Next Article in Journal
PolyX2: Fast Detection of Homorepeats in Large Protein Datasets
Next Article in Special Issue
New Insights into the Neuromyogenic Spectrum of a Gain of Function Mutation in SPTLC1
Previous Article in Journal
Macrophage Polarization in Atherosclerosis
Previous Article in Special Issue
High Concentration or Combined Treatment of Antisense Oligonucleotides for Spinal Muscular Atrophy Perturbed SMN2 Splicing in Patient Fibroblasts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Variability of Inflammation and Oxidative Stress Genes Affects Onset, Progression of the Disease and Survival of Patients with Amyotrophic Lateral Sclerosis

by
Metka Ravnik-Glavač
1,*,†,
Katja Goričar
1,†,
David Vogrinc
1,
Blaž Koritnik
2,3,
Jakob Gašper Lavrenčič
4,
Damjan Glavač
4,5 and
Vita Dolžan
1
1
Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
2
Institute of Clinical Neurophysiology, Division of Neurology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
3
Department of Neurology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
4
Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
5
Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2022, 13(5), 757; https://doi.org/10.3390/genes13050757
Submission received: 15 March 2022 / Revised: 20 April 2022 / Accepted: 24 April 2022 / Published: 25 April 2022
(This article belongs to the Special Issue Genetics of Motor Neuron Diseases)

Abstract

:
Inflammation and oxidative stress are recognized as important contributors to amyotrophic lateral sclerosis (ALS) disease pathogenesis. Our aim was to evaluate the impact of selected single-nucleotide polymorphisms in genes involved in inflammation and oxidative stress on ALS susceptibility and modification. One-hundred-and-eighty-five ALS patients and 324 healthy controls were genotyped for nine polymorphisms in seven antioxidant and inflammatory genes using competitive allele-specific PCR. Logistic regression; nonparametric tests and survival analysis were used in the statistical analysis. Investigated polymorphisms were not associated with ALS susceptibility. Carriers of at least one polymorphic SOD2 rs4880 T or IL1B rs1071676 C allele more often had bulbar ALS onset (p = 0.036 and p = 0.039; respectively). IL1B rs1071676 was also associated with a higher rate of disease progression (p = 0.015). After adjustment for clinical parameters; carriers of two polymorphic IL1B rs1071676 C alleles had shorter survival (HR = 5.02; 95% CI = 1.92–13.16; p = 0.001); while carriers of at least one polymorphic CAT rs1001179 T allele had longer survival (HR = 0.68; 95% CI = 0.47–0.99; p = 0.046). Our data suggest that common genetic variants in the antioxidant and inflammatory pathways may modify ALS disease. Such genetic information could support the identification of patients that may be responsive to the immune or antioxidant system—based therapies.

1. Introduction

Amyotrophic lateral sclerosis (ALS) is characterized by progressive degeneration of both upper and lower motor neurons, resulting in muscle weakness, atrophy, and gradual paralysis, leading to death due to respiratory failure usually within 2–3 years following the onset of symptoms [1]. The disease most typically begins in the limbs (spinal onset), but in approximately 20% of cases, oropharyngeal muscles weakness (bulbar onset) is the first evident symptom of the disease [2]. ALS has an incidence of 2–3 per 100,000 and a lifetime risk of 1 per 400 individuals [3], with some differences between populations [4].
About 5–10% of ALS cases have a familial background (FALS), but the vast majority are categorized as sporadic ALS (SALS) based on negative family history. FALS and SALS are mostly phenotypically indistinguishable, suggesting common pathways exist at the basis of neuronal death. In SALS, the lack of positive family history for ALS could be due to incomplete penetrance or oligogenic/polygenic inheritance [5].
To date, several genetic factors have been identified that drive motor neuron degeneration in ALS, increase susceptibility to the disease, or influence the rate of progression [6,7]. In the ALS database, around 150 genes have been reported that are, according to their contribution to ALS pathogenesis, classified as definitive ALS genes, clinical modifiers, and genes with strong or moderate evidence (https://alsod.ac.uk/. accessed on 15 March 2022). Although more than 30 of these genes strongly correlate with the disease, their exact roles in disease pathogenesis are still not completely understood [8]. The main proposed interrupted mechanisms by most genes mutated in ALS include disrupted proteostasis by increased protein aggregation (SOD1, TARDBP, FUS, C9orf72) decreased proteasomal degradation (VCP, UBQLN2), or impaired autophagy (OPTN, TBK1, CYLD, C9orf72, ALS2, SQSTM1, VCP, UBQLN2, CHMP2B). Several of these genes have also multiple functions. For example, mutations in SOD1 and SQSTM1 can lead to mitochondrial damage and oxidative stress [8].
Oxidative stress is a process where an accumulation of reactive oxygen species (ROS) leads to cellular damage and cell death due to an imbalance between free radical production and antioxidant defenses [9,10,11]. Cellular ROS levels may be reduced through the defense mechanisms of antioxidant enzymes [12]. Of these, manganese SOD (SOD2) is mainly involved in the elimination of highly reactive O2 in the cytosol and mitochondria to produce H2O2 [13]. Then H2O2 may be further removed by the action of glutathione peroxidases (GPX) and catalase (CAT) [14]. Studies have suggested that upregulation of GPX1 could be one of the protective responses against neuronal injury [14]. CAT is located mainly in peroxisomes and is responsible for the conversion of H2O2 to water and oxygen [12,13]. The contribution of catalase to the oxidative stress response is minor at low levels of H2O2 but becomes increasingly important at higher levels of H2O2 [12,15].
Oxidative stress is thought to increase with age, a major risk factor in ALS [16]. High levels of ROS can damage several different parts of the cellular machinery through lipid peroxidation and oxidation of proteins and/or DNA. In SOD1 G93A rodent ALS models as well as cell ALS models, researchers found oxidative damage to DNA, RNA, proteins, and lipids [17]. Markers of ROS damage have also been reported to be increased in cerebrospinal fluid, plasma, serum, and urine of SALS patients [18,19,20,21] and in post-mortem ALS spinal cord tissue [11,22,23,24].
Excessive production of ROS can result in reduced efficiency of cellular processes, excitotoxicity, protein aggregation and endoplasmic reticulum stress, and induction of inflammatory pathways that have all been directly involved in disease pathogenesis [9,15]. Inflammation/neuroinflammation has also been recognized as an important mediator of the pathogenesis of disease in ALS [25]. Inflammation can be triggered by aggregated proteins and impaired autophagy and/or oxidative stress and mitochondrial damage. The link between inflammation and other proposed ALS-related mechanisms is thus complex and multidirectional and can at some point initiate a vicious cycle of a pathogenic cascade of molecular events that gradually damages motor neurons to the point that cell deterioration is irreversible [8,26]. In addition, mutations in several genes that have been reported in ALS patients, including TBK1, OPTN, CYLD, and C9orf72, are directly linked to the immune response [8,27,28,29,30].
Several studies indicated that deregulation of immune response may in many cases occur early in the course of ALS. Namely, in mouse ALS models longitudinal live imaging studies revealed, in the very early presymptomatic stages of the disease, significant changes in activation of astrocytes and microglia [31,32]. Microglia are a central protagonist of the neuroinflammatory component of neurodegeneration in ALS. Macrophages from activated microglia are largely pro-inflammatory and secrete a number of proinflammatory cytokines such as tumor necrosis factor α (TNF-α), interferon γ (IFNγ), and interleukin 1β (IL-1β) [25]. In animal models, specific deletion of C9orf72 from myeloid cells in mice (specifically macrophages and microglia) resulted in lysosomal accumulation, hyperactive immune responses, and increased expression of cytokines IL-6 and IL-1β, altering the normal immune function of these cells [33].
Increasing evidence suggests that inflammatory response in the central nervous system (CNS) that includes proinflammatory cytokines contributes to the pathogenesis of ALS also in human patients [34]. Positron emission tomography imaging studies together with studies of proteins expressed by activated microglia revealed active gliosis in patients with ALS in vivo and demonstrated widespread microglial activation in the motor cortex, dorsolateral prefrontal cortex, and thalamus [35,36,37]. Increased microgliosis correlated positively with Upper Motor Neuron scores and negatively with ALS Functional Rating Scores (ALS-FRS) [35,38] thus indicating a positive relationship between neuroinflammation and disease severity. Infiltration of immune cells was observed in the CNS of ALS patients at sites of motor neuron injury, including macrophages and T-cells [39,40,41]. In addition, peripheral immune abnormalities including T-cells, cytokines, chemokines, and other markers of inflammation have been detected in blood in clinical studies [42]. ALS monocytes demonstrated a unique inflammation-related gene expression profile, including increased IL1B and IL8 expression [43].
Recent work further suggests that microglial release of inflammatory factors serves as a trigger for activated neurotoxic astrocytes [44]. Additionally, some miRNAs have been found to be involved in this process. For example, miR-146a was upregulated in IL-1β-stimulated human astrocytes and was associated with the regulation of an astrocyte-mediated inflammatory response [45]. Downregulation of miR-146a affected TLR/NF-κB signaling pathways in murine SOD1 astrocytes, thus contributing to neuroinflammation [46]. Banack et al. recently reported the upregulation of miR-146a-5p in the neural enriched extracellular vesicles of ALS patient samples compared to healthy controls [47].
The potential causative or disease driving role of inflammation and oxidative stress in ALS is not fully understood, as there are studies that support both a primary and secondary role during ALS disease progression [9,48]. Genetic factors can affect both inflammation and response to oxidative stress, but the role of common polymorphisms in ALS susceptibility or pathogenesis has not been well established. In this study, we aimed to further elucidate the role of inflammation and oxidative stress in the pathogenesis of ALS by genotyping ALS patients and controls for common genetic polymorphisms in genes involved in antioxidant (SOD2, GPX, and CAT) and inflammatory pathways (IL1B, TNF, IL6, MIR146A).

2. Subjects and Methods

2.1. Subjects

The patient cohort consisted of 185 SALS patients diagnosed and collected between 2012 and 2019 at the tertiary Ljubljana ALS Centre which takes care of the majority of Slovenian ALS patients. Patients’ age was 56 to 71 years and there were 94 male and 91 female (Table 1). In our previous study, we screened 85 of these SALS patients for common mutations in four major ALS-associated genes, SOD1, TARDBP, FUS, and C9orf72 [49]. Functional impairment of the patients was assessed routinely by the ALS-FRS-R during the out-patient visits every three months. ALS-FRS-R data were not available for 21 patients who did not have an out-patient visit at the time of blood collection. We obtained approval for this study from the National Medical Ethics Committee of the Republic of Slovenia (Approval number: 68/12/13 and 0120-120/2018/8) and all participants provided written informed consent. The Control group consisted of 324 unrelated healthy Slovenian blood donors without any systemic disease aged 40 to 65 years, 238 were male, and 86 were female.

2.2. DNA Extraction and Genotyping

DNA from patients’ peripheral venous blood samples was isolated using QIAamp Blood Midi Kit (Qiagen, Hilden, Germany) following the manufacturer’s protocol. For healthy controls, genomic DNA was isolated from peripheral venous blood samples using Qiagen FlexiGene Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions.
Genotyping was performed for nine single nucleotide polymorphisms (SNPs) with known or predicted functional effects across seven genes important in antioxidant and inflammatory pathways [50]. SOD2 (rs4880), CAT (rs1001179), GPX1 (rs1050450), IL1B (rs1143623, rs16944, rs1071676), MIR146A (rs2910164), IL6 (rs1800795) and TNF (rs1800629) were genotyped using a fluorescent-based, competitive allele-specific polymerase chain reaction (KASP, LGC Genomics, Hoddesdon, UK) according to the manufacturer’s protocol. For quality control, 10% of samples were genotyped in duplicate and all results were concordant.
Genotype frequencies of all nine investigated SNPs in SOD2, CAT, GPX1, IL1B, IL6, and TNF genes among ALS patients and healthy controls are presented in Supplementary Table S1. Genotype frequencies of all investigated SNPs were in agreement with Hardy-Weinberg Equilibrium in both controls and ALS patients.

2.3. Statistical Analysis

Continuous variables were described with median and interquartile range (25–75%), while categorical variables were described with frequencies. Deviation from the Hardy-Weinberg equilibrium (HWE) was evaluated using the chi-square test. Both dominant and additive genetic models were used in the analysis. Logistic regression was used to evaluate the association of selected SNPs with binary categorical variables and to determine the odds ratios (ORs) and their 95% confidence intervals (CIs). Fisher’s exact test was used if there were no subjects within one of the groups or for dependent categorical variables with more than two categories, as well as to compare different subject groups. Nonparametric Mann-Whitney or Kruskal-Wallis test with post hoc Bonferroni corrections for pairwise comparisons were used to evaluate the association of SNPs with continuous variables.
Survival of ALS patients was defined as the time from disease onset to death. Patients without death at the time of the analysis were censored at the date of the last follow-up. Kaplan-Meier test was used to calculate median survival or follow-up time, while Cox regression was used to assess the role of selected SNPs and to calculate the hazard ratios (HR) with their 95% CIs. Clinical parameters used for adjustment in multivariable regression models were selected using stepwise forward-conditional regression. IBM SPSS Statistics version 27.0 (IBM Corporation, Armonk, NY, USA) was used for all analyses. All tests were two-sided, and the level of significance was set at 0.05.

3. Results

We included in our study 185 ALS patients and 324 healthy controls. The level of functional impairment according to ALS-FRS-R data was available for 164 ALS patients. Clinical characteristics of all ALS patients and patients with available ALS-FRS-R data are presented in Table 1. Most patients had spinal onset ALS (133, 72.3%) and 153 (86.0%) died by the time of the analysis. C9orf72 mutation was found in six (3.2%) patients. Median survival was 39.7 (26.6–68.4) months and median follow-up was 104.9 (90.7–156.6) months.
Among healthy controls, 238 (73.5%) were male and 86 (26.5%) female. The gender distribution differed significantly between healthy controls and ALS patients (p = 0.001). The median age of healthy controls was 49 (44–55) years, which was significantly younger compared to ALS patients (p < 0.001).

3.1. Association of Investigated SNPs with ALS Susceptibility and ALS Type

None of the investigated SNPs was associated with ALS susceptibility, not even after adjustment for gender and age (Table 2).
On the other hand, some of the genotype frequencies differed between groups of patients with spinal, bulbar, and other ALS types at disease onset (Table 3). Carriers of at least one polymorphic SOD2 rs4880 T allele more often had bulbar onset ALS when compared to carriers of two C alleles that had more often spinal onset ALS (p = 0.036). Similarly, bulbar onset ALS was more frequent in carriers of at least one polymorphic IL1B rs1071676 C allele compared to carriers of two G alleles (p = 0.039). Conversely, spinal onset ALS was more frequent in carriers of at least one polymorphic IL1B rs16944 C allele, while other ALS types were more common in carriers of two T alleles, but the difference did not reach statistical significance (p = 0.051).

3.2. Association of Investigated SNPs with Level of Functional Impairment and Rate of Disease Progression in ALS Patients

Among the investigated SNPs, only IL1B rs1071676 was associated with the rate of disease progression (p = 0.015, Table 4, Figure 1). After adjustment for multiple comparisons, carriers of two polymorphic IL1B rs1071676 C alleles had a higher rate of disease progression compared to carriers of two G alleles (Padj = 0.015) and heterozygotes (Padj = 0.012).
However, none of the investigated SNPs was associated with a level of functional impairment in ALS patients in our study (Table 4).

3.3. Association of Investigated SNPs with Survival of ALS Patients

Among the investigated clinical parameters, shorter survival was observed in older patients (HR = 1.04, 95% CI = 1.02–1.06, p < 0.001). Bulbar onset ALS (HR = 1.85, 95% CI = 1.27–2.69, p = 0.001) and other onset ALS types (HR = 3.19, 95% CI = 1.38–7.35, p = 0.007) were associated with shorter survival compared to spinal onset ALS. Slower rate of disease progression was also significantly associated with longer survival (HR = 0.60, 95% CI = 0.53–0.68, p < 0.001). In forward conditional analysis, age at onset (HR = 1.04, 95% CI = 1.02–1.05, p < 0.001) and rate of disease progression (HR = 0.63, 95% CI = 0.56–0.72, p < 0.001) remained associated with survival and were used for adjustment in multivariable models.
In univariable analysis, none of the investigated SNPs was significantly associated with the survival of ALS patients (Table 5). Carriers of at least one polymorphic CAT rs1001179 T allele had longer survival compared to carriers of two C alleles (47.6 months compared to 37.0 months), but the association was only significant after adjustment for clinical parameters (HR = 0.68, 95% CI = 0.47–0.99, p = 0.046; Figure 2A). Carriers of two polymorphic IL1B rs1071676 C alleles had shorter survival compared to carriers of two G alleles (24.3 months compared to 39.7 months), but the association was only significant after adjustment for clinical parameters (HR = 5.02, 95% CI = 1.92–13.16, p = 0.001; Figure 2B).

4. Discussion

Inflammation/neuroinflammation and oxidative stress are important mediators of the pathogenesis of disease in ALS and have both been reported to be involved in the early stages of the disease [8,9]. However, results that indicate a causative or significant role of oxidative stress and immune system components in ALS pathogenesis are limited and more results support their involvement in exacerbating the disease progression [9,48,51]. Although oxidative stress and inflammation are probably not the main triggers of the disease, an important challenge to ALS research is also to find out how endogenous genetic variants of inflammation and antioxidant factors modify the disease that accounts for different disease courses. To further elucidate this challenging topic, we have genotyped a representative cohort of SALS patients and controls for common genetic polymorphisms in genes involved in antioxidant and inflammatory pathways. In the present study, IL1B rs1071676 polymorphism was associated with disease onset, rate of disease progression, and survival of ALS patients. Selected polymorphisms in antioxidant genes were also associated with some clinical characteristics, while no association with ALS susceptibility was observed. To our knowledge, these selected polymorphisms have not been previously studied in connection with ALS.
Our results revealed that none of the investigated SNPs was associated with ALS disease risk. Although there are few studies on how polymorphisms in oxidative stress response and inflammatory pathways influence ALS pathogenesis, they are all in agreement with our results. In previous studies investigating antioxidant genes, mutation, or individual common polymorphisms in SOD2, glutathione S-transferase P1 (GSTP1) or paraoxonase 1 (PON1) genes were not associated with ALS susceptibility [52,53,54]. Regarding inflammatory processes, previous studies did not investigate genetic variability in the genes included in our study. On the other hand, functional variants of the human CX3CR1 gene, also known as fractalkine receptor, were also not associated with ALS disease risk [55,56]. Fractalkine signaling affects inflammatory responses and has been discovered to play a role in the migration of microglia to their synaptic targets, where phagocytosis and synaptic refinement occur during the development of the central nervous system [57].
The key result of our study is the association of IL1B rs1071676 with several different patients’ characteristics: disease onset, rate of disease progression, and survival of ALS patients. On the other hand, no significant associations with ALS were observed for common IL1B promotor polymorphisms rs1143623 and rs16944.
IL1B rs1071676 occurs in the 3′-UTR region (c.*505G > C) of the gene and is predicted to alter the miRNA binding site with potential functional effect [58]. In our study, carriers of two polymorphic IL1B rs1071676 C alleles had a higher rate of disease progression compared to carriers of two G alleles and heterozygotes. Similarly, carriers of two polymorphic IL1B rs1071676 C alleles had a 15.4-month shorter median survival compared to carriers of two G alleles, but the association was only significant after adjustment for clinical parameters. In addition, carriers of at least one polymorphic IL1B rs1071676 C allele had more bulbar onset of ALS. Consistently, patients with bulbar onset had shorter survival compared to patients with spinal ALS onset. To the best of our knowledge, this polymorphism was previously not investigated in ALS and its role is not well known. However, it was previously already associated with brain patterns after hypoxic-ischemic encephalopathy [59], fatigue in adults with HIV/AIDS [60], and response to anti-TNF therapy in Crohn’s disease [58]. Further studies are therefore needed to elucidate the role of this polymorphism in IL-1β expression.
On the other hand, the role of IL-1β in ALS was already previously reported. The NLRP3 inflammasome is required for the activation of caspase 1 and the processing and release of IL-1β and IL-18 [61]. Johann et al. investigated the expression of the NLRP3 inflammasome in SOD1 mutated ALS animal model and in post-mortem tissue of ALS patients [61]. They detected NLRP3 and IL-1β in spinal cord astrocytes already at a pre-symptomatic stage in SOD1 mutant mice. NLRP3 components and active caspase 1 levels were also increased in human ALS tissue compared to controls, which suggested that astroglial NLRP3 inflammasome complexes may contribute to neuroinflammation in ALS [61]. In SOD1 mutated mice models, an increase in IL-1β levels was consistently reported [62,63,64], further suggesting an association with disease progression through the promotion of neuroinflammation [62]. IL-1β was even proposed as a potential therapeutic target [62]. However, the results of studies evaluating blood or cerebrospinal fluid IL-1β in ALS patients were not always consistent [65,66,67]. Still, several studies observed an increase of IL-1β in ALS patients, even though its levels were not always above the limit of detection [65,66]. Importantly, increased IL-1β was observed in ALS patients with short survival, and increased expression was associated with shorter survival, especially in carriers of C9orf72 mutation [65]. These results are consistent with our results, suggesting IL-1β variability should be further examined in larger studies.
Among polymorphisms in antioxidant genes, we observed in our cohort differences in SOD2 rs4880 genotype frequencies distribution between spinal, bulbar, and other ALS onsets. Carriers of at least one polymorphic SOD2 rs4880 T allele more often had bulbar onset of ALS compared to carriers of two C alleles that had more spinal onset of ALS. This single C/T nucleotide polymorphism is the most characterized polymorphism in the signal sequence of the SOD2 gene that results in p.Ala16Val of the mitochondrial targeting sequence in the SOD2 protein. It was predicted that the valine-containing protein (rs4880 T allele) would form a β-sheet rather than the expected α-helix of alanine-containing protein (rs4880 C allele). It was shown, consistent with this prediction, that the p.16Val SOD2 had reduced import efficiency into the mitochondria as well as reduced mRNA stability and activity in the mitochondria, which in turn both increase oxidative stress [68,69,70]. It was also predicted that the association of SOD2 rs4880 gene polymorphism with various different diseases may be due to differential modulation of oxidative stress with other endogenous as well as exogenous antioxidants and may thus be partly influenced by environmental factors, like physical activity and diet [68,70,71]. However, further studies are needed to better evaluate the role of SOD2 polymorphisms in ALS. Verde et al. [72] reported similar results for rs662 (Q192R) in detoxifying enzyme PON1. They found PON1 minor allele G to be associated both with the bulbar onset and independently with reduced survival. Q192R modifies enzyme activity with regard to substrate specificity and catalytic efficiency. Authors hypothesized that 192R alloenzyme is less efficient in ameliorating certain endogenous damaging processes such as oxidative stress or in metabolizing some exogenous toxic substances [72].
Catalase plays a significant role in protecting cells against severe oxidative stress [12]. In our study, carriers of at least one polymorphic CAT rs1001179 T allele had significantly longer survival compared to carriers of two C alleles after adjustment for clinical parameters. This common C/T polymorphism is located 262 base pairs upstream from the transcription start site of the catalase gene and affects transcription factor binding and alters gene expression [73,74]. In blood samples, catalase levels were significantly higher in donors carrying the T allele in comparison to donors homozygous for the C allele [73], in concordance with our study, where this allele was associated with longer survival. Studies investigating this polymorphism in neurodegenerative diseases are scarce [75], while several associations were observed with other diseases, especially cancer [76,77]. On the other hand, catalase activity was consistently decreased in erythrocytes of ALS patients [78,79,80], suggesting its important role in oxidative stress response; however, further studies are needed to validate the role of catalase in ALS.
We were the first to comprehensively evaluate the role of common polymorphisms in antioxidant and inflammation genes in ALS susceptibility and pathogenesis. Our study group reflects well the characteristics of SALS patients. C9orf72 mutation was found in 3.2% of patients, similar to other studies [81]. Investigated clinical parameters in our study revealed shorter survival in older patients and shorter survival of patients with bulbar onset compared to spinal ALS onset, consistent with previous studies [82]. However, our study also has some limitations, mainly that ALS-FRS-R data were not available in all patients. Additionally, as C9orf72 mutations are rare in SALS, we could not evaluate the role of investigated polymorphisms in carriers of this mutation separately. Another limitation was that controls were younger than ALS patients as only individuals younger than 65 years can serve as blood donors and that only data on gender and age were available for the control group. However, the possible relevance of the diverse age of the ALS patients and controls for the clinical course of the disease was taken into account while performing the adjustment in logistic regression analysis.
Taken together, we have shown for the first time that some of the selected studied polymorphisms in genes involved in inflammation (IL1B rs1071676) and oxidative stress (SOD2 rs4880, CAT rs1001179), could be disease modifiers in ALS and that common genetic variants can influence ALS onset, rate of progression and survival. These types of studies are important to identify specific patient populations that may be responsive to the immune or antioxidant system—based therapies and to motivate future large-scale genetic research on modifiers of ALS progression for potential new treatments for ALS.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes13050757/s1, Table S1: Genotype frequencies of selected polymorphisms.

Author Contributions

Data curation, writing—original draft preparation, review & editing and supervision M.R.-G.; data curation, writing—original draft preparation K.G.; formal analysis, data curation D.V.; data curation, resources B.K.; formal analysis J.G.L.; supervision and funding acquisition D.G.; conceptualization and funding acquisition V.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Slovenian Research Agency (ARRS) under research core funding Nos. P3-0054, P1-0170 and P3-0338.

Institutional Review Board Statement

The study was approved by the National Medical Ethics Committee of the Republic of Slovenia. All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed Consent Statement

Informed consent was obtained from all individual participants included in the study.

Data Availability Statement

The datasets during and/or analyzed during the current study available from the corresponding author on reasonable request.

Acknowledgments

We thank all of the patients who were willing to cooperate in this research and made it possible. We thank Blood Transfusion Centre of Slovenia for collecting healthy controls.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rowland, L.P.; Shneider, N.A. Amyotrophic Lateral Sclerosis. N. Engl. J. Med. 2001, 344, 1688–1700. [Google Scholar] [CrossRef]
  2. Ringel, S.P.; Murphy, J.R.; Alderson, M.K.; Bryan, W.; England, J.D.; Miller, R.G.; Petajan, J.H.; Smith, S.A.; Roelofs, R.I.; Ziter, F.; et al. The natural history of amyotrophic lateral sclerosis. Neurology 1993, 43, 1316. [Google Scholar] [CrossRef]
  3. Brown, R.H.; Al-Chalabi, A. Amyotrophic Lateral Sclerosis. N. Engl. J. Med. 2017, 377, 162–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Chiò, A.; Logroscino, G.; Traynor, B.; Collins, J.; Simeone, J.; Goldstein, L.; White, L. Global Epidemiology of Amyotrophic Lateral Sclerosis: A Systematic Review of the Published Literature. Neuroepidemiology 2013, 41, 118–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Brenner, D.; Weishaupt, J.H. Update on amyotrophic lateral sclerosis genetics. Curr. Opin. Neurol. 2019, 32, 735–739. [Google Scholar] [CrossRef]
  6. Taylor, J.P.; Brown, R.H., Jr.; Cleveland, D.W. Decoding ALS: From genes to mechanism. Nature 2016, 539, 197–206. [Google Scholar] [CrossRef] [Green Version]
  7. White, M.A.; Sreedharan, J. Amyotrophic lateral sclerosis: Recent genetic highlights. Curr. Opin. Neurol. 2016, 29, 557–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Béland, L.-C.; Markovinovic, A.; Jakovac, H.; De Marchi, F.; Bilic, E.; Mazzini, L.; Kriz, J.; Munitic, I. Immunity in amyotrophic lateral sclerosis: Blurred lines between excessive inflammation and inefficient immune responses. Brain Commun. 2020, 2, fcaa124. [Google Scholar] [CrossRef]
  9. Obrador, E.; Salvador-Palmer, R.; López-Blanch, R.; Jihad-Jebbar, A.; Vallés, S.; Estrela, J. The Link between Oxidative Stress, Redox Status, Bioenergetics and Mitochondria in the Pathophysiology of ALS. Int. J. Mol. Sci. 2021, 22, 6352. [Google Scholar] [CrossRef]
  10. Jurcau, A. Insights into the Pathogenesis of Neurodegenerative Diseases: Focus on Mitochondrial Dysfunction and Oxidative Stress. Int. J. Mol. Sci. 2021, 22, 1847. [Google Scholar] [CrossRef]
  11. Harley, J.; Clarke, B.; Patani, R. The Interplay of RNA Binding Proteins, Oxidative Stress and Mitochondrial Dysfunction in ALS. Antioxidants 2021, 10, 552. [Google Scholar] [CrossRef] [PubMed]
  12. Gandhi, S.; Abramov, A.Y. Mechanism of oxidative stress in neurodegeneration. Oxid. Med. Cell. Longev. 2012, 2012, 428010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Dasuri, K.; Zhang, L.; Keller, J.N. Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free Radic. Biol. Med. 2013, 62, 170–185. [Google Scholar] [CrossRef] [PubMed]
  14. Power, J.H.T.; Blumbergs, P.C. Cellular glutathione peroxidase in human brain: Cellular distribution, and its potential role in the degradation of Lewy bodies in Parkinson’s disease and dementia with Lewy bodies. Acta Neuropathol. 2008, 117, 63–73. [Google Scholar] [CrossRef] [PubMed]
  15. Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef]
  16. Chang, C.-K.; Chiang, M.-H.; Toh, E.K.-W.; Chang, C.-F.; Huang, T.-H. Molecular mechanism of oxidation-induced TDP-43 RRM1 aggregation and loss of function. FEBS Lett. 2013, 587, 575–582. [Google Scholar] [CrossRef] [Green Version]
  17. Barber, S.C.; Shaw, P.J. Oxidative stress in ALS: Key role in motor neuron injury and therapeutic target. Free Radic. Biol. Med. 2010, 48, 629–641. [Google Scholar] [CrossRef]
  18. Tohgi, H.; Abe, T.; Yamazaki, K.; Murata, T.; Ishizaki, E.; Isobe, C. Remarkable increase in cerebrospinal fluid 3-nitrotyrosine in patients with sporadic amyotrophic lateral sclerosis. Ann. Neurol. 1999, 46, 129–131. [Google Scholar] [CrossRef]
  19. Bogdanov, M.; Brown, R.H.; Matson, W.; Smart, R.; Hayden, D.; O’Donnell, H.; Beal, M.F.; Cudkowicz, M. Increased oxidative damage to DNA in ALS patients. Free Radic. Biol. Med. 2000, 29, 652–658. [Google Scholar] [CrossRef]
  20. Mitsumoto, H.; Santella, R.M.; Liu, X.; Bogdanov, M.; Zipprich, J.; Wu, H.-C.; Mahata, J.; Kilty, M.; Bednarz, K.; Bell, D.; et al. Oxidative stress biomarkers in sporadic ALS. Amyotroph. Lateral Scler. 2008, 9, 177–183. [Google Scholar] [CrossRef]
  21. Simpson, E.P.; Henry, Y.K.; Henkel, J.S.; Smith, R.G.; Appel, S.H. Increased lipid peroxidation in sera of ALS patients: A potential biomarker of disease burden. Neurology 2004, 62, 1758–1765. [Google Scholar] [CrossRef] [PubMed]
  22. Shibata, N.; Nagai, R.; Uchida, K.; Horiuchi, S.; Yamada, S.; Hirano, A.; Kawaguchi, M.; Yamamoto, T.; Sasaki, S.; Kobayashi, M. Morphological evidence for lipid peroxidation and protein glycoxidation in spinal cords from sporadic amyotrophic lateral sclerosis patients. Brain Res. 2001, 917, 97–104. [Google Scholar] [CrossRef]
  23. Fitzmaurice, P.; Shaw, I.C.; Kleiner, H.E.; Miller, R.T.; Monks, T.J.; Lau, S.S.; Mitchell, J.D.; Lynch, P.G. Evidence for DNA damage in amyotrophic lateral sclerosis. Muscle Nerve 1996, 19, 797–798. [Google Scholar] [PubMed]
  24. Ferrante, R.J.; Browne, S.E.; Shinobu, L.A.; Bowling, A.C.; Baik, M.J.; MacGarvey, U.; Kowall, N.W.; Brown, R.H., Jr.; Beal, M.F. Evidence of Increased Oxidative Damage in Both Sporadic and Familial Amyotrophic Lateral Sclerosis. J. Neurochem. 1997, 69, 2064–2074. [Google Scholar] [CrossRef]
  25. Hooten, K.G.; Beers, D.R.; Zhao, W.; Appel, S.H. Protective and Toxic Neuroinflammation in Amyotrophic Lateral Sclerosis. Neurotherapeutics 2015, 12, 364–375. [Google Scholar] [CrossRef] [Green Version]
  26. Smith, E.F.; Shaw, P.; De Vos, K.J. The role of mitochondria in amyotrophic lateral sclerosis. Neurosci. Lett. 2019, 710, 132933. [Google Scholar] [CrossRef]
  27. Maruyama, H.; Morino, H.; Ito, H.; Izumi, Y.; Kato, H.; Watanabe, Y.; Kinoshita, Y.; Kamada, M.; Nodera, H.; Suzuki, H.; et al. Mutations of optineurin in amyotrophic lateral sclerosis. Nature 2010, 465, 223–226. [Google Scholar] [CrossRef]
  28. Cirulli, E.T.; Lasseigne, B.N.; Petrovski, S.; Sapp, P.C.; Dion, P.A.; Leblond, C.S.; Couthouis, J.; Lu, Y.-F.; Wang, Q.; Krueger, B.J.; et al. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science 2015, 347, 1436–1441. [Google Scholar] [CrossRef] [Green Version]
  29. Freischmidt, A.; Wieland, T.; Richter, B.; Ruf, W.P.; Schaeffer, V.; Müller, K.; Marroquin, N.; Nordin, F.; Hübers, A.; Weydt, P.; et al. Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat. Neurosci. 2015, 18, 631–636. [Google Scholar] [CrossRef]
  30. Dobson-Stone, C.; Hallupp, M.; Shahheydari, H.; Ragagnin, A.M.G.; Chatterton, Z.; Carew-Jones, F.; Shepherd, C.E.; Stefen, H.; Paric, E.; Fath, T.; et al. CYLD is a causative gene for frontotemporal dementia—Amyotrophic lateral sclerosis. Brain 2020, 143, 783–799. [Google Scholar] [CrossRef]
  31. Gravel, M.; Béland, L.-C.; Soucy, G.; Abdelhamid, E.; Rahimian, R.; Gravel, C.; Kriz, J. IL-10 Controls Early Microglial Phenotypes and Disease Onset in ALS Caused by Misfolded Superoxide Dismutase 1. J. Neurosci. 2016, 36, 1031–1048. [Google Scholar] [CrossRef]
  32. Keller, A.F.; Gravel, M.; Kriz, J. Treatment with minocycline after disease onset alters astrocyte reactivity and increases microgliosis in SOD1 mutant mice. Exp. Neurol. 2011, 228, 69–79. [Google Scholar] [CrossRef]
  33. O’Rourke, J.G.; Bogdanik, L.; Yáñez, A.; Lall, D.; Wolf, A.J.; Muhammad, A.K.M.G.; Ho, R.; Carmona, S.; Vit, J.P.; Zarrow, J.; et al. C9orf72 is required for proper macrophage and microglial function in mice. Science 2016, 351, 1324–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Calvo, A.; Moglia, C.; Balma, M.; Chio, A. Involvement of immune response in the pathogenesis of amyotrophic lateral sclerosis: A therapeutic opportunity? CNS Neurol. Disord.-Drug Targets 2010, 9, 325–330. [Google Scholar] [CrossRef] [PubMed]
  35. Turner, M.R.; Cagnin, A.; Turkheimer, F.E.; Miller, C.C.J.; Shaw, C.E.; Brooks, D.J.; Leigh, P.N.; Banati, R.B. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: An [11C](R)-PK11195 positron emission tomography study. Neurobiol. Dis. 2004, 15, 601–609. [Google Scholar] [CrossRef] [PubMed]
  36. McGeer, P.L.; McGeer, E.G. Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve 2002, 26, 459–470. [Google Scholar] [CrossRef] [PubMed]
  37. Winkeler, A.; Boisgard, R.; Martin, A.; Tavitian, B. Radioisotopic Imaging of Neuroinflammation: FIGURE 1. J. Nucl. Med. 2009, 51, 1–4. [Google Scholar] [CrossRef] [Green Version]
  38. Zürcher, N.R.; Loggia, M.L.; Lawson, R.; Chonde, D.B.; Izquierdo-Garcia, D.; Yasek, J.E.; Akeju, O.; Catana, C.; Rosen, B.R.; Cudkowicz, M.E.; et al. Increased in vivo glial activation in patients with amyotrophic lateral sclerosis: Assessed with [11C]-PBR28. NeuroImage: Clin. 2015, 7, 409–414. [Google Scholar] [CrossRef] [Green Version]
  39. Graves, M.C.; Fiala, M.; Dinglasan, L.A.V.; Liu, N.Q.; Sayre, J.; Chiappelli, F.; van Kooten, C.; Vinters, H.V. Inflammation in amyotrophic lateral sclerosis spinal cord and brain is mediated by activated macrophages, mast cells and T cells. Amyotroph. Lateral Scler. 2004, 5, 213–219. [Google Scholar] [CrossRef]
  40. Kawamata, T.; Akiyama, H.; Yamada, T.; McGeer, P.L. Immunologic reactions in amyotrophic lateral sclerosis brain and spinal cord tissue. Am. J. Pathol. 1992, 140, 691–707. [Google Scholar]
  41. McCombe, P.A.; Henderson, R.D. The Role of Immune and Inflammatory Mechanisms in ALS. Curr. Mol. Med. 2011, 11, 246–254. [Google Scholar] [CrossRef] [PubMed]
  42. Hu, Y.; Cao, C.; Qin, X.-Y.; Yu, Y.; Yuan, J.; Zhao, Y.; Cheng, Y. Increased peripheral blood inflammatory cytokine levels in amyotrophic lateral sclerosis: A meta-analysis study. Sci. Rep. 2017, 7, 9094. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, W.; Beers, D.R.; Hooten, K.G.; Sieglaff, D.H.; Zhang, A.; Kalyana-Sundaram, S.; Traini, C.M.; Halsey, W.S.; Hughes, A.M.; Sathe, G.M.; et al. Characterization of Gene Expression Phenotype in Amyotrophic Lateral Sclerosis Monocytes. JAMA Neurol. 2017, 74, 677–685. [Google Scholar] [CrossRef] [PubMed]
  44. Bai, Y.; Su, X.; Piao, L.; Jin, Z.; Jin, R. Involvement of Astrocytes and microRNA Dysregulation in Neurodegenerative Diseases: From Pathogenesis to Therapeutic Potential. Front. Mol. Neurosci. 2021, 14, 556215. [Google Scholar] [CrossRef]
  45. Iyer, A.; Zurolo, E.; Prabowo, A.; Fluiter, K.; Spliet, W.G.M.; Van Rijen, P.C.; Gorter, J.A.; Aronica, E. MicroRNA-146a: A Key Regulator of Astrocyte-Mediated Inflammatory Response. PLoS ONE 2012, 7, e44789. [Google Scholar] [CrossRef] [Green Version]
  46. Gomes, C.; Cunha, C.; Nascimento, F.; Ribeiro, J.A.; Vaz, A.R.; Brites, D. Cortical Neurotoxic Astrocytes with Early ALS Pathology and miR-146a Deficit Replicate Gliosis Markers of Symptomatic SOD1G93A Mouse Model. Mol. Neurobiol. 2018, 56, 2137–2158. [Google Scholar] [CrossRef]
  47. Banack, S.A.; Dunlop, R.A.; Cox, P.A. An miRNA fingerprint using neural-enriched extracellular vesicles from blood plasma: Towards a biomarker for amyotrophic lateral sclerosis/motor neuron disease. Open Biol. 2020, 10, 200116. [Google Scholar] [CrossRef]
  48. Staats, K.A.; Borchelt, D.R.; Tansey, M.G.; Wymer, J. Blood-based biomarkers of inflammation in amyotrophic lateral sclerosis. Mol. Neurodegener. 2022, 17, 11. [Google Scholar] [CrossRef]
  49. Vrabec, K.; Koritnik, B.; Leonardis, L.; Dolenc-Grošelj, L.; Zidar, J.; Smith, B.; Vance, C.; Shaw, C.; Rogelj, B.; Glavač, D.; et al. Genetic analysis of amyotrophic lateral sclerosis in the Slovenian population. Neurobiol. Aging 2014, 36, 1601.e17–1601.e20. [Google Scholar] [CrossRef]
  50. Redenšek, S.; Flisar, D.; Kojović, M.; Kramberger, M.G.; Georgiev, D.; Pirtošek, Z.; Trošt, M.; Dolžan, V. Genetic variability of inflammation and oxidative stress genes does not play a major role in the occurrence of adverse events of dopaminergic treatment in Parkinson’s disease. J. Neuroinflamm. 2019, 16, 50. [Google Scholar] [CrossRef] [Green Version]
  51. Lyon, M.S.; Wosiski-Kuhn, M.; Gillespie, R.; Caress, J.; Milligan, C. Inflammation, Immunity, and amyotrophic lateral sclerosis: I. Etiology and pathology. Muscle Nerve 2018, 59, 10–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Tomkins, J.; Banner, S.J.; McDermott, C.; Shaw, P. Mutation screening of manganese superoxide dismutase in amyotrophic lateral sclerosis. NeuroReport 2001, 12, 2319–2322. [Google Scholar] [CrossRef] [PubMed]
  53. Valdmanis, P.N.; Kabashi, E.; Dyck, A.; Hince, P.; Lee, J.; Dion, P.; D’Amour, M.; Souchon, F.; Bouchard, J.P.; Salachas, F.; et al. Association of paraoxonase gene cluster polymorphisms with ALS in France, Quebec, and Sweden. Neurology 2008, 71, 514–520. [Google Scholar] [CrossRef] [PubMed]
  54. Barros, J.B.D.S.; Santos, K.D.F.; Azevedo, R.M.; de Oliveira, R.P.D.; Leobas, A.C.D.; Bento, D.d.C.P.; Santos, R.D.S.; Reis, A.A.D.S. No association of GSTP1 rs1695 polymorphism with amyotrophic lateral sclerosis: A case-control study in the Brazilian population. PLoS ONE 2021, 16, e0247024. [Google Scholar] [CrossRef] [PubMed]
  55. López-López, A.; Gamez, J.; Syriani, E.; Morales, M.; Salvado, M.; Rodriguez, M.J.; Mahy, N.; Vidal-Taboada, J.M. CX3CR1 Is a Modifying Gene of Survival and Progression in Amyotrophic Lateral Sclerosis. PLoS ONE 2014, 9, e96528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Calvo, A.; Moglia, C.; Canosa, A.; Cammarosano, S.; Ilardi, A.; Bertuzzo, D.; Traynor, B.J.; Brunetti, M.; Barberis, M.; Mora, G.; et al. Common polymorphisms of chemokine (C-X3-C motif) receptor 1 gene modify amyotrophic lateral sclerosis outcome: A population-based study. Muscle Nerve 2017, 57, 212–216. [Google Scholar] [CrossRef]
  57. Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic Pruning by Microglia Is Necessary for Normal Brain Development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [Green Version]
  58. Walczak, M.; Lykowska-Szuber, L.; Plucinska, M.; Stawczyk-Eder, K.; Zakerska-Banaszak, O.; Eder, P.; Krela-Kazmierczak, I.; Michalak, M.; Zywicki, M.; Karlowski, W.M.; et al. Is Polymorphism in the Apoptosis and Inflammatory Pathway Genes Associated With a Primary Response to Anti-TNF Therapy in Crohn’s Disease Patients? Front. Pharmacol. 2020, 11, 1207. [Google Scholar] [CrossRef]
  59. Esih, K.; Goričar, K.; Rener-Primec, Z.; Dolžan, V.; Soltirovska-Šalamon, A. CARD8 and IL1B Polymorphisms Influence MRI Brain Patterns in Newborns with Hypoxic-Ischemic Encephalopathy Treated with Hypothermia. Antioxidants 2021, 10, 96. [Google Scholar] [CrossRef]
  60. Lee, K.A.; Gay, C.L.; Lerdal, A.; Pullinger, C.R.; Aouizerat, B.E. Cytokine polymorphisms are associated with fatigue in adults living with HIV/AIDS. Brain Behav. Immun. 2014, 40, 95–103. [Google Scholar] [CrossRef] [Green Version]
  61. Johann, S.; Heitzer, M.; Kanagaratnam, M.; Goswami, A.; Rizo, T.; Weis, J.; Troost, D.; Beyer, C. NLRP3 inflammasome is expressed by astrocytes in the SOD1 mouse model of ALS and in human sporadic ALS patients. Glia 2015, 63, 2260–2273. [Google Scholar] [CrossRef] [PubMed]
  62. Meissner, F.; Molawi, K.; Zychlinsky, A. Mutant superoxide dismutase 1-induced IL-1β accelerates ALS pathogenesis. Proc. Natl. Acad. Sci. USA 2010, 107, 13046–13050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Li, M.; Ona, V.O.; Guégan, C.; Chen, M.; Jackson-Lewis, V.; Andrews, L.J.; Olszewski, A.J.; Stieg, P.E.; Lee, J.-P.; Przedborski, S.; et al. Functional Role of Caspase-1 and Caspase-3 in an ALS Transgenic Mouse Model. Science 2000, 288, 335–339. [Google Scholar] [CrossRef] [PubMed]
  64. Bellezza, I.; Grottelli, S.; Costanzi, E.; Scarpelli, P.; Pigna, E.; Morozzi, G.; Mezzasoma, L.; Peirce, M.J.; Moresi, V.; Adamo, S.; et al. Peroxynitrite Activates the NLRP3 Inflammasome Cascade in SOD1(G93A) Mouse Model of Amyotrophic Lateral Sclerosis. Mol. Neurobiol. 2017, 55, 2350–2361. [Google Scholar] [CrossRef] [Green Version]
  65. Olesen, M.N.; Wuolikainen, A.; Nilsson, A.C.; Wirenfeldt, M.; Forsberg, K.; Madsen, J.S.; Lillevang, S.T.; Brandslund, I.; Andersen, P.M.; Asgari, N. Inflammatory profiles relate to survival in subtypes of amyotrophic lateral sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7, e697. [Google Scholar] [CrossRef] [Green Version]
  66. Polverino, A.; Rucco, R.; Stillitano, I.; Bonavita, S.; Grimaldi, M.; Minino, R.; Pesoli, M.; Trojsi, F.; D’Ursi, A.M.; Sorrentino, G.; et al. In Amyotrophic Lateral Sclerosis Blood Cytokines Are Altered, but Do Not Correlate with Changes in Brain Topology. Brain Connect. 2020, 10, 411–421. [Google Scholar] [CrossRef]
  67. Italiani, P.; Carlesi, C.; Giungato, P.; Puxeddu, I.; Borroni, B.; Bossù, P.; Migliorini, P.; Siciliano, G.; Boraschi, D. Evaluating the levels of interleukin-1 family cytokines in sporadic amyotrophic lateral sclerosis. J. Neuroinflamm. 2014, 11, 94. [Google Scholar] [CrossRef] [Green Version]
  68. Bresciani, G.; Cruz, I.B.M.; De Paz, J.A.; Cuevas, M.J.; González-Gallego, J. The MnSOD Ala16Val SNP: Relevance to human diseases and interaction with environmental factors. Free Radic. Res. 2013, 47, 781–792. [Google Scholar] [CrossRef]
  69. Sutton, A.; Imbert, A.; Igoudjil, A.; Descatoire, V.; Cazanave, S.; Pessayre, D.; Degoul, F. The manganese superoxide dismutase Ala16Val dimorphism modulates both mitochondrial import and mRNA stability. Pharm. Genom. 2005, 15, 311–319. [Google Scholar] [CrossRef]
  70. Ekoue, D.N.; He, C.; Diamond, A.M.; Bonini, M.G. Manganese superoxide dismutase and glutathione peroxidase-1 contribute to the rise and fall of mitochondrial reactive oxygen species which drive oncogenesis. Biochim. Biophys. Acta 2017, 1858, 628–632. [Google Scholar] [CrossRef]
  71. Bresciani, G.; González-Gallego, J.; da Cruz, I.B.; de Paz, J.A.; Cuevas, M.J. The Ala16Val MnSOD gene polymorphism modulates oxidative response to exercise. Clin. Biochem. 2012, 46, 335–340. [Google Scholar] [CrossRef] [PubMed]
  72. Verde, F.; Tiloca, C.; Morelli, C.; Doretti, A.; Poletti, B.; Maderna, L.; Messina, S.; Gentilini, D.; Fogh, I.; Ratti, A.; et al. PON1 is a disease modifier gene in amyotrophic lateral sclerosis: Association of the Q192R polymorphism with bulbar onset and reduced survival. Neurol. Sci. 2019, 40, 1469–1473. [Google Scholar] [CrossRef] [PubMed]
  73. Forsberg, L.; Lyrenäs, L.; Morgenstern, R.; de Faire, U. A common functional C-T substitution polymorphism in the promoter region of the human catalase gene influences transcription factor binding, reporter gene transcription and is correlated to blood catalase levels. Free Radic. Biol. Med. 2001, 30, 500–505. [Google Scholar] [CrossRef]
  74. Esih, K.; Goričar, K.; Dolžan, V.; Rener-Primec, Z. The association between antioxidant enzyme polymorphisms and cerebral palsy after perinatal hypoxic-ischaemic encephalopathy. Eur. J. Paediatr. Neurol. 2016, 20, 704–708. [Google Scholar] [CrossRef]
  75. Goulas, A.; Fidani, L.; Kotsis, A.; Mirtsou, V.; Petersen, R.C.; Tangalos, E.; Hardy, J. An association study of a functional catalase gene polymorphism, −262C→T, and patients with Alzheimer’s disease. Neurosci. Lett. 2002, 330, 210–212. [Google Scholar] [CrossRef]
  76. Liu, K.; Liu, X.; Wang, M.; Wang, X.; Kang, H.; Lin, S.; Yang, P.; Dai, C.; Xu, P.; Li, S.; et al. Two common functional catalase gene polymorphisms (rs1001179 and rs794316) and cancer susceptibility: Evidence from 14,942 cancer cases and 43,285 controls. Oncotarget 2016, 7, 62954–62965. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, C.-D.; Sun, Y.; Chen, N.; Huang, L.; Huang, J.-W.; Zhu, M.; Wang, T.; Ji, Y.-L. The Role of Catalase C262T Gene Polymorphism in the Susceptibility and Survival of Cancers. Sci. Rep. 2016, 6, 26973. [Google Scholar] [CrossRef] [Green Version]
  78. Nikolić-Kokić, A.; Stević, Z.; Blagojević, D.; Davidović-Plavšić, B.; Jones, D.R.; Spasić, M. Alterations in anti-oxidative defence enzymes in erythrocytes from sporadic amyotrophic lateral sclerosis (SALS) and familial ALS patients. Clin. Chem. Lab. Med. (CCLM) 2006, 44, 589–593. [Google Scholar] [CrossRef]
  79. Golenia, A.; Leśkiewicz, M.; Regulska, M.; Budziszewska, B.; Szczęsny, E.; Jagiełła, J.; Wnuk, M.; Ostrowska, M.; Lasoń, W.; Basta-Kaim, A.; et al. Catalase activity in blood fractions of patients with sporadic ALS. Pharmacol. Rep. 2014, 66, 704–707. [Google Scholar] [CrossRef]
  80. Babu, G.N.; Kumar, A.; Chandra, R.; Puri, S.; Singh, R.; Kalita, J.; Misra, U. Oxidant–antioxidant imbalance in the erythrocytes of sporadic amyotrophic lateral sclerosis patients correlates with the progression of disease. Neurochem. Int. 2008, 52, 1284–1289. [Google Scholar] [CrossRef]
  81. Zou, Z.-Y.; Zhou, Z.-R.; Che, C.-H.; Liu, C.-Y.; He, R.-L.; Huang, H.-P. Genetic epidemiology of amyotrophic lateral sclerosis: A systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2017, 88, 540–549. [Google Scholar] [CrossRef] [PubMed]
  82. Verma, A. Clinical Manifestation and Management of Amyotrophic Lateral Sclerosis. In Amyotrophic Lateral Sclerosis; Araki, T., Ed.; Exon Publications: Brisbane, Australia, 2021. [Google Scholar] [CrossRef]
Figure 1. Association of IL1B rs1071676 with ALS-FRS-R points per month progression rate. In the boxplots, empty circles represent outliers between 1.5 and 3-times interquartile range away from the 1st or 3rd quartiles, while outliers beyond 3-times interquartile range are presented with stars (*).
Figure 1. Association of IL1B rs1071676 with ALS-FRS-R points per month progression rate. In the boxplots, empty circles represent outliers between 1.5 and 3-times interquartile range away from the 1st or 3rd quartiles, while outliers beyond 3-times interquartile range are presented with stars (*).
Genes 13 00757 g001
Figure 2. Association of CAT rs1001179 (A) and IL1B rs1071676 (B) with survival of ALS patients. Carriers of at least one polymorphic CAT rs1001179 T allele had longer survival after adjustment for age at onset and rate of disease progression (HR = 0.68, 95% CI = 0.47–0.99, p = 0.046). Carriers of two polymorphic IL1B rs1071676 C alleles had shorter survival after adjustment for age at onset and rate of disease progression (HR = 5.02, 95% CI = 1.92–13.16, p = 0.001).
Figure 2. Association of CAT rs1001179 (A) and IL1B rs1071676 (B) with survival of ALS patients. Carriers of at least one polymorphic CAT rs1001179 T allele had longer survival after adjustment for age at onset and rate of disease progression (HR = 0.68, 95% CI = 0.47–0.99, p = 0.046). Carriers of two polymorphic IL1B rs1071676 C alleles had shorter survival after adjustment for age at onset and rate of disease progression (HR = 5.02, 95% CI = 1.92–13.16, p = 0.001).
Genes 13 00757 g002
Table 1. Clinical characteristics of ALS patients for the whole cohort (N = 185) and for patients with available ALS-FRS-R data (N = 164).
Table 1. Clinical characteristics of ALS patients for the whole cohort (N = 185) and for patients with available ALS-FRS-R data (N = 164).
CharacteristicCategory/UnitWhole CohortPatients with ALS-FRS-R Data
GenderMale, N (%)94 (50.8)84 (51.2)
Female, N (%)91 (49.2)81 (48.8)
Age at onsetYears, Median (25–75%)63 (56–71) [2]63 (57–71)
Age at the time of blood collectionYears, Median (25–75%)65 (58–72) [2]65 (59–73) [2]
ALS onsetSpinal133 (72.3) [1]121 (73.8)
Bulbar45 (24.5)40 (24.4)
Other6 (3.3)3 (1.8)
C9orf72 mutationNo, N (%)179 (96.8)159 (97.0)
Yes, N (%)6 (3.2)5 (3.0)
DeathNo, N (%)25 (14.0) [7]24 (14.9) [3]
Yes, N (%)153 (86.0)137 (85.1)
SurvivalMonths, Median (25–75%)39.7 (26.6–68.4) [7]42.3 (28.0–72.3) [3]
Follow-up timeMonths, Median (25–75%)104.9 (90.7–156.6) [7]124.2 (89.7–156.6) [3]
Level of functional impairment aPoints, Median (25–75%)34.94 (29.50–39.99) [21]34.94 (29.50–39.99)
Rate of disease progression bMedian (25–75%)−0.80 (−1.40 to −0.30) [37]−0.80 (−1.40 to −0.30) [16]
a ALS-FRS-R, b Slope of the linear regression line for ALS-FRS-R points per month. Number of missing data is presented in [] brackets. ALS, amyotrophic lateral sclerosis; ALS-FRS-R, ALS functional rating scale revised.
Table 2. Association between the investigated polymorphisms and ALS risk.
Table 2. Association between the investigated polymorphisms and ALS risk.
GeneSNPGenotypeOR (95% CI)POR (95% CI)adjPadj
SOD2rs4880CCReference Reference
CT1.07 (0.69–1.66)0.7531.01 (0.58–1.74)0.978
TT1.06 (0.64–1.77)0.8100.84 (0.44–1.58)0.586
CT + TT1.07 (0.71–1.62)0.7480.95 (0.57–1.58)0.834
CATrs1001179CCReference Reference
CT0.92 (0.63–1.35)0.6790.94 (0.57–1.53)0.792
TT0.84 (0.38–1.87)0.6761.03 (0.38–2.74)0.958
CT + TT0.91 (0.63–1.32)0.6180.95 (0.60–1.51)0.826
GPX1rs1050450CCReference Reference
CT0.95 (0.64–1.40)0.7881.05 (0.65–1.70)0.849
TT0.97 (0.54–1.76)0.9211.11 (0.51–2.40)0.788
CT + TT0.95 (0.66–1.37)0.7951.06 (0.67–1.67)0.799
IL1Brs1143623GGReference Reference
GC0.80 (0.55–1.19)0.2720.74 (0.46–1.21)0.235
CC1.07 (0.57–2.01)0.8320.93 (0.42–2.04)0.850
GC + CC0.85 (0.59–1.23)0.3900.78 (0.49–1.23)0.282
rs16944TTReference Reference
TC0.99 (0.56–1.76)0.9790.99 (0.48–2.04)0.984
CC1.05 (0.59–1.85)0.8731.06 (0.52–2.17)0.872
TC + CC1.02 (0.60–1.74)0.9421.03 (0.52–2.01)0.939
rs1071676GGReference Reference
GC0.93 (0.63–1.37)0.7141.11 (0.68–1.80)0.685
CC0.47 (0.19–1.21)0.1180.54 (0.18–1.61)0.269
GC + CC0.86 (0.59–1.25)0.4241.01 (0.63–1.60)0.981
MIR146Ars2910164GGReference Reference
GC1.05 (0.72–1.55)0.7930.85 (0.52–1.39)0.518
CC0.74 (0.32–1.75)0.4980.54 (0.18–1.62)0.273
GC + CC1.01 (0.70–1.46)0.9700.81 (0.50–1.29)0.364
IL6rs1800795GGReference Reference
GC1.18 (0.78–1.77)0.4310.83 (0.49–1.39)0.467
CC0.97 (0.58–1.63)0.9141.01 (0.53–1.91)0.985
GC + CC1.11 (0.76–1.63)0.5840.88 (0.54–1.42)0.593
TNFrs1800629GGReference Reference
GA0.88 (0.59–1.32)0.5420.97 (0.58–1.60)0.897
AA0.94 (0.31–2.87)0.9160.77 (0.20–2.99)0.707
GA + AA0.89 (0.60–1.31)0.5480.95 (0.58–1.54)0.826
Adj: adjusted for age and gender; CI, confidence interval; OR, odds ratio; SNP, single nucleotide polymorphism.
Table 3. Comparison of genotype frequencies among patients with different types of ALS onset.
Table 3. Comparison of genotype frequencies among patients with different types of ALS onset.
GeneSNPGenotypeSpinal
N (%)
Bulbar
N (%)
Other
N (%)
p
SOD2rs4880CC37 (80.4)6 (13)3 (6.5)0.153
CT62 (68.1)27 (29.7)2 (2.2)
TT33 (71.7)12 (26.1)1 (2.2)
CT + TT95 (69.3)39 (28.5)3 (2.2)Pdom = 0.036
CATrs1001179CC81 (74.3)23 (21.1)5 (4.6)0.585
CT44 (68.8)19 (29.7)1 (1.6)
TT7 (70)3 (30)0 (0)
CT + TT51 (68.9)22 (29.7)1 (1.4)Pdom = 0.244
GPX1rs1050450CC71 (76.3)19 (20.4)3 (3.2)0.492
CT46 (64.8)22 (31)3 (4.2)
TT16 (80)4 (20)0 (0)
CT + TT62 (68.1)26 (28.6)3 (3.3)Pdom = 0.446
IL1Brs1143623GG76 (76)23 (23)1 (1)0.249
GC44 (68.8)16 (25)4 (6.3)
CC12 (63.2)6 (31.6)1 (5.3)
GC + CC56 (67.5)22 (26.5)5 (6)Pdom = 0.128
rs16944TT15 (62.5)6 (25)3 (12.5)0.063
TC55 (71.4)19 (24.7)3 (3.9)
CC62 (75.6)20 (24.4)0 (0)
TC + CC117 (73.6)39 (24.5)3 (1.9)Pdom = 0.051
rs1071676GG87 (77)21 (18.6)5 (4.4)0.132
GC41 (64.1)22 (34.4)1 (1.6)
CC4 (66.7)2 (33.3)0 (0)
GC + CC45 (64.3)24 (34.3)1 (1.4)Pdom = 0.039
MIR146Ars2910164GG80 (72.1)28 (25.2)3 (2.7)0.665
GC48 (73.8)14 (21.5)3 (4.6)
CC4 (57.1)3 (42.9)0 (0)
GC + CC52 (72.2)17 (23.6)3 (4.2)Pdom = 0.872
IL6rs1800795GG43 (69.4)17 (27.4)2 (3.2)0.655
GC61 (70.1)23 (26.4)3 (3.4)
CC28 (82.4)5 (14.7)1 (2.9)
GC + CC89 (73.6)28 (23.1)4 (3.3)Pdom = 0.861
TNFrs1800629GG91 (71.7)32 (25.2)4 (3.1)1.000
GA37 (72.5)12 (23.5)2 (3.9)
AA4 (80)1 (20)0 (0)
GA + AA41 (73.2)13 (23.2)2 (3.6)Pdom = 0.951
SNP, single nucleotide polymorphism; Pdom: p-value for dominant genetic model. The bold represents statistical significance.
Table 4. Association of investigated SNPs with level of functional impairment and rate of disease progression.
Table 4. Association of investigated SNPs with level of functional impairment and rate of disease progression.
GeneSNPGenotypeLevel of Functional Impairment
Median (25–75%)
pRate of Disease Progression
Median (25–75%)
p
SOD2rs4880CC36.94 (31.07–40.86)0.199−0.66 (−1.25 to −0.27)0.455
CT34 (29–39.6) −0.77 (−1.47 to −0.28)
TT34.18 (28.55–39.53) −0.97 (−1.70 to −0.40)
CT + TT34 (29–39.6)Pdom = 0.073−0.83 (−1.50 to −0.31)Pdom = 0.263
CATrs1001179CC35 (29.21–39.98)0.111−0.81 (−1.42 to −0.34)0.584
CT33.89 (29.06–39.23) −0.71 (−1.54 to −0.28)
TT39.29 (34.7–43.84) −0.42 (−1.56 to −0.11)
CT + TT34.67 (30.1–40.31)Pdom = 0.690−0.67 (−1.49 to −0.27)Pdom = 0.847
GPX1rs1050450CC34.75 (30–40.71)0.606−0.66 (−1.31 to −0.33)0.323
CT35 (28.62–39.17) −0.83 (−1.50 to −0.25)
TT34.51 (29.75–40.51) −1.12 (−2.27 to −0.47)
CT + TT35 (29.12–39.48)Pdom = 0.470−0.92 (−1.57 to −0.26)Pdom = 0.421
IL1Brs1143623GG34.02 (29.5–40)0.887−0.72 (−1.34 to −0.25)0.498
GC35.14 (29–39.83) −0.76 (−1.75 to −0.32)
CC34.93 (29.66–40.82) −0.93 (−1.54 to −0.49)
GC + CC35.14 (29.38–39.83)Pdom = 0.843−0.80 (−1.56 to −0.33)Pdom = 0.307
rs16944TT33.13 (29.57–40.3)0.903−1.04 (−1.50 to −0.53)0.383
TC34.88 (28.74–39.83) −0.66 (−1.35 to −0.28)
CC35 (30–40) −0.82 (−1.38 to −0.21)
TC + CC35 (29–40)Pdom = 0.893−0.71 (−1.36 to −0.27)Pdom = 0.174
rs1071676GG34.39 (29.86–40.06)0.565−0.71 (−1.24 to −0.34)0.015
GC34.73 (28.99–39.99) −0.80 (−1.48 to −0.18)
CC36.96 (34.52–40.12) −2.40 (−3.10 to −1.69)
GC + CC35 (29–39.98)Pdom = 0.875−0.81 (−1.77 to −0.24)Pdom = 0.690
MIR146Ars2910164GG34.73 (29.86–39.78)0.548−0.68 (−1.54 to −0.27)0.695
GC34.38 (28.88–40.01) −0.83 (−1.45 to −0.41)
CC37.8 (31.05–41) −0.83 (−1.17 to −0.19)
GC + CC35 (29.12–40.09)Pdom = 0.851−0.83 (−1.39 to −0.39)Pdom = 0.479
IL6rs1800795GG34 (30–39.6)0.285−0.53 (−1.33 to −0.18)0.216
GC34 (28–39.83) −0.83 (−1.49 to −0.37)
CC36.95 (32.08–40.4) −0.83 (−1.64 to −0.28)
GC + CC34.94 (29–40.2)Pdom = 0.634−0.83 (−1.52 to −0.34)Pdom = 0.108
TNFrs1800629GG35.04 (30.53–40.24)0.512−0.82 (−1.39 to −0.31)0.827
GA34 (27.95–37.98) −0.67 (−2.07 to −0.27)
AA38.15 (27.46–39.98) −0.80 (−0.81 to −0.36)
GA + AA34 (27.91–38.54)Pdom = 0.272−0.68 (−1.49 to −0.27)Pdom = 0.962
SNP, single nucleotide polymorphism; Pdom: P-value for dominant genetic model. The bold represents statistical significance.
Table 5. Association of investigated SNPs with survival of ALS patients.
Table 5. Association of investigated SNPs with survival of ALS patients.
GeneSNPGenotypeMedian Survival (25–75%)HR (95% CI)pHR (95% CI)adjPadj
SOD2rs4880CC37.6 (25.7–76.0)Reference Reference
CT39.7 (27.7–69.2)1.00 (0.67–1.50)0.9851.16 (0.71–1.9)0.558
TT43.5 (23.7–63.0)1.20 (0.76–1.87)0.4351.18 (0.69–2.03)0.549
CT + TT40.7 (26.9–66.9)1.06 (0.73–1.55)0.7421.17 (0.73–1.86)0.520
CATrs1001179CC37.0 (26.6–65.8)Reference Reference
CT43.7 (26.5–72.3)0.83 (0.58–1.17)0.2830.70 (0.46–1.04)0.079
TT60.3 (56.7–85.6)0.61 (0.30–1.26)0.1790.62 (0.28–1.36)0.231
CT + TT47.6 (26.5–78.6)0.79 (0.57–1.1)0.1560.68 (0.47–0.99)0.046
GPX1rs1050450CC39.0 (28.2–69.2)Reference Reference
CT39.7 (25.3–72.8)0.89 (0.63–1.26)0.5070.85 (0.58–1.24)0.396
TT40.5 (29.0–65.8)0.99 (0.58–1.68)0.9691.06 (0.53–2.14)0.869
CT + TT39.7 (25.3–67.6)0.91 (0.66–1.26)0.5700.88 (0.61–1.26)0.477
IL1Brs1143623GG41.8 (26.5–74.5)Reference Reference
GC39.0 (25.3–68.4)1.12 (0.8–1.58)0.5031.21 (0.82–1.78)0.332
CC37.0 (34.0–56.7)1.32 (0.76–2.31)0.3220.78 (0.38–1.59)0.491
GC + CC39.0 (27.7–67.6)1.16 (0.84–1.6)0.3611.11 (0.77–1.6)0.572
rs16944TT37.0 (33.2–56.7)Reference Reference
TC43.7 (25.7–76.0)0.66 (0.4–1.11)0.1181.08 (0.55–2.09)0.827
CC39.4 (26.5–66.9)0.74 (0.45–1.23)0.2451.12 (0.59–2.13)0.722
TC + CC41.8 (26.5–72.3)0.70 (0.44–1.14)0.1501.1 (0.59–2.06)0.754
rs1071676GG39.7 (25.7–66.9)Reference Reference
GC41.8 (28.7–83.6)0.75 (0.53–1.07)0.1101.03 (0.69–1.53)0.900
CC24.3 (18.4–32.0)1.53 (0.62–3.77)0.3585.02 (1.92–13.16)0.001
GC + CC40.5 (28.0–78.6)0.79 (0.57–1.11)0.1711.13 (0.77–1.66)0.529
MIR146Ars2910164GG42.9 (25.7–72.3)Reference Reference
GC39.0 (27.7–65.8)1.03 (0.73–1.45)0.8601 (0.68–1.46)0.989
CC36.7 (31.4–60.3)1.26 (0.55–2.88)0.5841.42 (0.57–3.54)0.448
GC + CC39.0 (27.7–65.8)1.05 (0.76–1.46)0.7681.03 (0.71–1.49)0.876
IL6rs1800795GG37.0 (26.6–78.6)Reference Reference
GC39.7 (25.7–60.3)1.02 (0.84–1.73)0.3171.06 (0.69–1.62)0.793
CC40.7 (29.9–74.5)0.90 (0.56–1.43)0.6470.84 (0.48–1.46)0.527
GC + CC40.5 (26.5–66.9)1.10 (0.78–1.55)0.5851 (0.66–1.5)0.984
TNFrs1800629GG39.0 (25.7–67.6)Reference Reference
GA41.8 (29.8–74.5)0.92 (0.64–1.33)0.6700.78 (0.5–1.21)0.266
AA37.0 (32.5–57.6)1.25 (0.51–3.06)0.6311.45 (0.58–3.58)0.426
GA + AA40.7 (29.8–74.5)0.95 (0.67–1.35)0.7780.84 (0.56–1.27)0.411
Adj: adjusted for age at onset and rate of disease progression. The bold represents statistical significance.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ravnik-Glavač, M.; Goričar, K.; Vogrinc, D.; Koritnik, B.; Lavrenčič, J.G.; Glavač, D.; Dolžan, V. Genetic Variability of Inflammation and Oxidative Stress Genes Affects Onset, Progression of the Disease and Survival of Patients with Amyotrophic Lateral Sclerosis. Genes 2022, 13, 757. https://doi.org/10.3390/genes13050757

AMA Style

Ravnik-Glavač M, Goričar K, Vogrinc D, Koritnik B, Lavrenčič JG, Glavač D, Dolžan V. Genetic Variability of Inflammation and Oxidative Stress Genes Affects Onset, Progression of the Disease and Survival of Patients with Amyotrophic Lateral Sclerosis. Genes. 2022; 13(5):757. https://doi.org/10.3390/genes13050757

Chicago/Turabian Style

Ravnik-Glavač, Metka, Katja Goričar, David Vogrinc, Blaž Koritnik, Jakob Gašper Lavrenčič, Damjan Glavač, and Vita Dolžan. 2022. "Genetic Variability of Inflammation and Oxidative Stress Genes Affects Onset, Progression of the Disease and Survival of Patients with Amyotrophic Lateral Sclerosis" Genes 13, no. 5: 757. https://doi.org/10.3390/genes13050757

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