Svoboda | Graniru | BBC Russia | Golosameriki | Facebook
Next Article in Journal
Assessing Transcriptomic Responses to Oxidative Stress: Contrasting Wild-Type Arabidopsis Seedlings with dss1(I) and dss1(V) Gene Knockout Mutants
Previous Article in Journal
The Gap Junction Inhibitor Octanol Decreases Proliferation and Increases Glial Differentiation of Postnatal Neural Progenitor Cells
Previous Article in Special Issue
Modulation of the Circadian Rhythm and Oxidative Stress as Molecular Targets to Improve Vascular Dementia: A Pharmacological Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidative Stress Markers in Multiple Sclerosis

by
Félix Javier Jiménez-Jiménez
1,*,
Hortensia Alonso-Navarro
1,
Paula Salgado-Cámara
1,
Elena García-Martín
2 and
José A. G. Agúndez
2
1
Section of Neurology, Hospital Universitario del Sureste, Arganda del Rey, E-28500 Madrid, Spain
2
University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, E-10071 Cáceres, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(12), 6289; https://doi.org/10.3390/ijms25126289
Submission received: 29 January 2024 / Revised: 10 March 2024 / Accepted: 3 June 2024 / Published: 7 June 2024
(This article belongs to the Special Issue Peripheral Biomarkers in Neurodegenerative Diseases—4th Edition)

Abstract

:
The pathogenesis of multiple sclerosis (MS) is not completely understood, but genetic factors, autoimmunity, inflammation, demyelination, and neurodegeneration seem to play a significant role. Data from analyses of central nervous system autopsy material from patients diagnosed with multiple sclerosis, as well as from studies in the main experimental model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), suggest the possibility of a role of oxidative stress as well. In this narrative review, we summarize the main data from studies reported on oxidative stress markers in patients diagnosed with MS and in experimental models of MS (mainly EAE), and case–control association studies on the possible association of candidate genes related to oxidative stress with risk for MS. Most studies have shown an increase in markers of oxidative stress, a decrease in antioxidant substances, or both, with cerebrospinal fluid and serum/plasma malonyl-dialdehyde being the most reliable markers. This topic requires further prospective, multicenter studies with a long-term follow-up period involving a large number of patients with MS and controls.

1. Introduction

Multiple sclerosis (MS), which is characterized mainly by inflammation, demyelination, and neuronal degeneration, is considered to be a chronic autoimmune disease with a genetic predisposition affecting the central nervous system. To date, at least 200 loci with genome-wide significance have been associated with the risk for MS through genome-wide association studies (GWAS) [1,2]. Most of the described associations, however, show a modest odds ratio (OR) and explain close to half of its heritability [1,2], HLA (in particular, the HLA-DRB1*15:01 haplotype) being the only one that has shown a strong association with MS risk [1]. It has been suggested that, together with genetic predisposition, some environmental factors, gene–environment, and environment–environment interactions, including smoking, infections (mainly Epstein–Barr virus seropositivity or exposure), low sun exposure/low vitamin D levels, and obesity may be related to the etiopathogenesis of MS and with MS onset and progression [3,4,5]. Since it has been suggested that oxidative stress is closely related to inflammation (for example, in inflammatory conditions, immune cells can liberate reactive oxidant substances leading to oxidative stress and, on the other hand, the oxidative damage produced by free radicals can induce an inflammatory response through the Toll-like receptors and inflammasomes) [6,7], with MS being a prototype of inflammatory diseases, oxidative stress could also play a role in the etiopathogenesis of MS. Figure 1 depicts the possible interaction between the different mechanisms proposed in the etiopathogenesis of MS, including oxidative stress.
The term “oxidative stress” designates the imbalance between the production of reactive oxygen species (ROS) and the ability of a biological system to neutralize intermediate reagents or to repair the resulting damage. Biomarkers of oxidative stress can be divided into molecules modified by their interaction with ROS or free radicals derived from nitrogen (RNS) and into molecules of the antioxidant system in response to an increase in redox stress. These include lipid peroxidation, protein oxidation, DNA oxidation markers, enzymes or protein with antioxidant actions, other prooxidant and antioxidant substances, and global markers of oxidative processes, such as the total oxidant status/capacity (TOS/TOC), total antioxidant status/capacity (TAS/TAC), and oxidative stress index (OSI).
This narrative review aims to analyze the results of published studies on the possible role of oxidative stress in multiple sclerosis, mainly those related to oxidative stress markers in different tissues from patients diagnosed with MS, but also case–control association studies on the possible association of candidate genes related to oxidative stress with risk for MS, and studies showing the presence of oxidative stress in experimental models of multiple sclerosis. To this end, we performed a PubMed Database search from 1966 to 28 December 2023, crossing the terms “multiple sclerosis” and “oxidative stress”. The search retrieved 1672 references that were manually selected to include only those strictly related to the topic (a total of 201 references).

2. Oxidative Stress Markers in Patients with Multiple Sclerosis

2.1. Oxidative Stress Markers in the Brain and Spinal Cord

The results of studies on oxidative markers in the brains or spinal cord of patients with MS, with most of them compared to controls [8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31], are summarized in Table S1. Many autopsy studies describe an increase in various markers of lipid peroxidation [8,9,10,11,12,13,14,15], an increase in carbonylated proteins [16], markers of DNA damage [9,11,12,17], and nitrotyrosine (a marker of nitrosative stress) [8,11] in the brains of patients diagnosed with MS, especially in active MS plaques. The enzymatic activity of superoxide-dismutase 1 and 2 is upregulated in active demyelinating lesions [10] and in cerebellar gray matter of patients with MS [11], and catalase activity is increased in active demyelinating lesions [11]. In contrast, glutathione peroxidase (GPx) [10] and catalase [10] activities are similar to those of controls in cerebellar gray matter. Iron content has been found to decrease in MS inactive lesions [12], mitochondrial protein expression is increased, and mitochondrial complex IV activity is upregulated in MS lesions [18]. Studies with proton magnetic resonance spectroscopy (1HMRS) found decreased glutathione in some brain regions of MS patients [19,20,21].
Several studies have also reported the upregulation of multiple enzymes and proteins involved in oxidative processes, such as NAD(P)H:quinone oxidoreductase 1 (NQO1) [22], some subunits of NADPH oxidase 2 [24], nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1) [24], nicotinamide adenine dinucleotide phosphate oxidase organizer [24], heme oxygenase 1 (HO-1) [25], myeloperoxidase (MPO) [26], metallothionein I + II [11], peroxiredoxins (PRX) 2 [23] and 5 [27], endoplasmic reticulum stress-related signaling pathway molecules [28], transcription factor NF-E2-related factor 2 (Nrf2) [29], DJ-1 protein [29], genes involved in mitochondrial protein synthesis (MRPL18, 14, 23; MRPS15, 22) [24], genes involved in adenine nucleotide translocation (SLC25A4) [24], and genes induced by oxidative stress and involved in the oxidative stress defense (UCP3, GRPEL1, TXNRD2, ISCU, AASS, ACADL, DMGDH, and CADS) [24], in brain MS lesions in comparison to control brains. On the other hand, peptidases of the 20S and 26 proteasomes [31], regulatory caps 11S α and 19S [31], and nuclear-encoded genes of the respiratory chain [24], mitochondrial DNA-encoded gene rays (ND1, ND2, ND3, ND5, ND6, COX1, and CYTB) [24], were found to be decreased or downregulated when compared to controls. Finally, brain concentrations of 20S proteasome α, β1, β2, and β5 subunits [31], calpain [31], cathepsin B [31], and mitochondrial LonP [31] have been reported to be similar in MS patients compared to controls.

2.2. Oxidative Stress Markers in Cerebrospinal Fluid (CSF)

Table S1 summarizes the results of studies related to markers of oxidative stress in the CSF from MS patients compared to controls [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67]. Most of these studies found increased CSF levels of markers of lipid peroxidation, such as malonyl-dialdehyde, hydroxyalkenals, diene conjugates, 4-hydroxy-nonenal, oxidized phosphatidylcholine, and isoprostanes in patients diagnosed with MS compared to controls [13,32,33,34,35,37,38,39,40,45,46,47], with some exceptions [36]. CSF levels of certain prostaglandins were increased in patients with MS in most studies [35,41,42,43,44], but were similar to controls in others [36,37,38].
CSF protein carbonyl concentrations were increased in MS patients compared to controls in two studies [40,47], and were similar to controls in another [35]. Protein-linked neuroketals were found to be increased [35], and advanced glycoxidation end products were similar to controls [48], respectively, in two studies. Advanced oxidation protein products were increased in the CSF of MS patients compared to controls [49] and were similar for the different clinical subtypes of MS [50]. CSF levels of markers of DNA damage were increased in MS patients compared to controls [38,50]. Nitrotyrosine (a marker of nitrosative stress) was also increased in MS patients compared to controls [36].
CSF iron levels were reported to be similar in MS patients and controls [51], and transferrin was found to be decreased in MS patients with a shorter MS duration [52]. CSF concentrations of copper [51,53], ceruloplasmin [53], and matrix-metalloproteinase 9 (MMP9) [57] were increased, while ferric-reducing antioxidant power [50] and ferroxidase activity [54] were described as being similar in MS patients compared with controls. The total antioxidant status or capacity (TAS or TAC) were decreased in MS (especially in RRMS) patients compared to controls in three studies [37,43,56], increased in one study [34], and similar in another [37]. The CSF total thiol (SH) groups were decreased in MS patients [33,49]. GPx [32] and glutathione-reductase (GSSG-R) were found to be increased and aryl esterase activity was similar [58] in comparison with controls in isolated studies.
The CSF concentrations of several antioxidants, such as ascorbate [51] and the antiaging antioxidant protein Klotho [55], were decreased in MS patients. CSF levels of alpha-tocopherol were similar in MS patients and controls [59]. Two studies measuring CSF uric acid concentrations found increased levels of this antioxidant and its precursors hypoxanthine and xanthine in MS patients compared to controls [63,64], while another study described similar values of uric acid and its metabolite allantoine in MS and controls [62].
CSF levels of the excitatory amino acid L-glutamate were decreased in MS patients compared to controls [60]. CSF levels of nitric oxide (NO) metabolites were reported to be increased in MS patients compared to controls in three studies [36,42,57], and similar to those of controls in another [61]. CSF levels of trace metals involved in oxidative stress processes were the subject of a single study, which described increased lead, decreased magnesium, and similar calcium, manganese, and zinc levels in patients with primary progressive MS (PPMS) compared to those with secondary progressive MS (SPMS) and controls [51]. The CSF human serum albumin (HAS), mercaptoalbumin (HMA), and non-mercaptoalbumins 1 and 2 (HNA1 and HNA2) [65] levels were reported to be similar in MS patients and controls. CSF neutrophil gelatinase-associated lipocalin (NGAL) was increased [39] or similar [38] compared to controls in two studies by the same group. Finally, DJ-1 [66], periredoxins 2 (PRX2) [38,39], and β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) [67] were increased in the CSF of patients with MS compared to controls.

2.3. Oxidative Stress Markers in Blood Cells

The results of studies addressing concentrations of oxidative stress markers in blood cells (erythrocytes, leukocytes, peripheral blood mononuclear cells/lymphocytes, and platelets) [68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84] are summarized in Table S1. MDA/TBA/TBARS levels were described as increased in erythrocytes [68] and leukocytes [72] from patients diagnosed with MS compared with controls, and were higher in patients with a more severe disease [72]. Radical oxygen species production was increased as well in platelets from patients with SPMS compared to controls [83]. However, other lipid peroxidation markers, such as diene conjugate and fatty acid patterns of phospholipids, were reported as being similar in MS patients and controls in erythrocytes [69] and leukocytes [69,73,74]. Advanced oxidation protein products were increased in erythrocytes from patients with CIS and RRSS (being even higher in RRSS and patients with a more severe disease) [68], and in platelets from SPMS patients (higher in patients with a more severe disease) [83] in comparison with controls, 3-nitrotyrosine was found to be increased in platelets from SPMS patients compared to controls [83], and DNA damage was increased in leukocytes from patients with RRMS [75] and in PBMC from patients with MS compared to controls [81]. Global mitochondrial activity PBMC levels were similar in MS patients and controls in one study [80], while another study found a significant increase in mitochondrial respiratory chain complexes I, II, III, IV, and V [78], and another found a decreased complex IV in MS patients compared to controls [82].
Regarding antioxidant enzyme activities or levels of antioxidant substances, in patients with MS compared to controls:
  • SOD activity was decreased in erythrocytes [68,70], leukocytes [70], and peripheral blood mononuclear cells (PBMC) [77]. Related to this finding, superoxide anion (O2) production in MS patients was increased in PBMC [78] and in platelets [73], but was similar in leukocytes [76] from MS patients compared to controls.
  • GPx activity was decreased in erythrocytes [69] and decreased [72] or similar [69] in leukocytes.
  • Catalase activity was decreased [71] or similar [69] in erythrocytes, similar in leukocytes [69], and increased in PBMC [77].
  • Myeloperoxidase activity was similar [76].
  • Reduced (GSH) and oxidized glutathione (GSSG) concentrations were reported as being similar in erythrocytes [69] and leukocytes [69,72].
  • Coenzyme Q10 concentrations were similar in erythrocytes [69] and decreased in leukocytes [69], and alpha-tocopherol levels were similar in erythrocytes [69] and leukocytes [69].
  • The TAS was similar [73,74] and total antiradical activity was increased [73,74] in leukocytes.
  • Thiol group concentrations were decreased [83], and NADPH oxidase (NOX-1), cytochrome c oxidase subunit 1 expression and glyceraldehyde 3-phosphate dehydrogenase activity (GADPH) were increased in platelets [83].
Finally, HO-1 [40], HsC70 [40], Hsp72 [40], and Trx concentrations [40], polyADP ribose (PAR) synthesis [81], and polyADP ribose polymerase-1 (PARP1) expression [81] were increased, HO-2 [40] and Hsp70-2 concentrations [80] were similar, and sirtuin 1 concentrations were increased [40] or similar [77] in PBMC from patients with MS compared with controls.

2.4. Oxidative Stress Markers in Serum/Plasma

The results of studies addressing serum and/or plasmatic levels of oxidative stress markers in patients diagnosed with MS compared to controls [33,34,36,40,44,48,49,50,51,52,53,54,56,58,59,62,63,64,65,66,69,72,73,75,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161] are summarized in Table S1.
Regarding lipid peroxidation markers, serum/plasma concentrations of MDA/TBA/TBARS were increased in MS patients compared to controls in 16 studies [33,64,72,75,85,86,87,88,90,91,92,93,94,96,97,98], but were similar in 4 studies [36,89,95,100] and decreased in 2 other studies [34,99]. Serum levels of MDA + hydroxyalkenals [101], “lipid oxidizability” [102], lipid peroxides [103], and fluorescent lipid peroxidation products [104], and lipid hydroperoxides [105,106,107,108] were increased in MS patients in most studies, and similar in others [89,103]. Serum tert-butyl hydroperoxide and hydroperoxide concentrations were reported to be similar [110,111,112,115] or increased in MS patients [109,114]. Serum HNE [40] and fatty acid patterns of phospholipids [69] levels were increased in MS patients compared to controls, while prostaglandin-F2alpha and F2-isoprostane were reported to be increased [36,116] or decreased [44].
Serum/plasma levels of protein carbonyls were described as increased in MS or RRMS patients compared to controls in nine studies [40,75,92,96,109,117,118,119,120], and similar between two groups in three other studies [108,110,115]. Advanced glycoxidation end products were normal [48,91] or increased [120], and advanced oxidation protein products were increased in MS according to eight studies [49,106,112,115,120,121,122,124], with this increase being more marked in patients with a more severe disease [115], higher ferritin levels [110], and with a trend towards a decrease during follow-up [123]. Other authors reported similar serum/plasma levels of advanced oxidation protein products in MS patients and controls (with a trend towards lower values in patients with low vitamin D levels) [111] and a lack in differences among the different evolutive types of MS [50]. Serum fructosamine levels were reported as increased in MS patients in a single study [91]. Serum/plasma levels of markers of oxidative stress/damage of DNA, such as 8-OHdG [105,118,125] and DNA single-strand breaks [99], were increased in MS patients.
Serum/plasma ferric-reducing antioxidant power (FRAP) and total-reducing antioxidant power (TRAP) were decreased [93,108,110,111,122,127] or similar [50,91,124] in MS patients compared to controls, and were similar in MS patients with different degrees of severity [107]. In comparison to controls, MS patients showed decreased [129,132,133] or similar [51,110,114,128,131,134] serum/plasma iron levels, increased ferritin levels [107,108,110,133] or those similar to controls [110], decreased [52,97] or similar [114,128] transferrin levels, higher soluble transferrin receptor levels [118,131], and similar lactoferrin levels to controls [135]. The serum/plasma total ferroxidase activity was decreased [56,136], ceruloplasmin increased [114,119,137], and copper increased [53,114,132] or similar [51,129,134] in MS compared to controls.
Serum/plasma TAS/TAC or total antiradical activity were reported to be decreased [34,77,78,87,91,94,96,102,109,114,118,130,132,138,139,141] or similar [56,73,85,89,95,97,100,126,142], TOS increased [87,97,103,118,126,129,130,132,139,140,143] or similar [103,129,140], and oxidative stress index (OSI) increased [87,97,126,139] or similar [103] in patients with MS compared to controls.
Serum/plasma total thiol group concentrations were decreased in MS patients compared to controls in most studies [33,49,75,96,109,112,122,143], with one exception [144], and were similar in patients with MS with low vs. those with high ferritin levels. Native thiol levels were found to be decreased in one study [143] and similar to those of controls in another [144]. In comparison to controls, serum plasma SOD activity from MS patients was reported to be similar [90,100,103,146], increased in MS patients [86,97,118,145], or decreased in MS patients [75,143]; total glutathione was similar [89] or increased in MS patients [118]; GSH was similar [69,89,100] or increased in MS patients [118]: GSSG was similar [68] or decreased in MS patients [118]; GSSG-reductase activity was decreased in MS patients [118,146]; GPx activity was similar [69,100,146], increased in MS patients [101], or decreased in MS patients [103,118,135]; catalase activity was increased [75,97,103] or similar [69,100]; and GST was similar [146] or decreased in MS patients [118].
Serum/plasma paraoxonase (PON1) activity was decreased in most studies in patients with MS [58,105,127,146], while others reported an increase [148] or similarity to controls [93]. Arylesterase activity in MS patients was similar to controls in three studies [123,139,146] and decreased in another [58].
Serum/plasma concentrations of coenzyme Q10 were reported as being decreased in MS patients compared to controls [64,69,89] or similar in MS patients and controls [151]. Similarly, four studies showed lower serum/plasma alpha-tocopherol levels in MS patients [59,69,72,75] and another showed non-significant differences when compared to controls [69]. In comparison with controls, serum/plasma gamma-tocopherol levels were decreased in MS patients in a single study [64], beta-carotene was decreased in two studies [72,152] and similar in one [64], and ascorbic acid was decreased in two studies [75,86] and similar in another two [51,72].
Many studies have addressed serum/plasma levels of NO metabolites (nitrates + nitrites) in MS patients and controls; seven showed a significant decrease [36,87,107,108,109,110,111], one showed a non-significant trend towards a decrease [61], and five others reported a significant increase [86,99,101,103,112] of these parameters in MS patients. Serum nitrotyrosine levels were increased in MS patients compared to controls [36,92,117].
Several studies have addressed serum/plasma trace metal concentrations in MS patients and controls, described in detail in Table S1, with varying results. The most consistent findings were increased cadmium [129,132,154,155], aluminum [129,132,134], molybdenum [129,132,134], tin [129,132], zirconium [129,132], and arsenic levels in MS patients [88,93,134,155], and a lack in differences in serum/plasma levels of lead [129,132,154,155], mercury [129,132], strontium [129,130,132], vanadium [129,132,134], and wolframium [129,132].
Serum/plasma levels of uric acid were decreased in patients with MS compared to controls in three studies [97,158,159], increased in another two [63,86], and similar to controls in two more [109,156], while the serum levels of the related substances hypoxanthine, xanthine, and uridine were found to be increased [158] and allantoine was similar in MS patients compared to controls. Serum/plasma uric acid levels from MS patients were decreased in current smokers compared with non-smokers and ex-smokers [160]. Serum/plasma bilirubin levels were decreased in MS patients [97,157]. Ischemia-modified albumin was increased [140] and irisin and nesfatin-1 were decreased in MS patients compared to controls [161]. Finally, serum/plasma levels of HAS [65], HMA [65], HNA1 [65], HNA2 [65], and DJ-1 [66] from MS patients did not differ significantly from those of controls.

2.5. Oxidative Stress Markers in Other Fluids

Several studies found increased levels of lipid peroxidation markers [85,162], increased levels of aluminum [162], decreased levels of silicon [162], increased 8-iso-prostaglandin (PG-)F2alpha levels [163], neopterin/creatinine ratio [164], and prolyl oligopeptidase levels [165] and decreased levels of alpha2-macroglobulin [165] in urine from MS patients compared to controls (Table S1).
Karlík et al. [91] described an increase in salivary levels of TBA/TBARS and advanced glycation end-products, decreased FRAP, and similar advanced oxidation protein products and TAS in patients with MS compared to controls (Table S1).

3. Genetic Variants of Genes Related to Oxidative Stress in Patients with Multiple Sclerosis

The possible association between single nucleotide polymorphisms (SNPs) or deletions in genes related to oxidative stress and the risk of developing MS has been the subject of several case–control association studies, which are summarized in Table S2 [113,155,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180]. Most of these studies found a lack of a direct association between these SNPs and multiple sclerosis, including the most common SNPs in CYP2D6 [166], GSTP1 [167,168,169], GSTM1 [113,155,167,170], GSST1 [167,170], GSTM3 [167], PON1 rs662 [173,174], PON1 rs854560 [173], NQO1 rs1800556 [176], HMOX1 2071747 [177], HMOX2 rs270363 [177], HMOX2 rs1051308 [177], NCF1 D7S2518 [178], NCF2 [178], NCF4 [178], CYBA [178], CYBB rs9330580 [178], NOS1 rs1879417 [179], NOS3 rs2070744 [180], and HNF1A-AS1 rs7953249 [181] genes.
Other authors reported a significant association for GSTM1 null polymorphism [171], GSTT1 null polymorphism [171], MPO rs2333227 [172], PON1 rs854560 [174], GLO1 rs1049346 [173], NQO1 rs1800566 [169,175], OGG1 rs1052133 [113], NCF1 D7S1870 [178], CYBB rs5963310 [178], NOS2 rs2297518 [179], CAT rs7943316 [179], and TRPP2-AS rs933151 [181] with the risk of developing MS, and a decreased risk related with SOD2 rs187947 [179] and GPX4 rs713041 [179]. Mann et al. [167] described an association between the combination of GSTM1 null polymorphism and GSTP1 rs1695 alleles and the presence of GSTM3 rs1799735 with severe disability in patients with an MS duration longer than 10 years [167]. Alexoudi et al. [169] described an interaction between GSTP1 rs1695 and NQO1 rs1800665 and the risk for MS. Finally, Agúndez et al. [178] described an association of the HMOX2 rs1051308AA genotype and rs1051308 with risk for MS in males.

4. Data from Experimental Models of Multiple Sclerosis

4.1. Lipid Peroxidation Markers

Perianes-Cachero et al. [182] described an increase in lipid peroxidation, and in SOD, GPx, GSSG-reductase activities (assessed using spectrophotometry), and a decrease in catalase activity (assessed using spectrophotometry) and GSH concentrations (assessed using a fluorometric method) in the hippocampus of 6-week-old female Lewis rats with chronic relapsing experimental autoimmune encephalomyelitis (EAE), the most important animal model of MS.
Dimitrijević et al. [183] reported an increased MDA (assessed using a colorimetric method) and superoxide anion levels, decreased GSH concentrations, decreased SOD activity (assessed using spectrophotometry), increased NOS3 and xanthine oxidase (an enzyme responsible for the synthesis of uric acid) expression (assessed using quantitative real time-polymerase chain reaction—qRT-PCR), and increased AOPP in the spinal cord from Dark Agouti rats with EAE, and increased plasma AOPP levels (assessed using high-performance size-exclusion matrix chromatography) in the same MS model.
Jhelum et al. [184] reported increased peroxidation, increased mRNA levels of several ferroptosis genes, increased nuclear receptor coactivator 4 (NCOA4) expression (assessed using qRT-PCR), decreased GPx4 activity (assessed using Western blot analysis), and decreased total glutathione (assessed using spectrophotometry) in the brain of female mice with EAE of C57BL/6. C57BL/6 OlaHSD [15] and C57BL/6 female mice with EAE [185] showed increased levels of acrolein or their metabolites (assessed using Western blot [15] or liquid chromatography/tandem mass spectrometry [185]) in the spinal cord and urine. A decrease in mRNA expression was described for the cytoplasmic isoform of GPx4 (assessed using RT-PCR) in the spinal cord from female C57BL/6 mice with EAE [186]. Smerjac and Bizzozzero [187] described increased lipid peroxidation markers (assessed by a colorimetric method) before the appearance of neurological symptoms, in the spinal cord of seven-week-old male Lewis rats with acute EAE.

4.2. Protein Oxidation Markers

Smerjac and Bizzozzero [187] described increased protein carbonylation and degradation (assessed using Western blotting) at the time of maximal clinical disability and a decreased glutathione concentration (assessed using spectrophotometry) in the spinal cord of seven-week-old male Lewis rats with acute EAE. The same group described an increased protein carbonylation (using the OxyBlot™ protein oxidation detection kit) within cerebellar astrocytes, which was maximal in the acute phase and decreased in the chronic phase of the disease [188], and in the spinal cord [189] of eight-week-old female C57BL/6 mice with EAE.
Castegna et al. [190] described an increased protein oxidation (assessed with OxyBlot and mass spectrometry analyses), increased glutamate/glutamine, and decreased natural antioxidant levels (assessed with liquid chromatography-tandem mass spectrometry analysis, LC-MS/MS), which paralleled disease activity in the brain of female 10–11-week-old PLSJL mice with EAE.

4.3. Heme Oxygenase 1 (HO-1)

Several authors have reported the increased expression of heme oxygenase 1 (HO-1) by using the Western blot analysis [184,191,192,193] and decreased expression of NADPH cytochrome P450 reductase (which is required for the catalytic activity of HO-1) expression [191] in the brain of female C57BL/6 mice [184], female SJL mice [191], pregnant Sprague-Dawley rats [192], and adult male Lewis rats [193] with EAE. An increase in HO-1 expression in the EAE brain was inhibited and exacerbated, respectively, through the coadministration of inducers of inhibitors of HO-1 [193]. HO-1 was increased in the spinal cord of the EAE rodents [192]. In addition, a significant upregulation in HO-1 and the iron storage protein ferritin (assessed with RT-PCR) in a demyelination model of mutant rats (dmv rats) was described in comparison with a hypomyelination model (mv rats) [194].

4.4. NAD(P)H Oxidases (NOX)

NAD(P)H oxidase (NOX) enzymes (assessed using a fluorescence lifetime imaging method with a two-photon laser-scanning microscope) were also found to be overactivated in inflammatory monocytes, activated microglia, and astrocytes of the brain and peripheral CD11b(+) cells of CerTN L15 x LysM tdRFP mice with EAE [195]. In addition, NOX2 deletion or deficiency could prevent EAE induction in female C57BL/6 gp91phox−/− [Nox2 KO mice (gp91Cybbtm1Din/J)] mice (assessment performed using qRT-PCR) [196,197].

4.5. Other Markers

Hasseldam et al. [198] reported a loss in mitochondrial membrane potential and oxidative changes (assessed using a spectrophotometric method) in the brains of female Dark Agouti rats with EAE, present approximately 10 days before clinical onset. Other authors, using two-photon imaging, described increased mitochondrial oxidation in oligodendrocytes from MOG-cre mice, CCR2-RFP x CX3CR1-GFP mice, mito-roGFP2-Orp1 mice, and Ai14 reporter mice with EAE [199].
Aheng et al. [200] described a higher severity of EAE in a model of mice lacking the inducible nitric oxide synthase (iNOS) gene, while mice lacking simultaneously uncoupling protein 2 (UCP2) and iNOS genes developed milder EAE.
Johnson et al. [201] described a relationship between the deficiency of nuclear-factor-erythroid-2-related factor 2 (Nrf2) (assessed using qPCR) and a more severe clinical course, a more rapid onset, and a greater percentage of Biozzi ABH mice back-crossed onto Nrf2-KO mice developing EAE after the administration of myelin oligodendrocyte glycoprotein (MOG 35-55), suggesting a role of Nrf2 in modulating the neuroinflammatory response.
Honorat et al. [202] reported an increased expression of xanthine oxidase (assessed using a fluorometric assay) in infiltrating macrophages and microglia of the spinal cord from 8-week-old female SJL/J mice with EAE, which decreased with the preadministration of a potent xanthine oxidase inhibitor, thereby, suggesting a role of xanthine oxidase in the pathogenesis of EAE.
Metabolomic studies in plasma (using ultra-high-performance liquid chromatography-orbitrap-mass spectrometry—UHPLC-Orbitrap-MS) from C57BL/6J EAE mice showed a downregulation in glycerophospholipids and fatty acyls and upregulation in glycolipids, taurine-conjugated bile acids, and sphingolipids, and an increase in NOX activity and MMP9 during disease progression [203]. Increased superoxide anion concentrations and upregulation in NOS3 in the pituitary and adrenal glands were reported, as well as an increase in MDA and GSH levels and in catalase activity (assessed using electron paramagnetic resonance spectroscopy) in adrenal glands of 2-month-old female rats of Dark Agouti with EAE [204]. Plasma concentrations of IgG antibodies with peroxidase [205], oxidoreductase [205], and catalase [206] activities (assessed using spectrophotometry) were increased at different stages of EAE in C57BL/6, Th, 2D2 mice [205], and C57BL/6 mice [206].

4.6. Effects of Exposure of Neuronal Cultures to Pathological Products of MS Patients

Vidaurre et al. [207], in a study comparing 13 MS patients with 10 HC, showed that acute exposure of culture neurons to the CSF from MS patients induced oxidative stress and decreased the expression of neuroprotective genes (assessed using RT-PCR and a quantitative lipidomic analysis), which was attributed to an increased content of the ceramides C16:0 and C24:0. Finally, the injection of cultured neurons and oligodendrocytes killed by oxidized phosphatidylcholines obtained from MS lesions induced focal demyelinating lesions with prominent axonal loss in the spinal cord of mice [14].

5. Discussion and Conclusions

Many findings point to the possible role of oxidative stress in the pathogenesis of MS. This is supported by a demonstrated increase in markers of oxidation of lipids, proteins, and DNA, and markers of nitrosative stress, together with changes in the activity of enzymes and proteins involved in oxidative stress, both in the brain and/or the spinal cord of MS patients (in samples from autopsies) and in experimental models of MS, mainly in different strains of rodents with EAE (although data from these experimental models could have a low predictive value). Most studies on CSF and blood cells have also shown an increase in several markers of oxidative stress and a decrease in several antioxidant substances in MS patients compared to controls. However, a recent meta-analysis of oxidative stress markers in CSF confirmed only a significant association of CSF MDA levels, but not of other potential markers, with MS [208]. Even though studies of concentrations of markers of oxidative stress in serum/plasma, urine, and other tissues have not shown conclusive results, most studies and the results of a meta-analysis showed an increase in serum/plasma MDA and albumin concentrations in patients diagnosed with MS compared to controls [208]. Similarly, the majority of studies showed a decrease in serum/plasma TAS/TAC and serum/plasma levels of SH groups and an increase in TOS and OSI in patients with MS.
In summary, the main alterations found in studies addressing oxidative stress markers in patients with MS included the following;
  • Increased markers for lipid peroxidation, protein oxidation, and DNA oxidation.
  • Increased mitochondrial activity.
  • Increased NO nitrotyrosine (therefore, increased nitrosative stress).
  • Increased TOS and OSI and decreased TAS.
  • Decreased iron, copper, and ceruloplasmin.
  • Increased SOD and catalase activities.
  • Decreased GSH levels and normal or decreased GPx activity.
  • Increased NQO1, NOX1, NOX2, and HO-1 activities.
  • Increased myeloperoxidase and peroxiredoxins 1 and 2 activities.
  • Increased endoplasmic reticulum stress proteins.
  • Decreased concentrations of uric acid and related substances.
  • Decreased ascorbate concentrations.
To date, none of the studied variants in genes related to oxidative stress have shown an unequivocal association with MS. Potential reasons for the controversies seen between the results of various studies, both in those addressing biochemical parameters and studies on genes related to oxidative stress, include sample size, differences in sample collection and the methods used, and possibly factors associated with the participants (for example, some studies were not matched by age and/or sex or treatment with disease-modifying therapies). Moreover, in the case of genetic case–control association studies, there was a lack of replication studies.

6. Future Directions

We suggest that future studies aiming to establish the possible role of oxidative stress in the pathogenesis of MS should fulfill, at least, the following conditions:
  • Design prospective and multicenter studies with a long-term follow-up period (1 year).
  • The recruitment of a large number of patients diagnosed with MS according to standardized criteria [209], not exposed to any therapy for this disease, and a similar number of age- and sex-matched healthy controls who do not fulfill clinical criteria for the diagnosis of MS and without a family history of MS.
  • Both MS patients and controls involved in such studies should not have obesity or undernutrition, should not suffer from oncologic, acute infectious diseases, kidney, liver, thyroid, or parathyroid disease, have no recent history of traumatism or surgery, and no atypical dietary habits (i.e., diets consisting exclusively of one type of foodstuffs, such as vegetables, and others). They should not use therapy with steroids, diuretics, diphosphonates vitamins, calcium or mineral supplements, or drugs that could affect oxidative stress. In addition, pregnant women should be excluded.
  • It would be desirable to collect plasma/serum and blood cells for the analysis of multiple oxidative stress biomarkers and to obtain blood DNA for genetic studies of genes related to oxidative stress, both in MS patients and controls, at baseline.
  • Patients with MS should undergo periodic clinical evaluations every 3–4 months to evaluate the evolutive type and severity of the disease according to standardized scales such as the EDSS [210].
  • A new collection of plasma/serum and blood cells should be performed for the analysis of multiple oxidative stress biomarkers at the end of the follow-up to evaluate the changes induced by the different treatments used for MS.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25126289/s1.

Author Contributions

F.J.J.-J.: Conceptualization, Methodology, Investigation, Validation, Formal Analysis, Investigation; Writing—Original Draft; Writing—Review and Editing; Project Administration. H.A.-N.: Conceptualization, Methodology, Investigation, Validation, Formal Analysis, Investigation; Writing—Original Draft; Writing—Review and Editing; Project Administration. P.S.-C.: Conceptualization, Methodology, Investigation, Validation, Formal Analysis, Investigation; Writing—Original Draft; Writing—Review and Editing; Project Administration. E.G.-M.: Conceptualization, Methodology, Investigation, Validation, Formal Analysis, Investigation; Writing—Original Draft; Writing—Review and Editing, Project Administration, Obtaining Funding. J.A.G.A.: Conceptualization, Methodology, Investigation, Validation, Formal Analysis, Investigation; Writing—Original Draft; Writing—Review and Editing, Project Administration, Obtaining Funding. All authors have read and agreed to the published version of the manuscript.

Funding

The work at the authors’ laboratory was supported in part by grants PI18/00540 and PI21/01683 from Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Madrid, Spain, and IB20134 and GR21073 from Junta de Extremadura, Mérida, Spain. Partially funded with FEDER funds.

Acknowledgments

We recognize the effort of the personnel of the Library of Hospital Universitario del Sureste, Arganda del Rey, who retrieved an important number of papers for us. James McCue, a native English speaker with expertise in editing scientific articles, extensively revised the English text.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Patsopoulos, N.A.; De Jager, P.-L. Genetic and gene expression signatures in multiple sclerosis. Mult. Scler. 2020, 26, 576–581. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, W.; Patsopoulos, N.A. Genetics and functional genomics of multiple sclerosis. Semin. Immunopathol. 2022, 44, 63–79. [Google Scholar] [CrossRef] [PubMed]
  3. Mechelli, R.; Umeton, R.; Manfrè, G.; Romano, S.; Buscarinu, M.C.; Rinaldi, V.; Bellucc, I.G.; Bigi, R.; Ferraldeschi, M.; Salvetti, M.; et al. Reworking GWAS Data to Understand the Role of Nongenetic Factors in MS Etiopathogenesis. Genes 2020, 11, 97. [Google Scholar] [CrossRef] [PubMed]
  4. Zarghami, A.; Li, Y.; Claflin, S.B.; van der Mei, I.; Taylor, B.V. Role of environmental factors in multiple sclerosis. Expert Rev. Neurother. 2021, 21, 1389–1408. [Google Scholar] [CrossRef] [PubMed]
  5. Waubant, E.; Lucas, R.; Mowry, E.; Graves, J.; Olsson, T.; Alfredsson, L.; Langer-Gould, A. Environmental and genetic risk factors for MS: An integrated review. Ann. Clin. Transl. Neurol. 2019, 6, 1905–1922. [Google Scholar] [CrossRef] [PubMed]
  6. McGarry, T.; Biniecka, M.; Veale, D.J.; Fearo, N.U. Hypoxia, oxidative stress and inflammation. Free Radic. Biol. Med. 2018, 125, 15–24. [Google Scholar] [CrossRef] [PubMed]
  7. Gambini, J.; Stromsnes, K. Oxidative Stress and Inflammation, From Mechanisms to Therapeutic Approaches. Biomedicines 2022, 10, 753. [Google Scholar] [CrossRef] [PubMed]
  8. Penkowa, M.; Espejo, C.; Ortega-Aznar, A.; Hidalgo, J.; Montalban, X.; Martínez Cáceres, E.M. Metallothionein expression in the central nervous system of multiple sclerosis patients. Cell. Mol. Life Sci. 2003, 60, 1258–1266. [Google Scholar] [CrossRef] [PubMed]
  9. Haider, L.; Fischer, M.T.; Frischer, J.M.; Bauer, J.; Höftberger, R.; Botond, G.; Esterbauer, H.; Binder, C.J.; Witztum, J.L.; Lassmann, H. Oxidative damage in multiple sclerosis lesions. Brain 2011, 134, 1914–1924. [Google Scholar] [CrossRef]
  10. Kemp, K.; Redondo, J.; Hares, K.; Rice, C.; Scolding, N.; Wilkins, A. Oxidative injury in multiple sclerosis cerebellar grey matter. Brain Res. 2016, 1642, 452–460. [Google Scholar] [CrossRef]
  11. van Horssen, J.; Schreibelt, G.; Drexhage, J.; Hazes, T.; Dijkstra, C.D.; van der Valk, P.; de Vries, H.E. Severe oxidative damage in multiple sclerosis lesions coincides with enhanced antioxidant enzyme expression. Free Radic. Biol. Med. 2008, 45, 1729–1737. [Google Scholar] [CrossRef] [PubMed]
  12. Haider, L.; Simeonidou, C.; Steinberger, G.; Hametner, S.; Grigoriadis, N.; Deretzi, G.; Kovacs, G.G.; Kutzelnigg, A.; Lassmann, H.; Frischer, J.-M. Multiple sclerosis deep grey matter, the relation between demyelination, neurodegeneration, inflammation and iron. J. Neurol. Neurosurg. Psychiatry 2014, 85, 1386–1395. [Google Scholar] [CrossRef] [PubMed]
  13. Qin, J.; Goswami, R.; Balabanov, R.; Dawson, G. Oxidized phosphatidylcholine is a marker for neuroinflammation in multiple sclerosis brain. J. Neurosci. Res. 2007, 85, 977–984. [Google Scholar] [CrossRef]
  14. Dong, Y.; D’Mello, C.; Pinsky, W.; Lozinski, B.M.; Kaushik, D.K.; Ghorbani, S.; Moezzi, D.; Brown, D.; Melo, F.C.; Zandee, S.; et al. Oxidized phosphatidylcholines found in multiple sclerosis lesions mediate neurodegeneration and are neutralized by microglia. Nat. Neurosci. 2021, 24, 489–503. [Google Scholar] [CrossRef]
  15. Spaas, J.; Franssen, W.M.A.; Keytsman, C.; Blancquaert, L.; Vanmierlo, T.; Bogie, J.; Broux, B.; Hellings, N.; van Horssen, J.; Posa, D.K.; et al. Carnosine quenches the reactive carbonyl acrolein in the central nervous system and attenuates autoimmune neuroinflammation. J. Neuroinflamm. 2021, 18, 255. [Google Scholar] [CrossRef]
  16. Bizzozero, O.A.; DeJesus, G.; Callahan, K.; Pastuszyn, A. Elevated protein carbonylation in the brain white matter and gray matter of patients with multiple sclerosis. J. Neurosci. Res. 2005, 81, 687–695. [Google Scholar] [CrossRef]
  17. Vladimirova, O.; O’Connor, J.; Cahill, A.; Alder, H.; Butunoi, C.; Kalman, B. Oxidative damage to DNA in plaques of MS brains. Mult. Scler. 1998, 4, 413–418. [Google Scholar] [CrossRef] [PubMed]
  18. Witte, M.E.; Bø, L.; Rodenburg, R.J.; Belien, J.A.; Musters, R.; Hazes, T.; Wintjes, L.T.; Smeitink, J.A.; Geurts, J.J.; De Vries, H.E.; et al. Enhanced number and activity of mitochondria in multiple sclerosis lesions. J. Pathol. 2009, 219, 193–204. [Google Scholar] [CrossRef] [PubMed]
  19. Choi, I.Y.; Lee, S.P.; Denney, D.R.; Lynch, S.G. Lower levels of glutathione in the brains of secondary progressive multiple sclerosis patients measured by 1H magnetic resonance chemical shift imaging at 3 T. Mult. Scler. 2011, 17, 289–296. [Google Scholar] [CrossRef]
  20. Choi, I.Y.; Lee, P.; Hughes, A.J.; Denney, D.R.; Lynch, S.G. Longitudinal changes of cerebral glutathione (GSH) levels associated with the clinical course of disease progression in patients with secondary progressive multiple sclerosis. Mult. Scler. 2017, 23, 956–962. [Google Scholar] [CrossRef]
  21. Choi, I.Y.; Lee, P.; Adany, P.; Hughes, A.J.; Belliston, S.; Denney, D.R.; Lynch, S.G. In vivo evidence of oxidative stress in brains of patients with progressive multiple sclerosis. Mult. Scler. 2018, 24, 1029–1038. [Google Scholar] [CrossRef]
  22. van Horssen, J.; Schreibelt, G.; Bö, L.; Montagne, L.; Drukarch, B.; van Muiswinkel, F.L.; de Vries, H.E. NAD(P)H,quinone oxidoreductase 1 expression in multiple sclerosis lesions. Free Radic. Biol. Med. 2006, 41, 311–317. [Google Scholar] [CrossRef] [PubMed]
  23. Voigt, D.; Scheidt, U.; Derfuss, T.; Brück, W.; Junker, A. Expression of the Antioxidative Enzyme Peroxiredoxin 2 in Multiple Sclerosis Lesions in Relation to Inflammation. Int. J. Mol. Sci. 2017, 18, 760. [Google Scholar] [CrossRef] [PubMed]
  24. Fischer, M.T.; Sharma, R.; Lim, J.L.; Haider, L.; Frischer, J.M.; Drexhage, J.; Mahad, D.; Bradl, M.; van Horssen, J.; Lassmann, H. NADPH oxidase expression in active multiple sclerosis lesions in relation to oxidative tissue damage and mitochondrial injury. Brain 2012, 135, 886–899. [Google Scholar] [CrossRef] [PubMed]
  25. Stahnke, T.; Stadelmann, C.; Netzler, A.; Brück, W.; Richter-Landsberg, C. Differential upregulation of heme oxygenase-1 (HSP32) in glial cells after oxidative stress and in demyelinating disorders. J. Mol. Neurosci. 2007, 32, 25–37. [Google Scholar] [CrossRef] [PubMed]
  26. Gray, E.; Thomas, T.L.; Betmouni, S.; Scolding, N.; Love, S. Elevated activity and microglial expression of myeloperoxidase in demyelinated cerebral cortex in multiple sclerosis. Brain Pathol. 2008, 18, 86–95. [Google Scholar] [CrossRef]
  27. Holley, J.E.; Newcombe, J.; Winyard, P.G.; Gutowski, N.J. Peroxiredoxin V in multiple sclerosis lesions, predominant expression by astrocytes. Mult. Scler. 2007, 13, 955–961. [Google Scholar] [CrossRef] [PubMed]
  28. Mháille, A.N.; McQuaid, S.; Windebank, A.; Cunnea, P.; McMahon, J.; Samali, A.; FitzGerald, U. Increased expression of endoplasmic reticulum stress-related signaling pathway molecules in multiple sclerosis lesions. J. Neuropathol. Exp. Neurol. 2008, 67, 200–211. [Google Scholar] [CrossRef]
  29. van Horssen, J.; Drexhage, J.A.; Flor, T.; Gerritsen, W.; van der Valk, P.; de Vries, H.E. Nrf2 and DJ1 are consistently upregulated in inflammatory multiple sclerosis lesions. Free Radic. Biol. Med. 2010, 49, 1283–1289. [Google Scholar] [CrossRef]
  30. Licht-Mayer, S.; Wimmer, I.; Traffehn, S.; Metz, I.; Brück, W.; Bauer, J.; Bradl, M.; Lassmann, H. Cell type-specific Nrf2 expression in multiple sclerosis lesions. Acta Neuropathol. 2015, 130, 263–277. [Google Scholar] [CrossRef]
  31. Zheng, J.; Bizzozero, O.A. Decreased activity of the 20S proteasome in the brain white Matter and gray matter of patients with multiple sclerosis. J. Neurochem. 2011, 117, 143–153. [Google Scholar] [CrossRef] [PubMed]
  32. Calabrese, V.; Raffaele, R.; Cosentino, E.; Rizza, V. Changes in cerebrospinal fluid levels of malondialdehyde and glutathione reductase activity in multiple sclerosis. Int. J. Clin. Pharmacol. Res. 1994, 14, 119–123. [Google Scholar] [PubMed]
  33. Calabrese, V.; Bella, R.; Testa, D.; Spadaro, F.; Scrofani, A.; Rizza, V.; Pennisi, G. Increased cerebrospinal fluid and plasma levels of ultraweak chemiluminescence are associated with changes in the thiol pool and lipid-soluble fluorescence in multiple sclerosis, the pathogenic role of oxidative stress. Drugs Exp. Clin. Res. 1998, 24, 125–131. [Google Scholar] [PubMed]
  34. Ghabaee, M.; Jabedari, B.; Al-E-Eshagh, N.; Ghaffarpour, M.; Asadi, F. Serum and cerebrospinal fluid antioxidant activity and lipid peroxidation in Guillain-Barre syndrome and multiple sclerosis patients. Int. J. Neurosci. 2010, 120, 301–304. [Google Scholar] [CrossRef] [PubMed]
  35. Gonzalo, H.; Brieva, L.; Tatzber, F.; Jové, M.; Cacabelos, D.; Cassanyé, A.; Lanau-Angulo, L.; Boada, J.; Serrano, J.C.; González, C.; et al. Lipidome analysis in multiple sclerosis reveals protein lipoxidative damage as a potential pathogenic mechanism. J. Neurochem. 2012, 123, 622–634. [Google Scholar] [CrossRef] [PubMed]
  36. Seven, A.; Aslan, M.; Incir, S.; Altıntaş, A. Evaluation of oxidative and nitrosative stress in relapsing remitting multiple sclerosis, effect of corticosteroid therapy. Folia Neuropathol. 2013, 51, 58–64. [Google Scholar] [CrossRef]
  37. Bartova, R.; Petrlenicova, D.; Oresanska, K.; Prochazkova, L.; Liska, B.; Turecky, L.; Durfinova, M. Changes in levels of oxidative stress markers and some neuronal enzyme activities in cerebrospinal fluid of multiple sclerosis patients. Neuro. Endocrinol. Lett. 2016, 37, 102–106. [Google Scholar] [PubMed]
  38. Burgetova, A.; Dusek, P.; Uher, T.; Vaneckova, M.; Vejrazka, M.; Burgetova, R.; Horakova, D.; Srpova, B.; Krasensky, J.; Lambert, L. Oxidative Stress Markers in Cerebrospinal Fluid of Newly Diagnosed Multiple Sclerosis Patients and Their Link to Iron Deposition and Atrophy. Diagnostics 2022, 12, 1365. [Google Scholar] [CrossRef] [PubMed]
  39. Burgetova, A.; Dusek, P.; Uher, T.; Vaneckova, M.; Vejrazka, M.; Burgetova, R.; Horakova, D.; Srpova, B.; Kalousova, M.; Noskova, L.; et al. CSF Markers of Oxidative Stress Are Associated with Brain Atrophy and Iron Accumulation in a 2-Year Longitudinal Cohort of Early MS. Int. J. Mol. Sci. 2023, 24, 10048. [Google Scholar] [CrossRef]
  40. Pennisi, G.; Cornelius, C.; Cavallaro, M.M.; Salinaro, A.T.; Cambria, M.T.; Pennisi, M.; Bella, R.; Milone, P.; Ventimiglia, B.; Migliore, M.R.; et al. Redox regulation of cellular stress response in multiple sclerosis. Biochem. Pharmacol. 2011, 82, 1490–1499. [Google Scholar] [CrossRef]
  41. Greco, A.; Minghetti, L.; Sette, G.; Fieschi, C.; Levi, G. Cerebrospinal fluid isoprostane shows oxidative stress in patients with multiple sclerosis. Neurology 1999, 53, 1876–1879. [Google Scholar] [CrossRef]
  42. Greco, A.; Minghetti, L.; Puopolo, M.; Cannoni, S.; Romano, S.; Pozzilli, C.; Levi, G. Cerebrospinal fluid isoprostanes are not related to inflammatory activity in relapsing-remitting multiple sclerosis. J. Neurol. Sci. 2004, 224, 23–27. [Google Scholar] [CrossRef] [PubMed]
  43. Mir, F.; Lee, D.; Ray, H.; Sadiq, S.A. CSF isoprostane levels are a biomarker of oxidative stress in multiple sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2014, 1, e21. [Google Scholar] [CrossRef] [PubMed]
  44. Lam, M.A.; Maghzal, G.J.; Khademi, M.; Piehl, F.; Ratzer, R.; Romme Christensen, J.; Sellebjerg, F.T.; Olsson, T.; Stocker, R. Absence of systemic oxidative stress and increased CSF prostaglandin F2α in progressive MS. Neurol. Neuroimmunol. Neuroinflamm. 2016, 3, e256. [Google Scholar] [CrossRef] [PubMed]
  45. Mattsson, N.; Haghighi, S.; Andersen, O.; Yao, Y.; Rosengren, L.; Blennow, K.; Praticò, D.; Zetterberg, H. Elevated cerebrospinal fluid F2-isoprostane levels indicating oxidative stress in healthy siblings of multiple sclerosis patients. Neurosci. Lett. 2007, 414, 233–236. [Google Scholar] [CrossRef] [PubMed]
  46. Sbardella, E.; Greco, A.; Stromillo, M.L.; Prosperini, L.; Puopolo, M.; Cefaro, L.A.; Pantano, P.; De Stefano, N.; Minghetti, L.; Pozzilli, C. Isoprostanes in clinically isolated syndrome and early multiple sclerosis as biomarkers of tissue damage and predictors of clinical course. Mult. Scler. 2013, 19, 411–417. [Google Scholar] [CrossRef] [PubMed]
  47. Rommer, P.S.; Greilberger, J.; Salhofer-Polanyi, S.; Auff, E.; Leutmezer, F.; Herwig, R. Elevated levels of carbonyl proteins in cerebrospinal fluid of patients with neurodegenerative diseases. Tohoku J. Exp. Med. 2014, 234, 313–317. [Google Scholar] [CrossRef]
  48. Kalousová, M.; Havrdová, E.; Mrázová, K.; Spacek, P.; Braun, M.; Uhrová, J.; Germanová, A.; Zima, T. Advanced glycoxidation end products in patients with multiple sclerosis. Prague Med. Rep. 2005, 106, 167–174. [Google Scholar] [PubMed]
  49. Ljubisavljevic, S.; Stojanovic, I.; Vojinovic, S.; Stojanov, D.; Stojanovic, S.; Cvetkovic, T.; Savic, D.; Pavlovic, D. The patients with clinically isolated syndrome and relapsing remitting multiple sclerosis show different levels of advanced protein oxidation products and total thiol content in plasma and CSF. Neurochem. Int. 2013, 62, 988–997. [Google Scholar] [CrossRef]
  50. Pasquali, L.; Pecori, C.; Chico, L.; Iudice, A.; Siciliano, G.; Bonuccelli, U. Relation between plasmatic and cerebrospinal fluid oxidative stress biomarkers and intrathecal Ig synthesis in Multiple Sclerosis patients. J. Neuroimmunol. 2015, 283, 39–42. [Google Scholar] [CrossRef]
  51. Pomary, P.K.; Eichau, S.; Amigó, N.; Barrios, L.; Matesanz, F.; García-Valdecasas, M.; Hrom, I.; García Sánchez, M.I.; Garcia-Martin, M.L. Multifaceted Analysis of Cerebrospinal Fluid and Serum from Progressive Multiple Sclerosis Patients, Potential Role of Vitamin C and Metal Ion Imbalance in the Divergence of Primary Progressive Multiple Sclerosis and Secondary Progressive Multiple Sclerosis. J. Proteome Res. 2023, 22, 743–757. [Google Scholar] [PubMed]
  52. Zeman, D.; Adam, P.; Kalistová, H.; Sobek, O.; Kelbich, P.; Andel, J.; Andel, M. Transferrin in patients with multiple sclerosis, a comparison among various subgroups of multiple sclerosis patients. Acta Neurol. Scand. 2000, 101, 89–94. [Google Scholar] [CrossRef] [PubMed]
  53. De Riccardis, L.; Buccolieri, A.; Muci, M.; Pitotti, E.; De Robertis, F.; Trianni, G.; Manno, D.; Maffia, M. Copper and ceruloplasmin dyshomeostasis in serum and cerebrospinal fluid of multiple sclerosis subjects. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1828–1838. [Google Scholar] [CrossRef] [PubMed]
  54. Trentini, A.; Castellazzi, M.; Romani, A.; Squerzanti, M.; Baldi, E.; Caniatti, M.L.; Pugliatti, M.; Granieri, E.; Fainardi, E.; Bellini, T.; et al. Evaluation of total, ceruloplasmin-associated and type II ferroxidase activities in serum and cerebrospinal fluid of multiple sclerosis patients. J. Neurol. Sci. 2017, 377, 133–136. [Google Scholar] [CrossRef] [PubMed]
  55. Emami Aleagha, M.S.; Siroos, B.; Ahmadi, M.; Balood, M.; Palangi, A.; Haghighi, A.N.; Harirchian, M.H. Decreased concentration of Klotho in the cerebrospinal fluid of patients with relapsing-remitting multiple sclerosis. J. Neuroimmunol. 2015, 281, 5–8. [Google Scholar] [CrossRef] [PubMed]
  56. Voortman, M.M.; Damulina, A.; Pirpamer, L.; Pinter, D.; Pichler, A.; Enzinger, C.; Ropele, S.; Bachmaier, G.; Archelos, J.J.; Marsche, G.; et al. Decreased Cerebrospinal Fluid Antioxidative Capacity Is Related to Disease Severity and Progression in Early Multiple Sclerosis. Biomolecules 2021, 11, 1264. [Google Scholar] [CrossRef]
  57. Romme Christensen, J.; Börnsen, L.; Khademi, M.; Olsson, T.; Jensen, P.E.; Sørensen, P.S.; Sellebjerg, F. CSF inflammation and axonal damage are increased and correlate in progressive multiple sclerosis. Mult. Scler. 2013, 19, 877–884. [Google Scholar] [CrossRef]
  58. Castellazzi, M.; Trentini, A.; Romani, A.; Valacchi, G.; Bellini, T.; Bonaccorsi, G.; Fainardi, E.; Cavicchio, C.; Passaro, A.; Zuliani, G.; et al. Decreased arylesterase activity of paraoxonase-1 (PON-1) might be a common denominator of neuroinflammatory and neurodegenerative diseases. Int. J. Biochem. Cell Biol. 2016, 81, 356–363. [Google Scholar] [CrossRef]
  59. Jiménez-Jiménez, F.J.; de Bustos, F.; Molina, J.A.; de Andrés, C.; Gasalla, T.; Ortí-Pareja, M.; Zurdo, M.; Porta, J.; Castellano-Millán, F.; Arenas, J.; et al. Cerebrospinal fluid levels of alpha-tocopherol in patients with multiple sclerosis. Neurosci. Lett. 1998, 249, 65–67. [Google Scholar] [CrossRef]
  60. Stampanoni Bassi, M.; Nuzzo, T.; Gilio, L.; Miroballo, M.; Casamassa, A.; Buttari, F.; Bellantonio, P.; Fantozzi, R.; Galifi, G.; Furlan, R.; et al. Cerebrospinal fluid levels of L-glutamate signal central inflammatory neurodegeneration in multiple sclerosis. J. Neurochem. 2021, 159, 857–866. [Google Scholar] [CrossRef]
  61. de Bustos, F.; Navarro, J.A.; de Andrés, C.; Molina, J.A.; Jiménez-Jiménez, F.J.; Ortí-Pareja, M.; Gasalla, T.; Tallón-Barranco, A.; Martínez-Salio, A.; Arenas, J. Cerebrospinal fluid nitrate levels in patients with multiple sclerosis. Eur. Neurol. 1999, 41, 44–47. [Google Scholar] [CrossRef] [PubMed]
  62. Kastenbauer, S.; Kieseier, B.C.; Becker, B.F. No evidence of increased oxidative degradation of urate to allantoin in the CSF and serum of patients with multiple sclerosis. J. Neurol. 2005, 252, 611–612. [Google Scholar] [CrossRef] [PubMed]
  63. Amorini, A.M.; Petzold, A.; Tavazzi, B.; Eikelenboom, J.; Keir, G.; Belli, A.; Giovannoni, G.; Di Pietro, V.; Polman, C.; D’Urso, S.; et al. Increase of uric acid and purine comounds in biological fluids of multiple sclerosis patients. Clin. Biochem. 2009, 42, 1001–1006. [Google Scholar] [CrossRef] [PubMed]
  64. Kuračka, L.; Kalnovičová, T.; Kucharská, J.; Turčáni, P. Multiple sclerosis: Evaluation of purine nucleotide metabolism in central nervous system in association with serum levels of selected fat-soluble antioxidants. Mult. Scler. Int. 2014, 2014, 759808. [Google Scholar] [CrossRef]
  65. Paar, M.; Seifried, K.; Cvirn, G.; Buchmann, A.; Khalil, M.; Oettl, K. Redox State of Human Serum Albumin in Multiple Sclerosis, A Pilot Study. Int. J. Mol. Sci. 2022, 23, 15806. [Google Scholar] [CrossRef] [PubMed]
  66. Hirotani, M.; Maita, C.; Niino, M.; Iguchi-Ariga, S.; Hamada, S.; Ariga, H.; Sasaki, H. Correlation between DJ-1 levels in the cerebrospinal fluid and the progression of disabilities in multiple sclerosis patients. Mult. Scler. 2008, 14, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  67. Bruno, A.; Dolcetti, E.; Azzolini, F.; Buttari, F.; Gilio, L.; Iezzi, E.; Galifi, G.; Borrelli, A.; Furlan, R.; Finardi, A.; et al. BACE1 influences clinical manifestations and central inflammation in relapsing remitting multiple sclerosis. Mult. Scler. Relat. Disord. 2023, 71, 104528. [Google Scholar] [CrossRef] [PubMed]
  68. Ljubisavljevicm, S.; Stojanovicm, I.; Cvetkovicm, T.; Vojinovicm, S.; Stojanovm, D.; Stojanovicm, D.; Stefanovicm, N.; Pavlovicm, D. Erythrocytes’ antioxidative capacity as a potential marker of oxidative stress intensity in neuroinflammation. J. Neurol. Sci. 2014, 337, 8–13. [Google Scholar] [CrossRef]
  69. Syburra, C.; Passi, S. Oxidative stress in patients with multiple sclerosis. Ukr. Biokhim. Zh. 1999, 71, 112–115. [Google Scholar]
  70. Zagórski, T.; Dudek, I.; Berkan, L.; Mazurek, M.; Kedziora, J.; Chmielewski, H. Aktywność dysmutazy ponadtlenkowej (SOD-1) w erytrocytach chorych na stwardnienie rozsiane [Superoxide dismutase (SOD-1) activity in erythrocytes of patients with multiple sclerosis]. Neurol. Neurochir. Pol. 1991, 25, 725–730. [Google Scholar]
  71. PĂdureanu, R.; Albu, C.V.; PĂdureanu, V.; BugĂ, A.M. Oxidative Stress and Vitamin D as Predictors in Multiple Sclerosis. Curr. Health Sci. J. 2020, 46, 371–378. [Google Scholar] [PubMed]
  72. Naziroglu, M.; Kutluhan, S.; Ovey, I.S.; Aykur, M.; Yurekli, V.A. Modulation of oxidative stress, apoptosis, and calcium entry in leukocytes of patients with multiple sclerosis by Hypericum perforatum. Nutr. Neurosci. 2014, 17, 214–221. [Google Scholar] [CrossRef] [PubMed]
  73. Koch, M.; Ramsaransing, G.S.; Arutjunyan, A.V.; Stepanov, M.; Teelken, A.; Heersema, D.J.; De Keyser, J. Oxidative stress in serum and peripheral blood leukocytes in patients with different disease courses of multiple sclerosis. J. Neurol. 2006, 253, 483–487. [Google Scholar] [CrossRef] [PubMed]
  74. Koch, M.; Mostert, J.; Arutjunyan, A.; Stepanov, M.; Teelken, A.; Heersema, D.; De Keyser, J. Peripheral blood leukocyte NO production and oxidative stress in multiple sclerosis. Mult. Scler. 2008, 14, 159–265. [Google Scholar] [CrossRef] [PubMed]
  75. Polachini, C.R.; Spanevello, R.M.; Zanini, D.; Baldissarelli, J.; Pereira, L.B.; Schetinger, M.R.; da Cruz, I.B.; Assmann, C.E.; Bagatini, M.D.; Morsch, V.M. Evaluation of Delta-Aminolevulinic Dehydratase Activity, Oxidative Stress Biomarkers. and Vitamin D Levels in Patients with Multiple Sclerosis. Neurotox. Res. 2016, 29, 230–242. [Google Scholar] [CrossRef] [PubMed]
  76. Mossberg, N.; Movitz, C.; Hellstrand, K.; Bergström, T.; Nilsson, S.; Andersen, O. Oxygen radical production in leukocytes and disease severity in multiple sclerosis. J. Neuroimmunol. 2009, 213, 131–134. [Google Scholar] [CrossRef] [PubMed]
  77. Emamgholipour, S.; Hossein-Nezhad, A.; Sahraian, M.A.; Askarisadr, F.; Ansari, M. Evidence for possible role of melatonin in reducing oxidative stress in multiple sclerosis through its effect on SIRT1 and antioxidant enzymes. Life Sci. 2016, 145, 34–41. [Google Scholar] [CrossRef] [PubMed]
  78. Gonzalo, H.; Nogueras, L.; Gil-Sánchez, A.; Hervás, J.V.; Valcheva, P.; González-Mingot, C.; Martin-Gari, M.; Canudes, M.; Peralta, S.; Solana, M.J.; et al. Impairment of Mitochondrial Redox Status in Peripheral Lymphocytes of Multiple Sclerosis Patients. Front. Neurosci. 2019, 13, 938. [Google Scholar] [CrossRef] [PubMed]
  79. Borisovs, V.; Ļeonova, E.; Baumane, L.; Kalniņa, J.; Mjagkova, N.; Sjakste, N. Blood levels of nitric oxide and DNA breaks assayed in whole blood and isolated peripheral blood mononucleated cells in patients with multiple sclerosis. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2019, 843, 90–94. [Google Scholar] [CrossRef]
  80. Pistono, C.; Monti, M.C.; Boiocchi, C.; Berzolari, F.G.; Osera, C.; Mallucci, G.; Cuccia, M.; Pascale, A.; Montomoli, C.; Bergamaschi, R. Response to oxidative stress of peripheral blood mononuclear cells from multiple sclerosis patients and healthy controls. Cell Stress Chaperones 2020, 25, 81–91. [Google Scholar] [CrossRef]
  81. Grecchi, S.; Mazzini, G.; Lisa, A.; Armentero, M.T.; Bergamaschi, R.; Romani, A.; Blandini, F.; Di Perri, C.; Scovassi, A.I. Search for cellular stress biomarkers in lymphocytes from patients with multiple sclerosis: A pilot study. PLoS ONE 2012, 7, e44935. [Google Scholar] [CrossRef]
  82. Hargreaves, I.; Mody, N.; Land, J.; Heales, S. Blood Mononuclear Cell Mitochondrial Respiratory Chain Complex IV Activity Is Decreased in Multiple Sclerosis Patients, Effects of β-Interferon Treatment. J. Clin. Med. 2018, 7, 36. [Google Scholar] [CrossRef] [PubMed]
  83. Dziedzic, A.; Morel, A.; Miller, E.; Bijak, M.; Sliwinski, T.; Synowiec, E.; Ceremuga, M.; Saluk-Bijak, J. Oxidative Damage of Blood Platelets Correlates with the Degree of Psychophysical Disability in Secondary Progressive Multiple Sclerosis. Oxid. Med. Cell. Longev. 2020, 2020, 2868014. [Google Scholar] [CrossRef] [PubMed]
  84. Morel, A.; Bijak, M.; Miller, E.; Rywaniak, J.; Miller, S.; Saluk, J. Relationship between the Increased Haemostatic Properties of Blood Platelets and Oxidative Stress Level in Multiple Sclerosis Patients with the Secondary Progressive Stage. Oxid. Med. Cell. Longev. 2015, 2015, 240918. [Google Scholar] [CrossRef] [PubMed]
  85. Miller, E.; Mrowicka, M.; Saluk-Juszczak, J.; Ireneusz, M. The level of isoprostanes as a non-invasive marker for in vivo lipid peroxidation in secondary progressive multiple sclerosis. Neurochem. Res. 2011, 36, 1012–1016. [Google Scholar] [CrossRef]
  86. Tavazzi, B.; Batocchi, A.P.; Amorini, A.M.; Nociti, V.; D’Urso, S.; Longo, S.; Gullotta, S.; Picardi, M.; Lazzarino, G. Serum metabolic profile in multiple sclerosis patients. Mult. Scler. Int. 2011, 2011, 167156. [Google Scholar] [CrossRef] [PubMed]
  87. Acar, A.; Ugur Cevik, M.; Evliyaoglu, O.; Uzar, E.; Tamam, Y.; Arıkanoglu, A.; Yucel, Y.; Varol, S.; Onder, H.; Taşdemir, N. Evaluation of serum oxidant/antioxidant balance in multiple sclerosis. Acta Neurol. Belg. 2012, 112, 275–280. [Google Scholar] [CrossRef] [PubMed]
  88. Yousefi, B.; Ahmadi, Y.; Ghorbanihaghjo, A.; Faghfoori, Z.; Irannejad, V.S. Serum arsenic and lipid peroxidation levels in patients with multiple sclerosis. Biol. Trace Elem. Res. 2014, 158, 276–279. [Google Scholar] [CrossRef] [PubMed]
  89. Gironi, M.; Borgiani, B.; Mariani, E.; Cursano, C.; Mendozzi, L.; Cavarretta, R.; Saresella, M.; Clerici, M.; Comi, G.; Rovaris, M.; et al. Oxidative stress is differentially present in multiple sclerosis courses, early evident. and unrelated to treatment. J. Immunol. Res. 2014, 2014, 961863. [Google Scholar] [CrossRef]
  90. Adamczyk-Sowa, M.; Pierzchala, K.; Sowa, P.; Polaniak, R.; Kukla, M.; Hartel, M. Influence of melatonin supplementation on serum antioxidative properties and impact of the quality of life in multiple sclerosis patients. J. Physiol. Pharmacol. 2014, 65, 543–550. [Google Scholar]
  91. Karlík, M.; Valkovič, P.; Hančinová, V.; Krížová, L.; Tóthová, Ľ.; Celec, P. Markers of oxidative stress in plasma and saliva in patients with multiple sclerosis. Clin. Biochem. 2015, 48, 24–28. [Google Scholar] [CrossRef] [PubMed]
  92. Morel, A.; Bijak, M.; Niwald, M.; Miller, E.; Saluk, J. Markers of oxidative/nitrative damage of plasma proteins correlated with EDSS and BDI scores in patients with secondary progressive multiple sclerosis. Redox Rep. 2017, 22, 547–555. [Google Scholar] [CrossRef] [PubMed]
  93. Juybari, K.B.; Ebrahimi, G.; Momeni Moghaddam, M.A.; Asadikaram, G.; Torkzadeh-Mahani, M.; Akbari, M.; Mirzamohammadi, S.; Karimi, A.; Nematollahi, M.H. Evaluation of serum arsenic and its effects on antioxidant alterations in relapsing-remitting multiple sclerosis patients. Mult. Scler. Relat. Disord. 2018, 19, 79–84. [Google Scholar] [CrossRef] [PubMed]
  94. Padureanu, R.; Albu, C.V.; Mititelu, R.R.; Bacanoiu, M.V.; Docea, A.O.; Calina, D.; Padureanu, V.; Olaru, G.; Sandu, R.E.; Malin, R.D.; et al. Oxidative Stress and Inflammation Interdependence in Multiple Sclerosis. J. Clin. Med. 2019, 8, 1815. [Google Scholar] [CrossRef] [PubMed]
  95. Joodi Khanghah, O.; Nourazarian, A.; Khaki-Khatibi, F.; Nikanfar, M.; Laghousi, D.; Vatankhah, A.M.; Moharami, S. Evaluation of the Diagnostic and Predictive Value of Serum Levels of ANT1, ATG5, and Parkin in Multiple Sclerosis. Clin. Neurol. Neurosurg. 2020, 197, 106197. [Google Scholar] [CrossRef] [PubMed]
  96. Talebi, M.; Majdi, A.; Nasiri, E.; Naseri, A.; Sadigh-Eteghad, S. The correlation between circulating inflammatory, oxidative stress, and neurotrophic factors level with the cognitive outcomes in multiple sclerosis patients. Neurol. Sci. 2021, 42, 2291–2300. [Google Scholar] [CrossRef] [PubMed]
  97. Obradovic, D.; Andjelic, T.; Ninkovic, M.; Dejanovic, B.; Kotur-Stevuljevic, J. Superoxide dismutase (SOD), advanced oxidation protein products (AOPP), and disease-modifying treatment are related to better relapse recovery after corticosteroid treatment in multiple sclerosis. Neurol. Sci. 2021, 42, 3241–3247. [Google Scholar] [CrossRef]
  98. Ghonimi, N.A.M.; Elsharkawi, K.A.; Khyal, D.S.M.; Abdelghani, A.A. Serum malondialdehyde as a lipid peroxidation marker in multiple sclerosis patients and its relation to disease characteristics. Mult. Scler. Relat. Disord. 2021, 51, 102941. [Google Scholar] [CrossRef] [PubMed]
  99. Borisovs, V.; Bodrenko, J.; Kalnina, J.; Sjakste, N. Nitrosative stress parameters and the level of oxidized DNA bases in patients with multiple sclerosis. Metab. Brain Dis. 2021, 36, 1935–1941. [Google Scholar] [CrossRef]
  100. Naseri, A.; Forghani, N.; Sadigh-Eteghad, S.; Shanehbandi, D.; Asadi, M.; Nasiri, E.; Talebi, M. Circulatory antioxidant and oxidative stress markers are in correlation with demographics but not cognitive functions in multiple sclerosis patients. Mult. Scler. Relat. Disord. 2022, 57, 103432. [Google Scholar] [CrossRef]
  101. Ortiz, G.G.; Macías-Islas, M.A.; Pacheco-Moisés, F.P.; Cruz-Ramos, J.A.; Sustersik, S.; Barba, E.A.; Aguayo, A. Oxidative stress is increased in serum from Mexican patients with relapsing-remitting multiple sclerosis. Dis. Markers 2009, 26, 35–39. [Google Scholar] [CrossRef] [PubMed]
  102. Besler, H.T.; Comoğlu, S. Lipoprotein oxidation, plasma total antioxidant capacity and homocysteine level in patients with multiple sclerosis. Nutr. Neurosci. 2003, 6, 189–196. [Google Scholar] [CrossRef]
  103. Bizoń, A.; Chojdak-Łukasiewicz, J.; Kołtuniuk, A.; Budrewicz, S.; Pokryszko-Dragan, A.; Piwowar, A. Evaluation of Selected Oxidant/Antioxidant Parameters in Patients with Relapsing-Remitting Multiple Sclerosis Undergoing Disease-Modifying Therapies. Antioxidants 2022, 11, 2416. [Google Scholar] [CrossRef] [PubMed]
  104. Koch, M.; Mostert, J.; Arutjunyan, A.V.; Stepanov, M.; Teelken, A.; Heersema, D.; De Keyser, J. Plasma lipid peroxidation and progression of disability in multiple sclerosis. Eur. J. Neurol. 2007, 14, 529–533. [Google Scholar] [CrossRef] [PubMed]
  105. Ferretti, G.; Bacchetti, T.; Principi, F.; Di Ludovico, F.; Viti, B.; Angeleri, V.A.; Danni, M.; Provinciali, L. Increased levels of lipid hydroperoxides in plasma of patients with multiple sclerosis, a relationship with paraoxonase activity. Mult. Scler. 2005, 11, 677–682. [Google Scholar] [CrossRef]
  106. Oliveira, S.R.; Simão, A.N.; Kallaur, A.P.; de Almeida, E.R.; Morimoto, H.K.; Lopes, J.; Dichi, I.; Kaimen-Maciel, D.R.; Reiche, E.M. Disability in patients with multiple sclerosis: Influence of insulin resistance, adiposity, and oxidative stress. Nutrition 2014, 30, 268–273. [Google Scholar] [CrossRef] [PubMed]
  107. Kallaur, A.P.; Lopes, J.; Oliveira, S.R.; Simão, A.N.; Reiche, E.M.; de Almeida, E.R.; Morimoto, H.K.; de Pereira, W.L.; Alfieri, D.F.; Borelli, S.D.; et al. Immune-Inflammatory and Oxidative and Nitrosative Stress Biomarkers of Depression Symptoms in Subjects with Multiple Sclerosis; Increased Peripheral Inflammation but Less Acute Neuroinflammation. Mol. Neurobiol. 2016, 53, 5191–5202. [Google Scholar] [CrossRef] [PubMed]
  108. Kallaur, A.P.; Reiche, E.M.V.; Oliveira, S.R.; Simão, A.N.C.; Pereira, W.L.C.J.; Alfieri, D.F.; Flauzino, T.; Proença, C.M.; Lozovoy, M.A.B.; Kaimen-Maciel, D.R.; et al. Genetic, Immune-Inflammatory, and Oxidative Stress Biomarkers as Predictors for Disability and Disease Progression in Multiple Sclerosis. Mol. Neurobiol. 2017, 54, 31–44. [Google Scholar] [CrossRef]
  109. Oliveira, S.R.; Kallaur, A.P.; Simão, A.N.; Morimoto, H.K.; Lopes, J.; Panis, C.; Petenucci, D.L.; da Silva, E.; Cecchini, R.; Kaimen-Maciel, D.R.; et al. Oxidative stress in multiple sclerosis patients in clinical remission, association with the expanded disability status scale. J. Neurol. Sci. 2012, 321, 49–53. [Google Scholar] [CrossRef]
  110. Ferreira, K.P.Z.; Oliveira, S.R.; Kallaur, A.P.; Kaimen-Maciel, D.R.; Lozovoy, M.A.B.; de Almeida, E.R.D.; Morimoto, H.K.; Mezzaroba, L.; Dichi, I.; Reiche, E.M.V.; et al. Disease progression and oxidative stress are associated with higher serum ferritin levels in patients with multiple sclerosis. J. Neurol. Sci. 2017, 373, 236–241. [Google Scholar] [CrossRef]
  111. Oliveira, S.R.; Simão, A.N.C.; Alfieri, D.F.; Flauzino, T.; Kallaur, A.P.; Mezzaroba, L.; Lozovoy, M.A.B.; Sabino, B.S.; Ferreira, K.P.Z.; Pereira, W.L.C.J.; et al. Vitamin D deficiency is associated with disability and disease progression in multiple sclerosis patients independently of oxidative and nitrosative stress. J. Neurol. Sci. 2017, 381, 213–219. [Google Scholar] [CrossRef]
  112. Mezzaroba, L.; Simão, A.N.C.; Oliveira, S.R.; Flauzino, T.; Alfieri, D.F.; de Carvalho Jennings Pereira, W.L.; Kallaur, A.P.; Lozovoy, M.A.B.; Kaimen-Maciel, D.R.; Maes, M.; et al. Antioxidant and Anti-inflammatory Diagnostic Biomarkers in Multiple Sclerosis, A Machine Learning Study. Mol. Neurobiol. 2020, 57, 2167–2178. [Google Scholar] [CrossRef] [PubMed]
  113. Karahalil, B.; Orhan, G.; Ak, F. The impact of detoxifying and repair gene polymorphisms and the levels of serum ROS in the susceptibility to multiple sclerosis. Clin. Neurol. Neurosurg. 2015, 139, 288–294. [Google Scholar] [CrossRef] [PubMed]
  114. Siotto, M.; Filippi, M.M.; Simonelli, I.; Landi, D.; Ghazaryan, A.; Vollaro, S.; Ventriglia, M.; Pasqualetti, P.; Rongioletti, M.C.A.; Squitti, R.; et al. Oxidative Stress Related to Iron Metabolism in Relapsing Remitting Multiple Sclerosis Patients With Low Disability. Front. Neurosci. 2019, 13, 86. [Google Scholar] [CrossRef] [PubMed]
  115. Flauzino, T.; Simão, A.N.C.; de Carvalho Jennings Pereira, W.L.; Alfieri, D.F.; Oliveira, S.R.; Kallaur, A.P.; Lozovoy, M.A.B.; Kaimen-Maciel, D.R.; Maes, M.; Reiche, E.M.V. Disability in multiple sclerosis is associated with age and inflammatory, metabolic and oxidative/nitrosative stress biomarkers: Results of multivariate and machine learning procedures. Metab. Brain Dis. 2019, 34, 1401–1413. [Google Scholar] [CrossRef]
  116. Teunissen, C.E.; Sombekke, M.; van Winsen, L.; Killestein, J.; Barkhof, F.; Polman, C.H.; Dijkstra, C.D.; Blankenstein, M.A.; Pratico, D. Increased plasma 8,12-iso-iPF2alpha- VI levels in relapsing multiple sclerosis patients are not predictive of disease progression. Mult. Scler. 2012, 18, 1092–1098. [Google Scholar] [CrossRef]
  117. Miller, E.; Walczak, A.; Saluk, J.; Ponczek, M.B.; Majsterek, I. Oxidative modification of patient’s plasma proteins and its role in pathogenesis of multiple sclerosis. Clin. Biochem. 2012, 45, 26–30. [Google Scholar] [CrossRef]
  118. Tasset, I.; Agüera, E.; Sánchez-López, F.; Feijóo, M.; Giraldo, A.I.; Cruz, A.H.; Gascón, F.; Túnez, I. Peripheral oxidative stress in relapsing-remitting multiple sclerosis. Clin. Biochem. 2012, 45, 440–444. [Google Scholar] [CrossRef]
  119. Fiorini, A.; Koudriavtseva, T.; Bucaj, E.; Coccia, R.; Foppoli, C.; Giorgi A Schininà, M.E.; Di Domenico, F.; De Marco, F.; Perluigi, M. Involvement of oxidative stress in occurrence of relapses in multiple sclerosis, the spectrum of oxidatively modified serum proteins detected by proteomics and redox proteomics analysis. PLoS ONE 2013, 8, e65184. [Google Scholar] [CrossRef]
  120. Sadowska-Bartosz, I.; Adamczyk-Sowa, M.; Galiniak, S.; Mucha, S.; Pierzchala, K.; Bartosz, G. Oxidative modification of serum proteins in multiple sclerosis. Neurochem. Int. 2013, 63, 507–516. [Google Scholar] [CrossRef]
  121. Sadowska-Bartosz, I.; Adamczyk-Sowa, M.; Gajewska, A.; Bartosz, G. Oxidative modification of blood serum proteins in multiple sclerosis after interferon or mitoxantrone treatment. J. Neuroimmunol. 2014, 266, 67–74. [Google Scholar] [CrossRef] [PubMed]
  122. Pasquali, L.; Pecori, C.; Lucchesi, C.; LoGerfo, A.; Iudice, A.; Siciliano, G.; Bonuccelli, U. Plasmatic oxidative stress biomarkers in multiple sclerosis, relation with clinical and demographic characteristics. Clin. Biochem. 2015, 48, 19–23. [Google Scholar] [CrossRef] [PubMed]
  123. de Carvalho Jennings Pereira, W.L.; Flauzino, T.; Alfieri, D.F.; Oliveira, S.R.; Kallaur, A.P.; Simão, A.N.C.; Lozovoy, M.A.B.; Kaimen-Maciel, D.R.; Maes, M.; Reiche, E.M.V. Immune-Inflammatory, metabolic and hormonal biomarkers are associated with the clinical forms and disability progression in patients with multiple sclerosis: A follow-up study. J. Neurol. Sci. 2020, 410, 116630. [Google Scholar] [CrossRef] [PubMed]
  124. Bizoń, A.; Chojdak-Łukasiewicz, J.; Budrewicz, S.; Pokryszko-Dragan, A.; Piwowar, A. Exploring the Relationship between Antioxidant Enzymes, Oxidative Stress Markers, and Clinical Profile in Relapsing-Remitting Multiple Sclerosis. Antioxidants 2023, 12, 1638. [Google Scholar] [CrossRef] [PubMed]
  125. Ljubisavljevic, S.; Stojanovic, I.; Basic, J.; Pavlovic, D.A. The Validation Study of Neurofilament Heavy Chain and 8-hydroxy-2′-deoxyguanosine as Plasma Biomarkers of Clinical/Paraclinical Activity in First and Relapsing-Remitting Demyelination Acute Attacks. Neurotox. Res. 2016, 30, 530–538. [Google Scholar] [CrossRef]
  126. Vasić, M.; Topić, A.; Marković, B.; Milinković, N.; Dinčić, E. Oxidative stress-related risk of the multiple sclerosis development. J. Med. Biochem. 2023, 42, 1–8. [Google Scholar] [CrossRef]
  127. Jamroz-Wiśniewska, A.; Bełtowski, J.; Wójcicka, G.; Bartosik-Psujek, H.; Rejdak, K. Cladribine Treatment Improved Homocysteine Metabolism and Increased Total Serum Antioxidant Activity in Secondary Progressive Multiple Sclerosis Patients. Oxid. Med. Cell. Longev. 2020, 2020, 1654754. [Google Scholar] [CrossRef] [PubMed]
  128. Sfagos, C.; Makis, A.C.; Chaidos, A.; Hatzimichael, E.C.; Dalamaga, A.; Kosma, K.; Bourantas, K.L. Serum ferritin, transferrin and soluble transferrin receptor levels in multiple sclerosis patients. Mult. Scler. 2005, 11, 272–275. [Google Scholar] [CrossRef] [PubMed]
  129. Visconti, A.; Cotichini, R.; Cannoni, S.; Bocca, B.; Forte, G.; Ghazaryan, A.; Santucci, S.; D’Ippolito, C.; Stazi, M.A.; Salvetti, M.; et al. Concentration of elements in serum of patients affected by multiple sclerosis with first demyelinating episode: A six-month longitudinal follow-up study. Ann. Ist. Super. Sanita. 2005, 41, 217–222. [Google Scholar]
  130. Alimonti, A.; Ristori, G.; Giubilei, F.; Stazi, M.A.; Pino, A.; Visconti, A.; Brescianini, S.; Sepe Monti, M.; Forte, G.; Stanzione, P.; et al. Serum chemical elements and oxidative status in Alzheimer’s disease, Parkinson disease and multiple sclerosis. Neurotoxicology 2007, 28, 450–456. [Google Scholar] [CrossRef]
  131. Abo-Krysha, N.; Rashed, L. The role of iron dysregulation in the pathogenesis of multiple sclerosis, an Egyptian study. Mult. Scler. 2008, 14, 602–608. [Google Scholar] [CrossRef] [PubMed]
  132. Ristori, G.; Brescianini, S.; Pino, A.; Visconti, A.; Vittori, D.; Coarelli, G.; Cotichini, R.; Bocca, B.; Forte, G.; Pozzilli, C.; et al. Serum elements and oxidative status in clinically isolated syndromes, imbalance and predictivity. Neurology 2011, 76, 549–555. [Google Scholar] [CrossRef] [PubMed]
  133. Doğan, H.O.; Yildiz, Ö.K. Serum NADPH oxidase concentrations and the associations with iron metabolism in relapsing remitting multiple sclerosis. J. Trace Elem. Med. Biol. 2019, 55, 39–43. [Google Scholar] [CrossRef] [PubMed]
  134. Toczylowska, B.; Zieminska, E.; Podlecka-Pietowska, A.; Ruszczynska, A.; Chalimoniuk, M. Serum metabolic profiles and metal levels of patients with multiple sclerosis and patients with neuromyelitis optica spectrum disorders—NMR spectroscopy and ICP-MS studies. Mult. Scler. Relat. Disord. 2022, 60, 103672. [Google Scholar] [CrossRef] [PubMed]
  135. Sakai, T.; Inoue, A.; Koh, C.S.; Ikeda, S. [A study of free radical defense and oxidative stress in the sera of patients with neuroimmunological disorders]. Arerugi 2000, 49, 12–18. [Google Scholar]
  136. Cervellati, C.; Romani, A.; Fainardi, E.; Trentini, A.; Squerzanti, M.; Baldi, E.; Caniatti, M.L.; Granieri, E.; Bellini, T.; Castellazzi, M. Serum ferroxidase activity in patients with multiple sclerosis: A pilot study. In Vivo 2014, 28, 1197–1200. [Google Scholar] [PubMed]
  137. Adamczyk-Sowa, M.; Sowa, P.; Mucha, S.; Zostawa, J.; Mazur, B.; Owczarek, M.; Pierzchała, K. Changes in Serum Ceruloplasmin Levels Based on Immunomodulatory Treatments and Melatonin Supplementation in Multiple Sclerosis Patients. Med. Sci. Monit. 2016, 22, 2484–24891. [Google Scholar] [CrossRef] [PubMed]
  138. Hadžović-Džuvo, A.; Lepara, O.; Valjevac, A.; Avdagić, N.; Hasić, S.; Kiseljaković, E.; Ibragić, S.; Alajbegović, A. Serum total antioxidant capacity in patients with multiple sclerosis. Bosn. J. Basic. Med. Sci. 2011, 11, 33–36. [Google Scholar] [CrossRef] [PubMed]
  139. Kirbas, A.; Kirbas, S.; Anlar, O.; Efe, H.; Yilmaz, A. Serum paraoxonase and arylesterase activity and oxidative status in patients with multiple sclerosis. J. Clin. Neurosci. 2013, 20, 1106–1109. [Google Scholar] [CrossRef]
  140. Aydin, O.; Ellidag, H.Y.; Eren, E.; Kurtulus, F.; Yaman, A.; Yılmaz, N. Ischemia modified albumin is an indicator of oxidative stress in multiple sclerosis. Biochem. Med. 2014, 24, 383–389. [Google Scholar] [CrossRef]
  141. Siváková, M.; Siarnik, P.; Filippi, P.; Vlcek, M.; Imrich, R.; Turcani, P.; Zitnanova, I.; Penesova, A.; Radikova, Z.; Kollar, B. Oxidative stress in patients with newly diagnosed multiple sclerosis, any association with subclinical atherosclerosis? Neuro Endocrinol. Lett. 2019, 40, 135–140. [Google Scholar] [PubMed]
  142. Yevgi, R.; Demir, R. Oxidative stress activity of fingolimod in multiple sclerosis. Clin. Neurol. Neurosurg. 2021, 202, 106500. [Google Scholar] [CrossRef] [PubMed]
  143. Ozben, S.; Kucuksayan, E.; Koseoglu, M.; Erel, O.; Neselioglu, S.; Ozben, T. Plasma thiol/disulphide homeostasis changes in patients with relapsing-remitting multiple sclerosis. Int. J. Clin. Pract. 2021, 75, e14241. [Google Scholar] [CrossRef] [PubMed]
  144. Arslan, B.; Arslan, G.A.; Tuncer, A.; Karabudak, R.; Dinçel, A.S. Evaluation of Thiol Homeostasis in Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorders. Front. Neurol. 2021, 12, 716195. [Google Scholar] [CrossRef] [PubMed]
  145. Smirnova, L.P.; Mednova, I.A.; Krotenko, N.M.; Alifirova, V.M.; Ivanova, S.A. IgG-Dependent Dismutation of Superoxide in Patients with Different Types of Multiple Sclerosis and Healthy Subjects. Oxid. Med. Cell. Longev. 2020, 2020, 8171020. [Google Scholar] [CrossRef]
  146. Essenburg, C.; Browne, R.W.; Ghazal, D.; Tamaño-Blanco, M.; Jakimovski, D.; Weinstock-Guttman, B.; Zivadinov, R.; Ramanathan, M. Antioxidant defense enzymes in multiple sclerosis, A 5-year follow-up study. Eur. J. Neurol. 2023, 30, 2338–2347. [Google Scholar] [CrossRef]
  147. Chitsaz, N.; Dehghani, L.; Safi, A.; Esmalian-Afyouni, N.; Shaygannejad, V.; Rezvani, M.; Sohrabi, K.; Moridi, K.; Moayednia, M. Evaluation of glucose-6-phosphate dehydrogenase serum level in patients with multiple sclerosis and neuromyelitis optica. Iran J. Neurol. 2019, 18, 150–153. [Google Scholar] [CrossRef] [PubMed]
  148. Jamroz-Wiśniewska, A.; Bełtowski, J.; Bartosik-Psujek, H.; Wójcicka, G.; Rejdak, K. Processes of plasma protein N-homocysteinylation in multiple sclerosis. Int. J. Neurosci. 2017, 127, 709–715. [Google Scholar] [CrossRef]
  149. Kanesaka, T.; Mori, M.; Hattori, T.; Oki, T.; Kuwabara, S. Serum matrix metalloproteinase-3 levels correlate with disease activity in relapsing-remitting multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2006, 77, 185–188. [Google Scholar] [CrossRef]
  150. Mahmoudian, E.; Khalilnezhad, A.; Gharagozli, K.; Amani, D. Thioredoxin-1,redox factor-1 and thioredoxin-interacting protein, mRNAs are differentially expressed in Multiple Sclerosis patients exposed and non-exposed to interferon and immunosuppressive treatments. Gene 2017, 634, 29–36. [Google Scholar] [CrossRef]
  151. De Bustos, F.; Jiménez-Jiménez, F.J.; Molina, J.A.; Gómez-Escalonilla, C.; de Andrés, C.; del Hoyo, P.; Zurdo, M.; Tallón-Barranco, A.; Berbel, A.; Porta-Etessam, J.; et al. Serum levels of coenzyme Q10 in patients with multiple sclerosis. Acta Neurol. Scand. 2000, 101, 209–211. [Google Scholar] [CrossRef] [PubMed]
  152. de Bustos, F.; Jiménez-Jiménez, F.J.; Molina, J.A.; de Andrés, C.; Gasalla, T.; Ortí-Pareja, M.; Ayuso-Peralta, L.; Berbel, A.; Castellano-Millán, F.; Arenas, J.; et al. Serum levels of alpha-carotene.; beta-carotene.; and retinol in patients with multiple sclerosis. Acta Neurol. Belg. 2000, 100, 41–43. [Google Scholar] [PubMed]
  153. Ramsaransing, G.S.; Fokkema, M.R.; Teelken, A.; Arutjunyan, A.V.; Koch, M.; De Keyser, J. Plasma homocysteine levels in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2006, 77, 189–192. [Google Scholar] [CrossRef] [PubMed]
  154. Aliomrani, M.; Sahraian, M.A.; Shirkhanloo, H.; Sharifzadeh, M.; Khoshayand, M.R.; Ghahremani, M.H. Blood Concentrations of Cadmium and Lead in Multiple Sclerosis Patients from Iran. Iran J. Pharm. Res. 2016, 15, 825–833. [Google Scholar] [PubMed]
  155. Aliomrani, M.; Sahraian, M.A.; Shirkhanloo, H.; Sharifzadeh, M.; Khoshayand, M.R.; Ghahremani, M.H. Correlation between heavy metal exposure and GSTM1 polymorphism in Iranian multiple sclerosis patients. Neurol. Sci. 2017, 38, 1271–1278. [Google Scholar] [CrossRef] [PubMed]
  156. Massa, J.; O’Reilly, E.; Munger, K.L.; Delorenze, G.N.; Ascherio, A. Serum uric acid and risk of multiple sclerosis. J. Neurol. 2009, 256, 1643–1648. [Google Scholar] [CrossRef] [PubMed]
  157. Fuhua, P.; Xuhui, D.; Zhiyang, Z.; Ying, J.; Yu, Y.; Feng, T.; Jia, L.; Lijia, G.; Xueqiang, H. Antioxidant status of bilirubin and uric acid in patients with myasthenia gravis. Neuroimmunomodulation 2012, 9, 43–49. [Google Scholar] [CrossRef] [PubMed]
  158. Yang, D.; Weng, Y.; Lin, H.; Xie, F.; Yin, F.; Lou, K.; Zhou, X.; Han, Y.; Li, X.; Zhang, X. Serum uric acid levels in patients with myasthenia gravis are inversely correlated with disability. Neuroreport 2016, 27, 301–305. [Google Scholar] [CrossRef]
  159. Katarina, V.; Gordana, T.; Svetlana, M.D.; Milica, B. Oxidative stress and neuroinflammation should be both considered in the occurrence of fatigue and depression in multiple sclerosis. Acta Neurol. Belg. 2020, 120, 853–861. [Google Scholar] [CrossRef]
  160. Alrouji, M.; Manouchehrinia, A.; Aram, J.; Alotaibi, A.; Alhajlah, S.; Almuhanna, Y.; Alomeir, O.; Shamsi, A.; Gran, B.; Constantinescu, C.S. Investigating the Effect of Cigarette Smoking on Serum Uric Acid Levels in Multiple Sclerosis Patients: A Cross Sectional Study. Brain Sci. 2023, 13, 800. [Google Scholar] [CrossRef]
  161. Altas, M.; Uca, A.U.; Akdag, T.; Odabas, F.O.; Tokgoz, O.S. Serum levels of irisin and nesfatin-1 in multiple sclerosis. Arq. Neuropsiquiatr. 2022, 80, 161–167. [Google Scholar] [CrossRef] [PubMed]
  162. Exley, C.; Mamutse, G.; Korchazhkina, O.; Pye, E.; Strekopytov, S.; Polwart, A.; Hawkins, C. Elevated urinary excretion of aluminium and iron in multiple sclerosis. Mult. Scler. 2006, 12, 533–540. [Google Scholar] [CrossRef] [PubMed]
  163. Guan, J.Z.; Guan, W.P.; Maeda, T.; Guoqing, X.; Wan, G.Z.; Makino, N. Patients with multiple sclerosis show increased oxidative stress markers and somatic telomere length shortening. Mol. Cell. Biochem. 2015, 400, 183–187. [Google Scholar] [CrossRef]
  164. Khorami, H.; Neyestani, T.; Kadkhodaee, M.; Lotfi, J. Increased urinary neopterin, creatinine ratio as a marker of activation of cell-mediated immunity and oxidative stress in the Iranian patients with multiple sclerosis. Iran J. Allergy Asthma Immunol. 2003, 2, 155–158. [Google Scholar]
  165. Tenorio-Laranga, J.; Peltonen, I.; Keskitalo, S.; Duran-Torres, G.; Natarajan, R.; Männistö, P.T.; Nurmi, A.; Vartiainen, N.; Airas, L.; Elovaara, I.; et al. Alteration of prolyl oligopeptidase and activated α-2-macroglobulin in multiple sclerosis subtypes and in the clinically isolated syndrome. Biochem. Pharmacol. 2013, 85, 1783–1794. [Google Scholar] [CrossRef] [PubMed]
  166. Agúndez, J.A.; Arroyo, R.; Ledesma, M.C.; Martínez, C.; Ladero, J.M.; de Andrés, C.; Jiménez-Jiménez, F.J.; Molina, J.A.; Alvarez-Cermeño, J.C.; Varela de Seijas, E.; et al. Frequency of CYP2D6 allelic variants in multiple sclerosis. Acta Neurol. Scand. 1995, 92, 464–467. [Google Scholar] [CrossRef]
  167. Mann, C.L.; Davies, M.B.; Boggild, M.D.; Alldersea, J.; Fryer, A.A.; Jones, P.W.; Ko Ko, C.; Young, C.; Strange, R.C.; Hawkins, C.P. Glutathione S-transferase polymorphisms in MS, their relationship to disability. Neurology 2000, 54, 552–557. [Google Scholar] [CrossRef] [PubMed]
  168. Agúndez, J.A.; García-Martín, E.; Martínez, C.; Benito-León, J.; Millán-Pascual, J.; Díaz-Sánchez, M.; Calleja, P.; Pisa, D.; Turpín-Fenoll, L.; Alonso-Navarro, H.; et al. The GSTP1 gene variant rs1695 is not associated with an increased risk of multiple sclerosis. Cell. Mol. Immunol. 2015, 12, 777–779. [Google Scholar] [CrossRef]
  169. Alexoudi, A.; Zachaki, S.; Stavropoulou, C.; Chatzi, I.; Koumbi, D.; Stavropoulou, K.; Kollia, P.; Karageorgiou, C.E.; Sambani, C. Combined GSTP1 and NQO1 germline polymorphisms in the susceptibility to Multiple Sclerosis. Int. J. Neurosci. 2015, 125, 32–37. [Google Scholar] [CrossRef]
  170. Stavropoulou, C.; Korakaki, D.; Rigana, H.; Voutsinas, G.; Polyzoi, M.; Georgakakos, V.N.; Manola, K.N.; Karageorgiou, C.E.; Sambani, C. Glutathione-S-transferase T1 and M1 gene polymorphisms in Greek patients with multiple sclerosis: A pilot study. Eur. J. Neurol. 2007, 14, 572–574. [Google Scholar] [CrossRef]
  171. Parchami Barju, I.S.; Reiisi, S.; Bayati, A. Human glutathione s-transferase enzyme gene variations and risk of multiple sclerosis in Iranian population cohort. Mult. Scler. Relat. Disord. 2017, 17, 41–46. [Google Scholar] [CrossRef] [PubMed]
  172. Zakrzewska-Pniewska, B.; Styczynska, M.; Podlecka, A.; Samocka, R.; Peplonska, B.; Barcikowska, M.; Kwiecinski, H. Association of apolipoprotein E and myeloperoxidase genotypes to clinical course of familial and sporadic multiple sclerosis. Mult. Scler. 2004, 10, 266–271. [Google Scholar] [CrossRef] [PubMed]
  173. Sidoti, A.; Antognelli, C.; Rinaldi, C.; D’Angelo, R.; Dattola, V.; Girlanda, P.; Talesa, V.; Amato, A. Glyoxalase I A111E, paraoxonase 1 Q192R and L55M polymorphisms, susceptibility factors of multiple sclerosis? Mult. Scler. 2007, 13, 446–453. [Google Scholar] [CrossRef] [PubMed]
  174. Martínez, C.; García-Martín, E.; Benito-León, J.; Calleja, P.; Díaz-Sánchez, M.; Pisa, D.; Alonso-Navarro, H.; Ayuso-Peralta, L.; Torrecilla, D.; Agúndez, J.A.; et al. Paraoxonase 1 polymorphisms are not related with the risk for multiple sclerosis. Neuromolecular Med. 2010, 12, 217–223. [Google Scholar] [CrossRef] [PubMed]
  175. Stavropoulou, C.; Zachaki, S.; Alexoudi, A.; Chatzi, I.; Georgakakos, V.N.; Terzoudi, G.I.; Pantelias, G.E.; Karageorgiou, C.E.; Sambani, C. The C609T inborn polymorphism in NAD(P)H,quinone oxidoreductase 1 is associated with susceptibility to multiple sclerosis and affects the risk of development of the primary progressive form of the disease. Free Radic. Biol. Med. 2011, 51, 713–718. [Google Scholar] [CrossRef]
  176. Agúndez, J.A.; García-Martín, E.; Martínez, C.; Benito-León, J.; Millán-Pascual, J.; Calleja, P.; Díaz-Sánchez, M.; Pisa, D.; Turpín-Fenoll, L.; Alonso-Navarro, H.; et al. NQO1 gene rs1800566 variant is not associated with risk for multiple sclerosis. BMC Neurol. 2014, 14, 87. [Google Scholar] [CrossRef]
  177. Agúndez, J.A.; García-Martín, E.; Martínez, C.; Benito-León, J.; Millán-Pascual, J.; Calleja, P.; Díaz-Sánchez, M.; Pisa, D.; Turpín-Fenoll, L.; Alonso-Navarro, H.; et al. Heme Oxygenase-1 and 2 Common Genetic Variants and Risk for Multiple Sclerosis. Sci. Rep. 2016, 6, 20830. [Google Scholar] [CrossRef] [PubMed]
  178. Cardamone, G.; Paraboschi, E.M.; Soldà, G.; Duga, S.; Saarela, J.; Asselta, R. Genetic Association and Altered Gene Expression of CYBB in Multiple Sclerosis Patients. Biomedicines 2018, 6, 117. [Google Scholar] [CrossRef] [PubMed]
  179. Wigner, P.; Dziedzic, A.; Synowiec, E.; Miller, E.; Bijak, M.; Saluk-Bijak, J. Variation of genes encoding nitric oxide synthases and antioxidant enzymes as potential risks of multiple sclerosis development, a preliminary study. Sci. Rep. 2022, 12, 10603. [Google Scholar] [CrossRef]
  180. Agúndez, J.A.G.; García-Martín, E.; Rodríguez, C.; Benito-León, J.; Millán-Pascual, J.; Díaz-Sánchez, M.; Calleja, P.; Turpín-Fenoll, L.; Alonso-Navarro, H.; García-Albea, E.; et al. Endothelial nitric oxide synthase (NOS3) rs2070744 polymorphism and risk for multiple sclerosis. J. Neural Transm. 2020, 127, 1167–1175. [Google Scholar] [CrossRef]
  181. Bahrami, T.; Taheri, M.; Omrani, M.D.; Karimipoor, M. Associations Between Genomic Variants in lncRNA-TRPM2-AS and lncRNA-HNF1A-AS1 Genes and Risk of Multiple Sclerosis. J. Mol. Neurosci. 2020, 70, 1050–1055. [Google Scholar] [CrossRef]
  182. Perianes-Cachero, A.; Lobo, M.V.T.; Hernández-Pinto, A.M.; Busto, R.; Lasunción-Ripa, M.A.; Arilla-Ferreiro, E.; Puebla-Jiménez, L. Oxidative Stress and Lymphocyte Alterations in Chronic Relapsing Experimental Allergic Encephalomyelitis in the Rat Hippocampus and Protective Effects of an Ethanolamine Phosphate Salt. Mol. Neurobiol. 2020, 57, 860–878. [Google Scholar] [CrossRef]
  183. Dimitrijević, M.; Kotur-Stevuljević, J.; Stojić-Vukanić, Z.; Vujnović, I.; Pilipović, I.; Nacka-Aleksić, M.; Leposavić, G. Sex Difference in Oxidative Stress Parameters in Spinal Cord of Rats with Experimental Autoimmune Encephalomyelitis, Relation to Neurological Deficit. Neurochem. Res. 2017, 42, 481–492. [Google Scholar] [CrossRef] [PubMed]
  184. Jhelum, P.; Zandee, S.; Ryan, F.; Zarruk, J.G.; Michalke, B.; Venkataramani, V.; Curran, L.; Klement, W.; Prat, A.; David, S. Ferroptosis induces detrimental effects in chronic EAE and its implications for progressive MS. Acta Neuropathol. Commun. 2023, 11, 121. [Google Scholar] [CrossRef] [PubMed]
  185. Tully, M.; Tang, J.; Zheng, L.; Acosta, G.; Tian, R.; Hayward, L.; Race, N.; Mattson, D.; Shi, R. Systemic Acrolein Elevations in Mice With Experimental Autoimmune Encephalomyelitis and Patients With Multiple Sclerosis. Front. Neurol. 2018, 9, 420. [Google Scholar] [CrossRef]
  186. Hu, C.L.; Nydes, M.; Shanley, K.L.; Morales Pantoja, I.E.; Howard, T.A.; Bizzozero, O.A. Reduced expression of the ferroptosis inhibitor glutathione peroxidase-4 in multiple sclerosis and experimental autoimmune encephalomyelitis. J. Neurochem. 2019, 148, 426–439. [Google Scholar] [CrossRef] [PubMed]
  187. Smerjac, S.M.; Bizzozero, O.A. Cytoskeletal protein carbonylation and degradation in experimental autoimmune encephalomyelitis. J. Neurochem. 2008, 105, 763–772. [Google Scholar] [CrossRef]
  188. Zheng, J.; Bizzozero, O.A. Accumulation of protein carbonyls within cerebellar astrocytes in murine experimental autoimmune encephalomyelitis. J. Neurosci. Res. 2010, 88, 3376–3385. [Google Scholar] [CrossRef]
  189. Dasgupta, A.; Zheng, J.; Perrone-Bizzozero, N.I.; Bizzozero, O.A. Increased carbonylation.; protein aggregation and apoptosis in the spinal cord of mice with experimental autoimmune encephalomyelitis. ASN Neuro 2013, 5, e00111. [Google Scholar] [CrossRef] [PubMed]
  190. Castegna, A.; Palmieri, L.; Spera, I.; Porcelli, V.; Palmieri, F.; Fabis-Pedrini, M.J.; Kean, R.B.; Barkhouse, D.A.; Curtis, M.T.; Hooper, D.C. Oxidative stress and reduced glutamine synthetase activity in the absence of inflammation in the cortex of mice with experimental allergic encephalomyelitis. Neuroscience 2011, 185, 97–105. [Google Scholar] [CrossRef]
  191. Emerson, M.R.; LeVine, S.M. Heme oxygenase-1 and NADPH cytochrome P450 reductase expression in experimental allergic encephalomyelitis, an expanded view of the stress response. J. Neurochem. 2000, 75, 2555–2562. [Google Scholar] [CrossRef] [PubMed]
  192. Mehindate, K.; Sahlas, D.J.; Frankel, D.; Mawal, Y.; Liberman, A.; Corcos, J.; Dion, S.; Schipper, H.M. Proinflammatory cytokines promote glial heme oxygenase-1 expression and mitochondrial iron deposition, implications for multiple sclerosis. J. Neurochem. 2001, 77, 1386–1395. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, Y.; Zhu, B.; Luo, L.; Li, P.; Paty, D.W.; Cynader, M.S. Heme oxygenase-1 plays an important protective role in experimental autoimmune encephalomyelitis. Neuroreport 2001, 12, 1841–1845. [Google Scholar] [CrossRef] [PubMed]
  194. Izawa, T.; Yamate, J.; Franklin, R.J.; Kuwamura, M. Abnormal iron accumulation is involved in the pathogenesis of the demyelinating dmy rat but not in the hypomyelinating mv rat. Brain Res. 2010, 1349, 105–114. [Google Scholar] [CrossRef] [PubMed]
  195. Mossakowski, A.A.; Pohlan, J.; Bremer, D.; Lindquist, R.; Millward, J.M.; Bock, M.; Pollok, K.; Mothes, R.; Viohl, L.; Radbruch, M.; et al. Tracking CNS and systemic sources of oxidative stress during the course of chronic neuroinflammation. Acta Neuropathol. 2015, 130, 799–814. [Google Scholar] [CrossRef] [PubMed]
  196. Ravelli, K.L.G.; Santosk, G.D.; Dos Santos, N.B.; Munhoz, C.D.; Azzi-Nogueira, D.; Campos, A.C.; Pagano, R.L.; Britto, L.R.; Hernandes, M.S. Nox2-dependent Neuroinflammation in An EAE Model of Multiple Sclerosis. Transl. Neurosci. 2019, 10, 1–9. [Google Scholar] [CrossRef]
  197. Hu, C.F.; Wu, S.P.; Lin, G.J.; Shieh, C.C.; Hsu, C.S.; Chen, J.W.; Chen, S.H.; Hong, J.S.; Chen, S.J. Microglial Nox2 Plays a Key Role in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. Front. Immunol. 2021, 12, 638381. [Google Scholar] [CrossRef] [PubMed]
  198. Hasseldam, H.; Rasmussen, R.S.; Johansen, F.F. Oxidative damage and chemokine production dominate days before immune cell infiltration and EAE disease debut. J. Neuroinflamm. 2016, 13, 246. [Google Scholar] [CrossRef] [PubMed]
  199. Steudler, J.; Ecott, T.; Ivan, D.C.; Bouillet, E.; Walthert, S.; Berve, K.; Dick, T.P.; Engelhardt, B.; Locatelli, G. Autoimmune neuroinflammation triggers mitochondrial oxidation in oligodendrocytes. Glia 2022, 70, 2045–2061. [Google Scholar] [CrossRef]
  200. Aheng, C.; Ly, N.; Kelly, M.; Ibrahim, S.; Ricquier, D.; Alves-Guerra, M.C.; Miroux, B. Deletion of UCP2 in iNOS deficient mice reduces the severity of the disease during experimental autoimmune encephalomyelitis. PLoS ONE 2011, 6, e22841. [Google Scholar] [CrossRef]
  201. Johnson, D.A.; Amirahmadi, S.; Ward, C.; Fabry, Z.; Johnson, J.A. The absence of the pro-antioxidant transcription factor Nrf2 exacerbates experimental autoimmune encephalomyelitis. Toxicol. Sci. 2010, 114, 237–246. [Google Scholar] [CrossRef] [PubMed]
  202. Honorat, J.A.; Kinoshita, M.; Okuno, T.; Takata, K.; Koda, T.; Tada, S.; Shirakura, T.; Fujimura, H.; Mochizuki, H.; Sakoda, S.; et al. Xanthine oxidase mediates axonal and myelin loss in a murine model of multiple sclerosis. PLoS ONE 2013, 8, e71329. [Google Scholar] [CrossRef] [PubMed]
  203. Lee, G.; Hasan, M.; Kwon, O.S.; Jung, B.H. Identification of Altered Metabolic Pathways during Disease Progression in EAE Mice via Metabolomics and Lipidomics. Neuroscience 2019, 416, 74–87. [Google Scholar] [CrossRef] [PubMed]
  204. Trifunovic, S.; Stevanovic, I.; Milosevic, A.; Ristic, N.; Janjic, M.; Bjelobaba, I.; Savic, D.; Bozic, I.; Jakovljevic, M.; Tesovic, K.; et al. The Function of the Hypothalamic-Pituitary-Adrenal Axis During Experimental Autoimmune Encephalomyelitis, Involvement of Oxidative Stress Mediators. Front. Neurosci. 2021, 15, 649485. [Google Scholar] [CrossRef] [PubMed]
  205. Tolmacheva, A.S.; Aulova, K.S.; Urusov, A.E.; Doronin, V.B.; Nevinsky, G.A. Antibodies-Abzymes with Antioxidant Activities in Two Th and 2D2 Experimental Autoimmune Encephalomyelitis Mice during the Development of EAE Pathology. Molecules 2022, 27, 7527. [Google Scholar] [CrossRef] [PubMed]
  206. Urusov, A.E.; Tolmacheva, A.S.; Aulova, K.S.; Nevinsky, G.A. Autoantibody-Abzymes with Catalase Activity in Experimental Autoimmune Encephalomyelitis Mice. Molecules 2023, 28, 1330. [Google Scholar] [CrossRef] [PubMed]
  207. Vidaurre, O.G.; Haines, J.D.; Katz Sand, I.; Adula, K.P.; Huynh, J.L.; McGraw, C.A.; Zhang, F.; Varghese, M.; Sotirchos, E.; Bhargava, P.; et al. Cerebrospinal fluid ceramides from patients with multiple sclerosis impair neuronal bioenergetics. Brain 2014, 137, 2271–2286. [Google Scholar] [CrossRef] [PubMed]
  208. Zhang, S.Y.; Gui, L.N.; Liu, Y.Y.; Shi, S.; Cheng, Y. Oxidative Stress Marker Aberrations in Multiple Sclerosis, A Meta-Analysis Study. Front. Neurosci. 2020, 14, 823. [Google Scholar] [CrossRef]
  209. Thompson, A.J.; Banwell, B.L.; Barkhof, F.; Carroll, W.M.; Coetzee, T.; Comi, G.; Correale, J.; Fazekas, F.; Filippi, M.; Freedman, M.S.; et al. Diagnosis of multiple sclerosis, 2017 revisions of the McDonald criteria. Lancet Neurol. 2018, 17, 162–173. [Google Scholar] [CrossRef]
  210. Kurtzke, J.F. Rating neurologic impairment in multiple sclerosis, an expanded disability status scale (EDSS). Neurology 1983, 33, 1444–1452. [Google Scholar] [CrossRef]
Figure 1. Possible interactions between the different pathogenetic mechanisms in multiple sclerosis.
Figure 1. Possible interactions between the different pathogenetic mechanisms in multiple sclerosis.
Ijms 25 06289 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jiménez-Jiménez, F.J.; Alonso-Navarro, H.; Salgado-Cámara, P.; García-Martín, E.; Agúndez, J.A.G. Oxidative Stress Markers in Multiple Sclerosis. Int. J. Mol. Sci. 2024, 25, 6289. https://doi.org/10.3390/ijms25126289

AMA Style

Jiménez-Jiménez FJ, Alonso-Navarro H, Salgado-Cámara P, García-Martín E, Agúndez JAG. Oxidative Stress Markers in Multiple Sclerosis. International Journal of Molecular Sciences. 2024; 25(12):6289. https://doi.org/10.3390/ijms25126289

Chicago/Turabian Style

Jiménez-Jiménez, Félix Javier, Hortensia Alonso-Navarro, Paula Salgado-Cámara, Elena García-Martín, and José A. G. Agúndez. 2024. "Oxidative Stress Markers in Multiple Sclerosis" International Journal of Molecular Sciences 25, no. 12: 6289. https://doi.org/10.3390/ijms25126289

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