; Gerasymchuk, D.; Parkkinen, I.; Chmielarz, P.; Domanskyi, A. Interplay between MicroRNAs and Oxidative Stress in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 6055. [Google Scholar] [CrossRef] [Green Version]
  71. Singh, E.; Devasahayam, G. Neurodegeneration by oxidative stress: A review on prospective use of small molecules for neuroprotection. Mol. Biol. Rep. 2020, 47, 3133–3140. [Google Scholar] [CrossRef]
  72. Beal, M.F.; Ferrante, R.J.; Browne, S.E.; Matthews, R.T.; Kowall, N.W.; Brown, R.H., Jr. Increased 3-nitrotyrosine in Both Sporadic and Familial Amyotrophic Lateral Sclerosis. Ann. Neurol. 1997, 42, 644–654. [Google Scholar] [CrossRef]
  73. Kato, S.; Kato, M.; Abe, Y.; Matsumura, T.; Nishino, T.; Aoki, M.; Itoyama, Y.; Asayama, K.; Awaya, A.; Hirano, A.; et al. Redox system expression in the motor neurons in amyotrophic lateral sclerosis (ALS): Immunohistochemical studies on sporadic ALS, superoxide dismutase 1 (SOD1)-mutated familial ALS, and SOD1-mutated ALS animal models. Acta Neuropathol. 2005, 110, 101–112. [Google Scholar] [CrossRef]
  74. Mitsumoto, H.; Santella, R.M.; Liu, X.; Bogdanov, M.; Zipprich, J.; Wu, H.C.; Mahata, J.; Kilty, M.; Bednarz, K.; Bell, D.; et al. Oxidative stress biomarkers in sporadic ALS. Amyotroph. Lateral Scler. 2008, 9, 177–183. [Google Scholar] [CrossRef]
  75. Simpson, E.P.; Henry, Y.K.; Henkel, J.S.; Smith, R.G.; Appel, S.H. Increased lipid peroxidation in sera of ALS patients: A potential biomarker of disease burden. Neurology 2004, 62, 1758–1765. [Google Scholar] [CrossRef]
  76. Walczak, J.; Debska-Vielhaber, G.; Vielhaber, S.; Szymanski, J.; Charzynska, A.; Duszynski, J.; Szczepanowska, J. Distinction of sporadic and familial forms of ALS based on mitochondrial characteristics. FASEB J. 2019, 33, 4388–4403. [Google Scholar] [CrossRef]
  77. Ikawa, M.; Okazawa, H.; Tsujikawa, T.; Matsunaga, A.; Yamamura, O.; Mori, T.; Hamano, T.; Kiyono, Y.; Nakamoto, Y.; Yoneda, M. Increased oxidative stress is related to disease severity in the ALS motor cortex. A PET study. Neurology 2015, 84, 2033–2039. [Google Scholar] [CrossRef]
  78. Reddi, A.R.; Culotta, V.C. SOD1 integrates signals from oxygen and glucose to repress respiration. Cell 2013, 152, 224–235. [Google Scholar] [CrossRef] [Green Version]
  79. Rothstein, J.D. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann. Neurol. 2009, 65 (Suppl. 1), S3–S9. [Google Scholar] [CrossRef]
  80. Pare, B.; Lehmann, M.; Beaudin, M.; Nordstrom, U.; Saikali, S.; Julien, J.P.; Gilthorpe, J.D.; Marklund, S.L.; Cashman, N.R.; Andersen, P.M.; et al. Misfolded SOD1 pathology in sporadic Amyotrophic Lateral Sclerosis. Sci. Rep. 2018, 8, 14223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Tokuda, E.; Takei, Y.I.; Ohara, S.; Fujiwara, N.; Hozumi, I.; Furukawa, Y. Wild-type Cu/Zn-superoxide dismutase is misfolded in cerebrospinal fluid of sporadic amyotrophic lateral sclerosis. Mol. Neurodegener. 2019, 14, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Kaur, S.J.; McKeown, S.R.; Rashid, S. Mutant SOD1 mediated pathogenesis of Amyotrophic Lateral Sclerosis. Gene 2016, 577, 109–118. [Google Scholar] [CrossRef] [PubMed]
  83. Petrov, D.; Daura, X.; Zagrovic, B. Effect of Oxidative Damage on the Stability and Dimerization of Superoxide Dismutase 1. Biophys. J. 2016, 110, 1499–1509. [Google Scholar] [CrossRef] [Green Version]
  84. Tak, Y.J.; Park, J.H.; Rhim, H.; Kang, S. ALS-Related Mutant SOD1 Aggregates Interfere with Mitophagy by Sequestering the Autophagy Receptor Optineurin. Int. J. Mol. Sci. 2020, 21, 7525. [Google Scholar] [CrossRef]
  85. Farrawell, N.E.; Lambert-Smith, I.; Mitchell, K.; McKenna, J.; McAlary, L.; Ciryam, P.; Vine, K.L.; Saunders, D.N.; Yerbury, J.J. SOD1A4V aggregation alters ubiquitin homeostasis in a cell model of ALS. J. Cell Sci. 2018, 131. [Google Scholar] [CrossRef] [Green Version]
  86. Beckman, J.S.; Estévez, A.G.; Crow, J.P.; Barbeito, L. Superoxide dismutase and the death of motoneurons in ALS. Trends Neurosci. 2001, 24, S15–S20. [Google Scholar] [CrossRef]
  87. Cunha-Oliveira, T.; Montezinho, L.; Mendes, C.; Firuzi, O.; Saso, L.; Oliveira, P.J.; Silva, F.S.G. Oxidative Stress in Amyotrophic Lateral Sclerosis: Pathophysiology and Opportunities for Pharmacological Intervention. Oxid. Med. Cell. Longev. 2020, 2020, 5021694. [Google Scholar] [CrossRef]
  88. Kirby, J.; Halligan, E.; Baptista, M.J.; Allen, S.; Heath, P.R.; Holden, H.; Barber, S.C.; Loynes, C.A.; Wood-Allum, C.A.; Lunec, J.; et al. Mutant SOD1 alters the motor neuronal transcriptome: Implications for familial ALS. Brain 2005, 128, 1686–1706. [Google Scholar] [CrossRef]
  89. Sarlette, A.; Krampfl, K.; Grothe, C.; Neuhoff, N.; Dengler, R.; Petri, S. Nuclear erythroid 2-related factor 2-antioxidative response element signaling pathway in motor cortex and spinal cord in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2008, 67, 1055–1062. [Google Scholar] [CrossRef] [PubMed]
  90. Ferri, A.; Cozzolino, M.; Crosio, C.; Nencini, M.; Casciati, A.; Gralla, E.B.; Rotilio, G.; Valentine, J.S.; Carri, M.T. Familial ALS-superoxide dismutases associate with mitochondria and shift their redox potentials. Proc. Natl. Acad. Sci. USA 2006, 103, 13860–13865. [Google Scholar] [CrossRef] [Green Version]
  91. Uchino, M.; Ando, Y.; Tanaka, Y.; Nakamura, T.; Uyama, E.; Mita, S.; Murakami, T.; Ando, M. Decrease in Cu/Zn- and Mn-superoxide dismutase activities in brain and spinal cord of patients with amyotrophic lateral sclerosis. J. Neurol. Sci. 1994, 127, 61–67. [Google Scholar] [CrossRef]
  92. O’Reilly, S.A.; Roedica, J.; Nagy, D.; Hallewell, R.A.; Alderson, K.; Marklund, S.L.; Kuby, J.; Kushner, P.D. Motor neuron-astrocyte interactions and levels of Cu,Zn superoxide dismutase in sporadic amyotrophic lateral sclerosis. Exp. Neurol. 1995, 131, 203–210. [Google Scholar] [CrossRef]
  93. Brettschneider, J.; Del Tredici, K.; Toledo, J.B.; Robinson, J.L.; Irwin, D.J.; Grossman, M.; Suh, E.; Van Deerlin, V.M.; Wood, E.M.; Baek, Y.; et al. Stages of pTDP-43 pathology in amyotrophic lateral sclerosis. Ann. Neurol. 2013, 74, 20–38. [Google Scholar] [CrossRef] [PubMed]
  94. Tziortzouda, P.; Van Den Bosch, L.; Hirth, F. Triad of TDP43 control in neurodegeneration: Autoregulation, localization and aggregation. Nat. Rev. Neurosci. 2021, 22, 197–208. [Google Scholar] [CrossRef]
  95. Lagier-Tourenne, C.; Cleveland, D.W. Rethinking ALS: The FUS about TDP-43. Cell 2009, 136, 1001–1004. [Google Scholar] [CrossRef] [Green Version]
  96. Li, Q.; Yokoshi, M.; Okada, H.; Kawahara, Y. The cleavage pattern of TDP-43 determines its rate of clearance and cytotoxicity. Nat. Commun. 2015, 6, 6183. [Google Scholar] [CrossRef] [Green Version]
  97. Goh, C.W.; Lee, I.C.; Sundaram, J.R.; George, S.E.; Yusoff, P.; Brush, M.H.; Sze, N.S.K.; Shenolikar, S. Chronic oxidative stress promotes GADD34-mediated phosphorylation of the TAR DNA-binding protein TDP-43, a modification linked to neurodegeneration. J. Biol. Chem. 2018, 293, 163–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Cohen, T.J.; Hwang, A.W.; Restrepo, C.R.; Yuan, C.X.; Trojanowski, J.Q.; Lee, V.M. An acetylation switch controls TDP-43 function and aggregation propensity. Nat. Commun. 2015, 6, 5845. [Google Scholar] [CrossRef] [Green Version]
  99. Zuo, X.; Zhou, J.; Li, Y.; Wu, K.; Chen, Z.; Luo, Z.; Zhang, X.; Liang, Y.; Esteban, M.A.; Zhou, Y.; et al. TDP-43 aggregation induced by oxidative stress causes global mitochondrial imbalance in ALS. Nat. Struct. Mol. Biol. 2021, 28, 132–142. [Google Scholar] [CrossRef]
  100. Protter, D.S.W.; Parker, R. Principles and Properties of Stress Granules. Trends Cell Biol. 2016, 26, 668–679. [Google Scholar] [CrossRef] [Green Version]
  101. Cohen, T.J.; Hwang, A.W.; Unger, T.; Trojanowski, J.Q.; Lee, V.M. Redox signalling directly regulates TDP-43 via cysteine oxidation and disulphide cross-linking. EMBO J. 2012, 31, 1241–1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Walker, A.K.; Soo, K.Y.; Sundaramoorthy, V.; Parakh, S.; Ma, Y.; Farg, M.A.; Wallace, R.H.; Crouch, P.J.; Turner, B.J.; Horne, M.K.; et al. ALS-associated TDP-43 induces endoplasmic reticulum stress, which drives cytoplasmic TDP-43 accumulation and stress granule formation. PLoS ONE 2013, 8, e81170. [Google Scholar] [CrossRef] [Green Version]
  103. Dewey, C.M.; Cenik, B.; Sephton, C.F.; Dries, D.R.; Mayer, P., III; Good, S.K.; Johnson, B.A.; Herz, J.; Yu, G. TDP-43 is directed to stress granules by sorbitol, a novel physiological osmotic and oxidative stressor. Mol. Cell. Biol. 2011, 31, 1098–1108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Moujalled, D.; Grubman, A.; Acevedo, K.; Yang, S.; Ke, Y.D.; Moujalled, D.M.; Duncan, C.; Caragounis, A.; Perera, N.D.; Turner, B.J.; et al. TDP-43 mutations causing amyotrophic lateral sclerosis are associated with altered expression of RNA-binding protein hnRNP K and affect the Nrf2 antioxidant pathway. Hum. Mol. Genet. 2017, 26, 1732–1746. [Google Scholar] [CrossRef] [PubMed]
  105. Nishimura, S. Involvement of mammalian OGG1(MMH) in excision of the 8-hydroxyguanine residue in DNA. Free Radic. Biol. Med. 2002, 32, 813–821. [Google Scholar] [CrossRef]
  106. Radak, Z.; Boldogh, I. 8-Oxo-7,8-dihydroguanine: Links to gene expression, aging, and defense against oxidative stress. Free Radic. Biol. Med. 2010, 49, 587–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Mecocci, P.; MacGarvey, U.; Kaufman, A.E.; Koontz, D.; Shoffner, J.M.; Wallace, D.C.; Beal, M.F. Oxidative damage to mitochondrial DNA shows marked age-dependent increases in human brain. Ann. Neurol. 1993, 34, 609–616. [Google Scholar] [CrossRef] [PubMed]
  108. Shibata, N.; Nagai, R.; Miyata, S.; Jono, T.; Horiuchi, S.; Hirano, A.; Kato, S.; Sasaki, S.; Asayama, K.; Kobayashi, M. Nonoxidative protein glycation is implicated in familial amyotrophic lateral sclerosis with superoxide dismutase-1 mutation. Acta Neuropathol. 2000, 100, 275–284. [Google Scholar] [CrossRef]
  109. Kim, B.W.; Jeong, Y.E.; Wong, M.; Martin, L.J. DNA damage accumulates and responses are engaged in human ALS brain and spinal motor neurons and DNA repair is activatable in iPSC-derived motor neurons with SOD1 mutations. Acta Neuropathol. Commun. 2020, 8, 7. [Google Scholar] [CrossRef] [Green Version]
  110. Warita, H.; Hayashi, T.; Murakami, T.; Manabe, Y.; Abe, K. Oxidative damage to mitochondrial DNA in spinal motoneurons of transgenic ALS mice. Mol. Brain Res. 2001, 89, 147–152. [Google Scholar] [CrossRef]
  111. Aguirre, N.; Beal, M.F.; Matson, W.R.; Bogdanov, M.B. Increased oxidative damage to DNA in an animal model of amyotrophic lateral sclerosis. Free Radic. Res. 2005, 39, 383–388. [Google Scholar] [CrossRef] [PubMed]
  112. Martin, L.J.; Liu, Z.; Chen, K.; Price, A.C.; Pan, Y.; Swaby, J.A.; Golden, W.C. Motor neuron degeneration in amyotrophic lateral sclerosis mutant superoxide dismutase-1 transgenic mice: Mechanisms of mitochondriopathy and cell death. J. Comp. Neurol. 2007, 500, 20–46. [Google Scholar] [CrossRef]
  113. Barbosa, L.F.; Cerqueira, F.M.; Macedo, A.F.; Garcia, C.C.; Angeli, J.P.; Schumacher, R.I.; Sogayar, M.C.; Augusto, O.; Carri, M.T.; Di Mascio, P.; et al. Increased SOD1 association with chromatin, DNA damage, p53 activation, and apoptosis in a cellular model of SOD1-linked ALS. Biochim. Biophys. Acta 2010, 1802, 462–471. [Google Scholar] [CrossRef]
  114. Lopez-Gonzalez, R.; Lu, Y.; Gendron, T.F.; Karydas, A.; Tran, H.; Yang, D.; Petrucelli, L.; Miller, B.L.; Almeida, S.; Gao, F.B. Poly(GR) in C9ORF72-Related ALS/FTD Compromises Mitochondrial Function and Increases Oxidative Stress and DNA Damage in iPSC-Derived Motor Neurons. Neuron 2016, 92, 383–391. [Google Scholar] [CrossRef] [Green Version]
  115. Wang, H.; Guo, W.; Mitra, J.; Hegde, P.M.; Vandoorne, T.; Eckelmann, B.J.; Mitra, S.; Tomkinson, A.E.; Van Den Bosch, L.; Hegde, M.L. Mutant FUS causes DNA ligation defects to inhibit oxidative damage repair in Amyotrophic Lateral Sclerosis. Nat. Commun. 2018, 9, 3683. [Google Scholar] [CrossRef] [Green Version]
  116. Bradley, W.G.; Krasin, F. A new hypothesis of the etiology of amyotrophic lateral sclerosis. The DNA hypothesis. Arch. Neurol. 1982, 39, 677–680. [Google Scholar] [CrossRef] [PubMed]
  117. Angkeow, P.; Deshpande, S.S.; Qi, B.; Liu, Y.X.; Park, Y.C.; Jeon, B.H.; Ozaki, M.; Irani, K. Redox factor-1: An extra-nuclear role in the regulation of endothelial oxidative stress and apoptosis. Cell Death Differ. 2002, 9, 717–725. [Google Scholar] [CrossRef] [Green Version]
  118. Fishel, M.L.; Wu, X.; Devlin, C.M.; Logsdon, D.P.; Jiang, Y.; Luo, M.; He, Y.; Yu, Z.; Tong, Y.; Lipking, K.P.; et al. Apurinic/apyrimidinic endonuclease/redox factor-1 (APE1/Ref-1) redox function negatively regulates NRF2. J. Biol. Chem. 2015, 290, 3057–3068. [Google Scholar] [CrossRef] [Green Version]
  119. Shaikh, A.Y.; Martin, L.J. DNA base-excision repair enzyme apurinic/apyrimidinic endonuclease/redox factor-1 is increased and competent in the brain and spinal cord of individuals with amyotrophic lateral sclerosis. Neuromolecular. Med. 2002, 2, 47–60. [Google Scholar] [CrossRef] [PubMed]
  120. Li, J.; Song, M.; Moh, S.; Kim, H.; Kim, D.H. Cytoplasmic Restriction of Mutated SOD1 Impairs the DNA Repair Process in Spinal Cord Neurons. Cells 2019, 8, 1502. [Google Scholar] [CrossRef] [Green Version]
  121. Appleby-Mallinder, C.; Schaber, E.; Kirby, J.; Shaw, P.J.; Cooper-Knock, J.; Heath, P.R.; Highley, J.R. TDP43 proteinopathy is associated with aberrant DNA methylation in human amyotrophic lateral sclerosis. Neuropathol. Appl. Neurobiol. 2021, 47, 61–72. [Google Scholar] [CrossRef] [PubMed]
  122. Kisby, G.E.; Milne, J.; Sweatt, C. Evidence of reduced DNA repair in amyotrophic lateral sclerosis brain tissue. NeuroReport 1997, 8, 1337–1340. [Google Scholar] [CrossRef]
  123. Manabe, Y.; Warita, H.; Murakami, T.; Shiote, M.; Hayashi, T.; Nagano, I.; Shoji, M.; Abe, K. Early decrease of redox factor-1 in spinal motor neurons of presymptomatic transgenic mice with a mutant SOD1 gene. Brain Res. 2001, 915, 104–107. [Google Scholar] [CrossRef]
  124. Kikuchi, H.; Furuta, A.; Nishioka, K.; Suzuki, S.O.; Nakabeppu, Y.; Iwaki, T. Impairment of mitochondrial DNA repair enzymes against accumulation of 8-oxo-guanine in the spinal motor neurons of amyotrophic lateral sclerosis. Acta Neuropathol. 2002, 103, 408–414. [Google Scholar] [CrossRef] [PubMed]
  125. Kim, S.H.; Henkel, J.S.; Beers, D.R.; Sengun, I.S.; Simpson, E.P.; Goodman, J.C.; Engelhardt, J.I.; Siklós, L.; Appel, S.H. PARP expression is increased in astrocytes but decreased in motor neurons in the spinal cord of sporadic ALS patients. J. Neuropathol. Exp. Neurol 2003, 62, 88–103. [Google Scholar] [CrossRef] [Green Version]
  126. Farg, M.A.; Konopka, A.; Soo, K.Y.; Ito, D.; Atkin, J.D. The DNA damage response (DDR) is induced by the C9orf72 repeat expansion in amyotrophic lateral sclerosis. Hum. Mol. Genet. 2017, 26, 2882–2896. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, W.Y.; Pan, L.; Su, S.C.; Quinn, E.J.; Sasaki, M.; Jimenez, J.C.; Mackenzie, I.R.; Huang, E.J.; Tsai, L.H. Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons. Nat. Neurosci. 2013, 16, 1383–1391. [Google Scholar] [CrossRef]
  128. Bozzo, F.; Mirra, A.; Carri, M.T. Oxidative stress and mitochondrial damage in the pathogenesis of ALS: New perspectives. Neurosci. Lett. 2017, 636, 3–8. [Google Scholar] [CrossRef]
  129. Chang, Y.; Kong, Q.; Shan, X.; Tian, G.; Ilieva, H.; Cleveland, D.W.; Rothstein, J.D.; Borchelt, D.R.; Wong, P.C.; Lin, C.L. Messenger RNA oxidation occurs early in disease pathogenesis and promotes motor neuron degeneration in ALS. PLoS ONE 2008, 3, e2849. [Google Scholar] [CrossRef]
  130. Lenzken, S.C.; Romeo, V.; Zolezzi, F.; Cordero, F.; Lamorte, G.; Bonanno, D.; Biancolini, D.; Cozzolino, M.; Pesaresi, M.G.; Maracchioni, A.; et al. Mutant SOD1 and mitochondrial damage alter expression and splicing of genes controlling neuritogenesis in models of neurodegeneration. Hum. Mutat. 2011, 32, 168–182. [Google Scholar] [CrossRef] [Green Version]
  131. Pham, J.; Keon, M.; Brennan, S.; Saksena, N. Connecting RNA-Modifying Similarities of TDP-43, FUS, and SOD1 with MicroRNA Dysregulation Amidst A Renewed Network Perspective of Amyotrophic Lateral Sclerosis Proteinopathy. Int. J. Mol. Sci. 2020, 21, 3464. [Google Scholar] [CrossRef] [PubMed]
  132. Zhao, Y.; Dong, D.; Reece, E.A.; Wang, A.R.; Yang, P. Oxidative stress-induced miR-27a targets the redox gene nuclear factor erythroid 2-related factor 2 in diabetic embryopathy. Am. J. Obstet. Gynecol. 2018, 218, 131.e1–136.e10. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, N.; Zhang, L.; Lu, Y.; Zhang, M.; Zhang, Z.; Wang, K.; Lv, J. Down-regulation of microRNA-142-5p attenuates oxygen-glucose deprivation and reoxygenation-induced neuron injury through up-regulating Nrf2/ARE signaling pathway. Biomed. Pharmacother. 2017, 89, 1187–1195. [Google Scholar] [CrossRef]
  134. Pegoraro, V.; Merico, A.; Angelini, C. Micro-RNAs in ALS muscle: Differences in gender, age at onset and disease duration. J. Neurol. Sci. 2017, 380, 58–63. [Google Scholar] [CrossRef] [Green Version]
  135. Aschrafi, A.; Kar, A.N.; Natera-Naranjo, O.; MacGibeny, M.A.; Gioio, A.E.; Kaplan, B.B. MicroRNA-338 regulates the axonal expression of multiple nuclear-encoded mitochondrial mRNAs encoding subunits of the oxidative phosphorylation machinery. Cell. Mol. Life Sci. 2012, 69, 4017–4027. [Google Scholar] [CrossRef] [PubMed]
  136. De Felice, B.; Manfellotto, F.; Fiorentino, G.; Annunziata, A.; Biffali, E.; Pannone, R.; Federico, A. Wide-Ranging Analysis of MicroRNA Profiles in Sporadic Amyotrophic Lateral Sclerosis Using Next-Generation Sequencing. Front. Genet. 2018, 9, 310. [Google Scholar] [CrossRef]
  137. Li, C.; Wei, Q.; Gu, X.; Chen, Y.; Chen, X.; Cao, B.; Ou, R.; Shang, H. Decreased Glycogenolysis by miR-338-3p Promotes Regional Glycogen Accumulation Within the Spinal Cord of Amyotrophic Lateral Sclerosis Mice. Front. Mol. Neurosci. 2019, 12, 114. [Google Scholar] [CrossRef] [Green Version]
  138. De Felice, B.; Annunziata, A.; Fiorentino, G.; Borra, M.; Biffali, E.; Coppola, C.; Cotrufo, R.; Brettschneider, J.; Giordana, M.L.; Dalmay, T.; et al. miR-338-3p is over-expressed in blood, CFS, serum and spinal cord from sporadic amyotrophic lateral sclerosis patients. Neurogenetics 2014, 15, 243–253. [Google Scholar] [CrossRef]
  139. Li, N.; Muthusamy, S.; Liang, R.; Sarojini, H.; Wang, E. Increased expression of miR-34a and miR-93 in rat liver during aging, and their impact on the expression of Mgst1 and Sirt1. Mech. Ageing Dev. 2011, 132, 75–85. [Google Scholar] [CrossRef]
  140. Zhou, F.; Zhang, C.; Guan, Y.; Chen, Y.; Lu, Q.; Jie, L.; Gao, H.; Du, H.; Zhang, H.; Liu, Y.; et al. Screening the expression characteristics of several miRNAs in G93A-SOD1 transgenic mouse: Altered expression of miRNA-124 is associated with astrocyte differentiation by targeting Sox2 and Sox9. J. Neurochem. 2018, 145, 51–67. [Google Scholar] [CrossRef] [Green Version]
  141. Rizzuti, M.; Filosa, G.; Melzi, V.; Calandriello, L.; Dioni, L.; Bollati, V.; Bresolin, N.; Comi, G.P.; Barabino, S.; Nizzardo, M.; et al. MicroRNA expression analysis identifies a subset of downregulated miRNAs in ALS motor neuron progenitors. Sci. Rep. 2018, 8, 10105. [Google Scholar] [CrossRef]
  142. Di Pietro, L.; Baranzini, M.; Berardinelli, M.G.; Lattanzi, W.; Monforte, M.; Tasca, G.; Conte, A.; Logroscino, G.; Michetti, F.; Ricci, E.; et al. Potential therapeutic targets for ALS: MIR206, MIR208b and MIR499 are modulated during disease progression in the skeletal muscle of patients. Sci. Rep. 2017, 7, 9538. [Google Scholar] [CrossRef] [PubMed]
  143. Koval, E.D.; Shaner, C.; Zhang, P.; du Maine, X.; Fischer, K.; Tay, J.; Chau, B.N.; Wu, G.F.; Miller, T.M. Method for widespread microRNA-155 inhibition prolongs survival in ALS-model mice. Hum. Mol. Genet. 2013, 22, 4127–4135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Butovsky, O.; Jedrychowski, M.P.; Cialic, R.; Krasemann, S.; Murugaiyan, G.; Fanek, Z.; Greco, D.J.; Wu, P.M.; Doykan, C.E.; Kiner, O.; et al. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann. Neurol. 2015, 77, 75–99. [Google Scholar] [CrossRef] [PubMed]
  145. Butovsky, O.; Siddiqui, S.; Gabriely, G.; Lanser, A.J.; Dake, B.; Murugaiyan, G.; Doykan, C.E.; Wu, P.M.; Gali, R.R.; Iyer, L.K.; et al. Modulating inflammatory monocytes with a unique microRNA gene signature ameliorates murine ALS. J. Clin. Investig. 2012, 122, 3063–3087. [Google Scholar] [CrossRef] [PubMed]
  146. Cunha, C.; Santos, C.; Gomes, C.; Fernandes, A.; Correia, A.M.; Sebastiao, A.M.; Vaz, A.R.; Brites, D. Downregulated Glia Interplay and Increased miRNA-155 as Promising Markers to Track ALS at an Early Stage. Mol. Neurobiol. 2018, 55, 4207–4224. [Google Scholar] [CrossRef]
  147. Ito, H.; Wate, R.; Zhang, J.; Ohnishi, S.; Kaneko, S.; Ito, H.; Nakano, S.; Kusaka, H. Treatment with edaravone, initiated at symptom onset, slows motor decline and decreases SOD1 deposition in ALS mice. Exp. Neurol. 2008, 213, 448–455. [Google Scholar] [CrossRef]
  148. Song, Y.; Li, M.; Li, J.C.; Wei, E.Q. Edaravone protects PC12 cells from ischemic-like injury via attenuating the damage to mitochondria. J. Zhejiang Univ. Sci. B 2006, 7, 749–756. [Google Scholar] [CrossRef] [Green Version]
  149. Yamashita, T.; Shoge, M.; Oda, E.; Yamamoto, Y.; Giddings, J.C.; Kashiwagi, S.; Suematsu, M.; Yamamoto, J. The free-radical scavenger, edaravone, augments NO release from vascular cells and platelets after laser-induced, acute endothelial injury in vivo. Platelets 2006, 17, 201–206. [Google Scholar] [CrossRef]
  150. Qi, X.; Okuma, Y.; Hosoi, T.; Nomura, Y. Edaravone protects against hypoxia/ischemia-induced endoplasmic reticulum dysfunction. J. Pharmacol. Exp. Ther. 2004, 311, 388–393. [Google Scholar] [CrossRef]
  151. Luo, L.; Song, Z.; Li, X.; Zeng, Y.; He, J. Efficacy and safety of edaravone in treatment of amyotrophic lateral sclerosis-a systematic review and meta-analysis. Neurol. Sci. 2019, 40, 235–241. [Google Scholar] [CrossRef] [PubMed]
  152. Okada, M.; Yamashita, S.; Ueyama, H.; Ishizaki, M.; Maeda, Y.; Ando, Y. Long-Term effects of edaravone on survival of patients with amyotrophic lateral sclerosis. ENeurologicalSci 2018, 11, 11–14. [Google Scholar] [CrossRef]
  153. Park, J.M.; Kim, S.Y.; Park, D.; Park, J.S. Effect of edaravone therapy in Korean amyotrophic lateral sclerosis (ALS) patients. Neurol. Sci. 2020, 41, 119–123. [Google Scholar] [CrossRef] [PubMed]
  154. Yoshino, H.; Kimura, A. Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study). Amyotroph. Lateral Scler. 2006, 7, 241–245. [Google Scholar] [CrossRef]
  155. Ohta, Y.; Yamashita, T.; Nomura, E.; Hishikawa, N.; Ikegami, K.; Osakada, Y.; Matsumoto, N.; Kawahara, Y.; Yunoki, T.; Takahashi, Y.; et al. Improvement of a decreased anti-oxidative activity by edaravone in amyotrophic lateral sclerosis patients. J. Neurol. Sci. 2020, 415, 116906. [Google Scholar] [CrossRef]
  156. Fortuna, A.; Gizzi, M.; Bello, L.; Martinelli, I.; Bertolin, C.; Pegoraro, E.; Corbetta, M.; Soraru, G.; Edaravone Study, G. Safety and efficacy of edaravone compared to historical controls in patients with amyotrophic lateral sclerosis from North-Eastern Italy. J. Neurol. Sci. 2019, 404, 47–51. [Google Scholar] [CrossRef]
  157. Lunetta, C.; Moglia, C.; Lizio, A.; Caponnetto, C.; Dubbioso, R.; Giannini, F.; Mata, S.; Mazzini, L.; Sabatelli, M.; Siciliano, G.; et al. The Italian multicenter experience with edaravone in amyotrophic lateral sclerosis. J. Neurol. 2020, 267, 3258–3267. [Google Scholar] [CrossRef] [PubMed]
  158. Desnuelle, C.; Dib, M.; Garrel, C.; Favier, A. A double-blind, placebo-controlled randomized clinical trial of α-tocopherol (vitamin E) in the treatment of amyotrophic lateral sclerosis. ALS riluzole-tocopherol Study Group. Amyotroph. Lateral Scler. Motor Neuron Disord. 2001, 2, 9–18. [Google Scholar] [CrossRef]
  159. Fitzgerald, K.C.; O’Reilly, E.J.; Fondell, E.; Falcone, G.J.; McCullough, M.L.; Park, Y.; Kolonel, L.N.; Ascherio, A. Intakes of vitamin C and carotenoids and risk of amyotrophic lateral sclerosis: Pooled results from 5 cohort studies. Ann. Neurol. 2013, 73, 236–245. [Google Scholar] [CrossRef] [Green Version]
  160. Ferrante, K.L.; Shefner, J.; Zhang, H.; Betensky, R.; O’Brien, M.; Yu, H.; Fantasia, M.; Taft, J.; Beal, M.F.; Traynor, B.; et al. Tolerance of high-dose (3000 mg/day) coenzyme Q10 in ALS. Neurology 2005, 65, 1834–1836. [Google Scholar] [CrossRef]
  161. Ahmadi, M.; Agah, E.; Nafissi, S.; Jaafari, M.R.; Harirchian, M.H.; Sarraf, P.; Faghihi-Kashani, S.; Hosseini, S.J.; Ghoreishi, A.; Aghamollaii, V.; et al. Safety and Efficacy of Nanocurcumin as Add-On Therapy to Riluzole in Patients With Amyotrophic Lateral Sclerosis: A Pilot Randomized Clinical Trial. Neurotherapeutics 2018, 15, 430–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Chico, L.; Ienco, E.C.; Bisordi, C.; Lo Gerfo, A.; Petrozzi, L.; Petrucci, A.; Mancuso, M.; Siciliano, G. Amyotrophic Lateral Sclerosis and Oxidative Stress: A Double-Blind Therapeutic Trial After Curcumin Supplementation. CNS Neurol Disord Drug Targets 2018, 17, 767–779. [Google Scholar] [CrossRef] [PubMed]
  163. de la Rubia, J.E.; Drehmer, E.; Platero, J.L.; Benlloch, M.; Caplliure-Llopis, J.; Villaron-Casales, C.; de Bernardo, N.; AlarcOn, J.; Fuente, C.; Carrera, S.; et al. Efficacy and tolerability of EH301 for amyotrophic lateral sclerosis: A randomized, double-blind, placebo-controlled human pilot study. Amyotroph. Lateral Scler. Frontotemporal Degener. 2019, 20, 115–122. [Google Scholar] [CrossRef]
  164. Wang, Z.; Bai, Z.; Qin, X.; Cheng, Y. Aberrations in Oxidative Stress Markers in Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. Oxid. Med. Cell. Longev. 2019, 2019, 1712323. [Google Scholar] [CrossRef]
  165. Ihara, Y.; Nobukuni, K.; Takata, H.; Hayabara, T. Oxidative stress and metal content in blood and cerebrospinal fluid of amyotrophic lateral sclerosis patients with and without a Cu, Zn-superoxide dismutase mutation. Neurol. Res. 2005, 27, 105–108. [Google Scholar] [CrossRef]
  166. Blasco, H.; Garcon, G.; Patin, F.; Veyrat-Durebex, C.; Boyer, J.; Devos, D.; Vourc’h, P.; Andres, C.R.; Corcia, P. Panel of Oxidative Stress and Inflammatory Biomarkers in ALS: A Pilot Study. Can. J. Neurol. Sci. 2017, 44, 90–95. [Google Scholar] [CrossRef] [Green Version]
  167. Moumen, R.; Nouvelot, A.; Duval, D.; Lechevalier, B.; Viader, F. Plasma superoxide dismutase and glutathione peroxidase activity in sporadic amyotrophic lateral sclerosis. J. Neurol. Sci. 1997, 151, 35–39. [Google Scholar] [CrossRef]
  168. Baillet, A.; Chanteperdrix, V.; Trocme, C.; Casez, P.; Garrel, C.; Besson, G. The role of oxidative stress in amyotrophic lateral sclerosis and Parkinson’s disease. Neurochem. Res. 2010, 35, 1530–1537. [Google Scholar] [CrossRef] [PubMed]
  169. Tohgi, H.; Abe, T.; Yamazaki, K.; Murata, T.; Ishizaki, E.; Isobe, C. Remarkable increase in cerebrospinal fluid 3-nitrotyrosine in patients with sporadic amyotrophic lateral sclerosis. Ann. Neurol. 1999, 46, 129–131. [Google Scholar] [CrossRef]
  170. Ryberg, H.; Soderling, A.S.; Davidsson, P.; Blennow, K.; Caidahl, K.; Persson, L.I. Cerebrospinal fluid levels of free 3-nitrotyrosine are not elevated in the majority of patients with amyotrophic lateral sclerosis or Alzheimer’s disease. Neurochem. Int. 2004, 45, 57–62. [Google Scholar] [CrossRef]
  171. Ihara, Y.; Mori, A.; Hayabara, T.; Kawai, M.; Namba, R.; Nobukuni, K.; Sato, K.; Kibata, M. Superoxide dismutase and free radicals in sporadic amyotrophic lateral sclerosis: Relationship to clinical data. J. Neurol. Sci. 1995, 134, 51–56. [Google Scholar] [CrossRef]
  172. Bonnefont-Rousselot, D.; Lacomblez, L.; Jaudon, M.; Lepage, S.; Salachas, F.; Bensimon, G.; Bizard, C.; Doppler, V.; Delattre, J.; Meininger, V. Blood oxidative stress in amyotrophic lateral sclerosis. J. Neurol. Sci. 2000, 178, 57–62. [Google Scholar] [CrossRef]
  173. Przedborski, S.; Donaldson, D.M.; Murphy, P.L.; Hirsch, O.; Lange, D.; Naini, A.B.; McKenna-Yasek, D.; Brown, R.H., Jr. Blood superoxide dismutase, catalase and glutathione peroxidase activities in familial and sporadic amyotrophic lateral sclerosis. Neurodegeneration 1996, 5, 57–64. [Google Scholar] [CrossRef]
  174. Iwasaki, Y.; Ikeda, K.; Kinoshita, M. Decreased cerebrospinal-fluid superoxide dismutase in amyotrophic lateral sclerosis. Lancet 1993, 342, 1118. [Google Scholar] [CrossRef]
  175. Robberecht, W.; Sapp, P.; Viaene, M.K.; Rosen, D.; McKenna-Yasek, D.; Haines, J.; Horvitz, R.; Theys, P.; Brown, R., Jr. Cu/Zn superoxide dismutase activity in familial and sporadic amyotrophic lateral sclerosis. J. Neurochem. 1994, 62, 384–387. [Google Scholar] [CrossRef] [PubMed]
  176. Puymirat, J.; Cossette, L.; Gosselin, F.; Bouchard, J.P. Red blood cell Cu/Zn superoxide dismutase activity in sporadic amyotrophic lateral sclerosis. J. Neurol. Sci. 1994, 127, 121–123. [Google Scholar] [CrossRef]
  177. Tórsdóttir, G.; Kristinsson, J.; Gudmundsson, G.; Snaedal, J.; Jóhannesson, T. Copper, ceruloplasmin and superoxide dismutase (SOD) in amyotrophic lateral sclerosis. Pharmacol. Toxicol. 2000, 87, 126–130. [Google Scholar] [CrossRef]
  178. Mitchell, J.D.; Gatt, J.A.; Phillips, T.M.; Houghton, E.; Rostron, G.; Wignall, C.; Whittington, J.; Shaw, I.C. Cu/Zn superoxide dismutase free radicals, and motoneuron disease. Lancet 1993, 342, 1051–1052. [Google Scholar] [CrossRef]
  179. Ehrhart, J.; Smith, A.J.; Kuzmin-Nichols, N.; Zesiewicz, T.A.; Jahan, I.; Shytle, R.D.; Kim, S.H.; Sanberg, C.D.; Vu, T.H.; Gooch, C.L.; et al. Humoral factors in ALS patients during disease progression. J. Neuroinflamm. 2015, 12, 127. [Google Scholar] [CrossRef] [Green Version]
  180. Oteiza, P.I.; Uchitel, O.D.; Carrasquedo, F.; Dubrovski, A.L.; Roma, J.C.; Fraga, C.G. Evaluation of antioxidants, protein, and lipid oxidation products in blood from sporadic amyotrophic lateral sclerosis patients. Neurochem. Res. 1997, 22, 535–539. [Google Scholar] [CrossRef]
  181. Molina, J.A.; de Bustos, F.; Jiménez-Jiménez, F.J.; Gómez-Escalonilla, C.; García-Redondo, A.; Esteban, J.; Guerrero-Sola, A.; del Hoyo, P.; Martínez-Salio, A.; Ramírez-Ramos, C.; et al. Serum levels of coenzyme Q10 in patients with amyotrophic lateral sclerosis. J. Neural. Transm. 2000, 107, 1021–1026. [Google Scholar] [CrossRef] [PubMed]
  182. Nagase, M.; Yamamoto, Y.; Miyazaki, Y.; Yoshino, H. Increased oxidative stress in patients with amyotrophic lateral sclerosis and the effect of edaravone administration. Redox Rep. 2016, 21, 104–112. [Google Scholar] [CrossRef]
  183. Wuolikainen, A.; Acimovic, J.; Lovgren-Sandblom, A.; Parini, P.; Andersen, P.M.; Bjorkhem, I. Cholesterol, oxysterol, triglyceride, and coenzyme Q homeostasis in ALS. Evidence against the hypothesis that elevated 27-hydroxycholesterol is a pathogenic factor. PLoS ONE 2014, 9, e113619. [Google Scholar] [CrossRef]
  184. Murata, T.; Ohtsuka, C.; Terayama, Y. Increased mitochondrial oxidative damage and oxidative DNA damage contributes to the neurodegenerative process in sporadic amyotrophic lateral sclerosis. Free Radic. Res. 2008, 42, 221–225. [Google Scholar] [CrossRef]
  185. Sohmiya, M.; Tanaka, M.; Suzuki, Y.; Tanino, Y.; Okamoto, K.; Yamamoto, Y. An increase of oxidized coenzyme Q-10 occurs in the plasma of sporadic ALS patients. J. Neurol. Sci. 2005, 228, 49–53. [Google Scholar] [CrossRef]
  186. Keizman, D.; Ish-Shalom, M.; Berliner, S.; Maimon, N.; Vered, Y.; Artamonov, I.; Tsehori, J.; Nefussy, B.; Drory, V.E. Low uric acid levels in serum of patients with ALS: Further evidence for oxidative stress? J. Neurol. Sci. 2009, 285, 95–99. [Google Scholar] [CrossRef] [PubMed]
  187. Ikeda, K.; Kawabe, K.; Iwasaki, Y. Do serum uric acid levels reflect oxidative stress in the progression of ALS? J. Neurol. Sci. 2009, 287, P294. [Google Scholar] [CrossRef] [PubMed]
  188. Ikeda, K.; Hirayama, T.; Takazawa, T.; Kawabe, K.; Iwasaki, Y. Relationships between disease progression and serum levels of lipid, urate, creatinine and ferritin in Japanese patients with amyotrophic lateral sclerosis: A cross-sectional study. Intern. Med. 2012, 51, 1501–1508. [Google Scholar] [CrossRef] [Green Version]
  189. Zoccolella, S.; Simone, I.L.; Capozzo, R.; Tortelli, R.; Leo, A.; D’Errico, E.; Logroscino, G. An exploratory study of serum urate levels in patients with amyotrophic lateral sclerosis. J. Neurol. 2011, 258, 238–243. [Google Scholar] [CrossRef]
  190. Oh, S.I.; Baek, S.; Park, J.S.; Piao, L.; Oh, K.W.; Kim, S.H. Prognostic Role of Serum Levels of Uric Acid in Amyotrophic Lateral Sclerosis. J. Clin. Neurol. 2015, 11, 376–382. [Google Scholar] [CrossRef] [Green Version]
  191. Zheng, Z.; Guo, X.; Wei, Q.; Song, W.; Cao, B.; Huang, R.; Ou, R.; Chen, X.; Shang, H. Serum uric acid level is associated with the prevalence but not with survival of amyotrophic lateral sclerosis in a Chinese population. Metab. Brain Dis. 2014, 29, 771–775. [Google Scholar] [CrossRef] [PubMed]
  192. Lv, Q.; Wang, Z.; Zhong, Z.; Huang, W. Role of Long Noncoding RNAs in Parkinson’s Disease: Putative Biomarkers and Therapeutic Targets. Parkinson’s Dis. 2020, 2020, 5374307. [Google Scholar] [CrossRef]
  193. Ricci, C.; Marzocchi, C.; Battistini, S. MicroRNAs as Biomarkers in Amyotrophic Lateral Sclerosis. Cells 2018, 7, 219. [Google Scholar] [CrossRef] [Green Version]
  194. Tasca, E.; Pegoraro, V.; Merico, A.; Angelini, C. Circulating microRNAs as biomarkers of muscle differentiation and atrophy in ALS. Clin. Neuropathol. 2016, 35, 22–30. [Google Scholar] [CrossRef]
  195. Xu, Q.; Zhao, Y.; Zhou, X.; Luan, J.; Cui, Y.; Han, J. Comparison of the extraction and determination of serum exosome and miRNA in serum and the detection of miR-27a-3p in serum exosome of ALS patients. Intractable Rare Dis. Res. 2018, 7, 13–18. [Google Scholar] [CrossRef] [Green Version]
  196. Waller, R.; Wyles, M.; Heath, P.R.; Kazoka, M.; Wollff, H.; Shaw, P.J.; Kirby, J. Small RNA Sequencing of Sporadic Amyotrophic Lateral Sclerosis Cerebrospinal Fluid Reveals Differentially Expressed miRNAs Related to Neural and Glial Activity. Front. Neurosci. 2017, 11, 731. [Google Scholar] [CrossRef]
  197. De Felice, B.; Guida, M.; Guida, M.; Coppola, C.; De Mieri, G.; Cotrufo, R. A miRNA signature in leukocytes from sporadic amyotrophic lateral sclerosis. Gene 2012, 508, 35–40. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Oxidative molecular modifications causing protein aggregation. Oxidative biomolecular modifications including proteins or nucleic acids occurred in both the nucleus and cytoplasm. Oxidative modifications of proteins can lead to conformational changes and protein misfolding, resulting in protein aggregation. Moreover, endoplasmic reticulum (ER) stress due to impairment of ER degradation and impairment of nuclear import results in protein aggregation. Oxidative damage of DNA and RNA, impairment of DNA repair pathways, and mitochondria dysfunction lead to apoptosis.
Figure 1. Oxidative molecular modifications causing protein aggregation. Oxidative biomolecular modifications including proteins or nucleic acids occurred in both the nucleus and cytoplasm. Oxidative modifications of proteins can lead to conformational changes and protein misfolding, resulting in protein aggregation. Moreover, endoplasmic reticulum (ER) stress due to impairment of ER degradation and impairment of nuclear import results in protein aggregation. Oxidative damage of DNA and RNA, impairment of DNA repair pathways, and mitochondria dysfunction lead to apoptosis.
Processes 09 01667 g001
Table 1. Promising biomarkers related to oxidative stress.
Table 1. Promising biomarkers related to oxidative stress.
Proteins
Related to Redox SystemNameKinds of Body FluidsChangesRelated to Parameter of ALSReference
Products of oxidative stress8-OHdGCSF
Plasma
Urine
Increase8-OHdG levels in urine were
negatively correlated with both the
rate of change of the ALSFRS-R and the FVC
[69,74,165,166]
MDAPlasmaIncreaseNot described[166,167,168]
HNEPlasma
CSF
IncreaseHNE levels in serum were
positively correlated with the
extent of disease
[75]
3-NTCSFIncrease or
no change
(inconsistent result)
Not described[169,170]
Antioxidant enzymeActivity of SODRBC
Plasma
CSF
Increase or reduce or no change (inconsistent
result)
The activity in bulbar onset
sporadic ALS was significantly higher than in spinal onset
sporadic ALS
[165,166,167,168,171,172,173,174,175,176,177]
activity of GPXWhole blood
RBC
Plasma
Reduce or
no change
(inconsistent result)
Not described[166,167,168,172,173,178]
GSHWhole blood
Plasma
Reduce or
no change
( inconsistent result)
No significant temporal change during 6 months was observed in sporadic ALS[166,168,179,180]
GSSGWhole bloodIncreaseNot described[166]
CoQ10Plasma
Serum
No changeCoQ10 levels were not influenced by clinical form, age at onset, and duration of disease[181,182,183]
Proportion of
oxidized forms of CoQ10
Serum
CSF
IncreaseProportion of oxidized forms of CoQ10 correlated with duration of disease[184,185]
UASerum
Plasma
Reduce or
no change (inconsistent result)
The levels of UA in bulbar onset sporadic ALS were significantly decreased compared with spinal onset sporadic ALS patients.
The baseline levels of UA were negatively correlated with the annual decline of ALSFRS-R.
[182,186,187,188,189,190,191]
RNAs
Related to Redox SystemNameKinds of Body FluidsChangesRelated to the Parameter of ALSReference
The NRF2-ARE pathway miR-27aSerumDecreaseDownregulation of miR-27a was correlated with the degree of muscle atrophy[194,195]
miR-142-5pCSFDecreaseNot described[145,196]
Mitochondrial
dysfunction and neuroinflammation
miR-155Peripheral monocyteIncreaseNot described[145,196]
Mitochondrial
dysfunction
miR-338-3pSerum
PBL
CSF
IncreaseNot described[138,197]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hosaka, T.; Tsuji, H.; Tamaoka, A. Biomolecular Modifications Linked to Oxidative Stress in Amyotrophic Lateral Sclerosis: Determining Promising Biomarkers Related to Oxidative Stress. Processes 2021, 9, 1667. https://doi.org/10.3390/pr9091667

AMA Style

Hosaka T, Tsuji H, Tamaoka A. Biomolecular Modifications Linked to Oxidative Stress in Amyotrophic Lateral Sclerosis: Determining Promising Biomarkers Related to Oxidative Stress. Processes. 2021; 9(9):1667. https://doi.org/10.3390/pr9091667

Chicago/Turabian Style

Hosaka, Takashi, Hiroshi Tsuji, and Akira Tamaoka. 2021. "Biomolecular Modifications Linked to Oxidative Stress in Amyotrophic Lateral Sclerosis: Determining Promising Biomarkers Related to Oxidative Stress" Processes 9, no. 9: 1667. https://doi.org/10.3390/pr9091667

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop