Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: Free Radic Biol Med. 2013 Jun 21;65:10.1016/j.freeradbiomed.2013.06.029. doi: 10.1016/j.freeradbiomed.2013.06.029

Clinical Perspective of Oxidative Stress in Sporadic ALS

Emanuele D’Amico 1, Pam Factor-Litvak 2, Regina M Santella 3, Hiroshi Mitsumoto 4
PMCID: PMC3859834  NIHMSID: NIHMS498379  PMID: 23797033

Abstract

Sporadic amyotrophic lateral sclerosis (sALS) is one of the most devastating neurological diseases; most patients die within 3 to 4 years after symptom onset. Oxidative stress is a disturbance in the pro-oxidative/anti-oxidative balance favoring the pro-oxidative state. Autopsy and laboratory studies in ALS indicate that oxidative stress plays a major role in motor neuron degeneration and astrocyte dysfunction. Oxidative stress biomarkers in cerebrospinal fluid, plasma, and urine, are elevated, suggesting that abnormal oxidative stress is generated outside of the central nervous system. Our review indicates that agricultural chemicals, heavy metals, military service, professional sports, excessive physical exertion, chronic head trauma, and certain foods might be modestly associated with ALS risk, with a stronger association between risk and smoking. At the cellular level, these factors are all involved in generating oxidative stress. Experimental studies indicate that a combination of insults that induce modest oxidative stress can exert additive deleterious effects on motor neurons, suggesting multiple exposures in real-world environments are important. As the disease progresses, nutritional deficiency, cachexia, psychological stress, and impending respiratory failure may further increase oxidative stress. Moreover, accumulating evidence suggests that ALS is possibly a systemic disease. Laboratory, pathologic, and epidemiologic evidence clearly support the hypothesis that oxidative stress is central in the pathogenic process, particularly in genetically susceptive individuals. If we are to improve ALS treatment, well-designed biochemical and genetic epidemiological studies, combined with a multidisciplinary research approach, are needed and will provide knowledge crucial to our understanding of ALS etiology, pathophysiology, and prognosis.

Keywords: sALS, oxidative stress, environmental epidemiology, phenotypic variation, disease prognosis

INTRODUCTION

Amyotrophic lateral sclerosis (ALS) is one of the most devastating neurological diseases. Patients with ALS develop relentlessly progressive paralysis that involves all the skeletal muscles, as well as the bulbar and respiratory muscles. This paralysis ultimately leads to the patient’s death on average 40 months after symptom onset. Riluzole is the only medication that has received Food and Drug Administration approval, but it has only modest benefits at best (13). More than a dozen molecular mutations have been discovered in familial ALS (fALS), which constitutes 5% to 10% of all ALS cases (49). In contrast, despite decades of intense research and a number of highly plausible hypotheses (1012), still little is known regarding factors related to the causes or risks of developing sporadic ALS (sALS), which we focus on in this review.

Our review is specifically focused on clinical, or patient-oriented, research in oxidative stress in sALS. Since discussing oxidative stress on the basic science level is not our objective, we provide a few excellent reviews here for this information (1315). First, we briefly discuss the effects of oxidative stress on motor neurons and the central nervous system (CNS). Moreover, we review currently available biomarkers that are useful for investigating oxidative stress in sALS and the potential consequences of oxidized products. Then, we examine epidemiological studies and how environmental and lifestyle factors potentially trigger oxidative stress in exposed individuals. We also discuss evidence that oxidative stress is not just an event in the CNS but rather a systemic process, although the CNS and motor neurons may be most vulnerable to systemic oxidative stress. Because oxidative stress results from pro- and anti-oxidative imbalance (15, 16), current knowledge of intrinsic anti-oxidative mechanisms is reviewed along with possible interactions between oxidative stress and modifier genes in sALS. We close with a call for molecular epidemiology studies in ALS. We hope that this review will stimulate more research in patients with ALS that investigates the relationship between sALS pathogenesis and environmental exposures related to oxidative stress, which will ultimately lead to novel therapeutic approaches and better clinical management.

MOTOR NEURON DEGENERATION IN ALS

The CNS as a whole is particularly susceptible to oxidative stress because the neuronal membrane contains a high abundance of polyunsaturated fatty acids, especially arachidonic and docosahexaenoic acids; it consumes oxygen at a high rate; and it contains high concentrations of redox-active transition metals but a relatively (compared to the oxidative stress level) low concentration of antioxidants (17, 18). In sALS, at the cellular level, genetic factors, excitotoxicity, apoptosis, inflammation, mitochondrial dysfunction, protein aggregates, and oxidative stress are among the primary hypotheses put forth to explain motor neuron degeneration (1012). Among these factors, oxidative stress appears intimately linked to a series of cellular events in motor neurons that contribute to neuronal degeneration and death (19). In fact, analyses of post-mortem neuronal tissue from patients with sALS consistently show oxidative damage to proteins, lipids, and DNA (2024), suggesting that oxidative injury is very likely to be one of the principal cellular mechanisms of motor neuron degeneration (25). Several excellent reviews on oxidative stress in ALS are available, mostly focusing on the molecular mechanisms of oxidative stress in neuronal degeneration (2531). In recent years, however, laboratory studies have revealed that astrocytic glial cells play a key role in neuronal degeneration (3236). When cultured astrocytes undergo oxidative damage, glutamate transport is impaired, likely resulting in excitotoxic neuronal injury (37). There are a number of in vitro studies, suggesting that experimental oxidative stress in astrocytes and oligodendroglia could lead to motoneuronal degeneration (3841).

OXIDATIVE STRESS BIOMARKERS AVAILABLE FOR STUDIES IN ALS

Whereas oxidative stress appears to be closely associated with motor neuron degeneration in ALS, it still remains unsettled whether oxidative stress is involved outside of the CNS such as in skeletal muscles (28, 42, 43). Reliable oxidative stress biomarkers are the first essential step to ascertaining such extra-CNS involvement (42). Oxidative stress damages critical cellular macromolecules, which can eventually lead to cell death by necrosis or apoptosis (44). The localization and effects of oxidative stress may be determined from analysis of discrete biomarkers of oxidative or nitrosative stress damage in tissues and biological fluids (13, 25, 45). Among the well-studied biomarkers are oxidized DNA, lipids, and proteins. To be truly useful, an oxidative stress biomarker must have some degree of predictive validity, but this relation still must be fully demonstrated for sALS (46), so such markers should be interpreted cautiously in cases of sALS (47). Here, we review the most thoroughly studied and reliable (and thus the most important) oxidative stress biomarkers to date, and how they are used in sALS studies.

DNA is clearly a preferred oxidative stress target. In particular, guanine is easily oxidized (48). 8-Oxo-deoxyguanosine (8-oxoG) is the most abundant type of oxidative DNA damage and is well-studied as an oxidative stress biomarker in vivo (49, 50). CNS lipids are also common targets of reactive oxygen species (ROS, e.g., H2O2, O2-, RO, OH, HOCl) and reactive nitrogen species (RNS, ONOO-, NO2, NO) (51).

Oxidative stress reactions can significantly alter membrane structure and intracellular lipids, resulting in altered fluidity, permeability, transport, and metabolic processes. Fatty acid peroxides can give rise to a variety of aldehydes, such as 4-hydroxy-2-nonenal (HNE), thiobarbituric acid-reactive substances (TBARS) like malondialdehyde (MDA), and isoprostanes (IsoPs) (52). These products are relatively stable and can diffuse within or exit the cell and attack targets far from the site of the original event. Therefore, they are not only end products and remnants of lipid peroxidation processes but also may act as “second cytotoxic messengers” (53, 54). Lipid peroxidation products can also be absorbed from the diet and excreted in urine. It follows that measurements of hydroxyl fatty acids in plasma total lipids as well as plasma or urinary MDA and HNE can be confounded by diet and should not be used as an index of whole-body lipid peroxidation unless diet is strictly controlled (55).

HNE is considered one of the most toxic molecules known to cause cellular damage and apoptosis (56, 57). Moreover, HNE is capable of disintegrating membranes and can diffuse between cell components (58). HNE interacts with the cell membrane and a wide array of amino acid residues present in receptors, chaperones, electron transport chain proteins, and proteins that repair oxidative damage (56). Other lipid biomarkers of oxidative stress include the F2-isoprostane family (F2-IsoPs) (59). F2-IsoPs are formed non-enzymatically as a result of the free radical-mediated peroxidation of arachidonic acid (60). F2-IsoPs have been found in measurable quantities in most of the biological fluids that have been analyzed. Plasma and urine are the sample types that are commonly analyzed, as they are the most convenient to obtain and the least invasive (61). F2-IsoPs are not only biomarkers of oxidative stress but exert biological effects themselves, suggesting they also might function as pathophysiologic mediators of oxidant injury (62). Most current knowledge on the biological actions of F2-IsoPs is limited to 15-F2t–IsoP, which is a potent vasoconstrictor. However, it is not known whether the IsoP concentrations reached locally in vivo is sufficient to exert biological effects. No specific inhibitor of their biological action exists (63); however, molecular modeling of phospholipids containing IsoPs reveals remarkably distorted molecules (64, 65). Thus, the formation of these abnormal phospholipids would be expected to exert profound effects on membrane fluidity and integrity, well-known sequellae of oxidative injury (60). Moreover, exposure to IsoPs is associated with neurotoxicity that modulates neuronal apoptosis (66).

Carbonylation and other ROS-associated post-translational modifications readily inactivate proteins, including enzymes (67, 68). Oxidation of the protein backbone is initiated by hydroxyl radical-mediated extraction of the α-hydrogen atom from the amino acid residues, forming a carbon-centered radical (67). All amino acid residues can be modified, resulting in the oxidation of amino acid side chains and protein–protein cross-linkages. Methionine and cysteine residues are more prone to oxidation than others due to the presence of sulfur. Tryptophan is also readily oxidized. Direct oxidation of lysine, arginine, tyrosine, proline, and threonine residues give rise to carbonyl derivatives, which, because they are easily detected, are most often used as a marker of proteins modified by oxidative stress (69, 70).

Peroxynitrite also affects protein activity by oxidizing amino acid residues as seen in nitrotyrosine, which is found in the CNS in human sALS, fALS and ALS-mouse models (7174). 3-nitrotyrosine (3-NT) is often used as a marker of nitrosative stress in CNS (cerebrospinal fluid, CSF) and peripheral blood samples (74, 75). Peroxynitrite-mediated tyrosine nitration has been suggested as a potential mechanism for the induction of neuronal degeneration in several neurodegenerative disorders including ALS (76, 77); however, the exact mechanisms underlying how free nitrotyrosine can cause neuronal degeneration remain to be elucidated (78).

Investigating the effects of oxidative stress on molecular targets and identifying reliable biomarkers involved in oxidative stress are two great challenges in sALS. A compelling study by Bogdanov et al (42), investigating 65 patients with newly diagnosed ALS (60 with sALS), 63 subjects without neurological disorders, and 37 patients with other neurological disorders, examined levels of 8-oxodG in plasma (n= 57, 21, and 14 patients, respectively), urine (n= 59, 56, and 35 patients, respectively) and CSF (n= 23, 19, and 10, respectively). They found that 8-oxodG was elevated in the CSF and urine of ALS patients compared to both control groups. Furthermore, plasma levels in ALS were significantly higher than in the group with no neurological disorders but not in the group with other neurological disorders. In all subjects, plasma and CSF 8-oxodG concentrations increased with age, confirming that oxidative damage has a role in normal aging. Plasma and urine 8-oxodG concentrations increased significantly with time (9 months from the baseline) within the ALS group (31 subjects had follow up) and the increase in urinary 8-oxodG was significant in the ALS group compared to the neurological disorders-free group (42). Our preliminary studies of oxidative stress in sALS progression have also revealed increased urinary 8-oxodG (43).

In 2004, Simpson et al (79) measured HNE in both serum and CSF in 63 patients with sALS, comparing the findings to 19 patients with other neurodegenerative disease, 14 patients with neurologic but non-neurodegenerative disease, and 16 healthy controls (serum only). They found that serum and CSF HNE levels were significantly elevated in sALS cases compared to both disease and healthy control groups. Further, HNE levels were 2.3-fold greater in CSF than serum in the sALS patients (P = 0.00005). In 42 of the patients with sALS, serum HNE was measured throughout the disease course; HNE concentration increased over time and positively correlated with advancing disease (r = 0.17, P = 0.03).

Chromatographic mass spectrometry has revealed increased CSF F2-IsoP concentration in Alzheimer disease but not in ALS (80). However, F2-IsoPs have been found to be significantly increased in urine as measured using an immunoassay technique in a cross-sectional pilot study of 50 participants with sALS compared to 46 healthy control subjects (P = 0.002) (43). Using logistic regression and controlling for sex and age, a positive association between sALS and the natural log of urinary F2-IsoP concentration (adjusted for creatinine) was found (OR = 3.24; 95%CI = 1.40–7.55; P = 0.006) for a doubling in the level of urinary F2-IsoPs (43).

Table 1 lists additional studies of potential oxidative stress biomarkers in sALS (74, 8190), not been described above.

Table 1.

Oxidative stress (OS) biomarker in ALS (other than described in the text)

Study ALS cases
(n)
Controls Results
Tohgi et al.
1999(81)
19 sALS 19 HC ↑CSF 3-NT and
3-NT/T ratio in sALS
Tohgi et al, 1999
(82)
18 sALS 14 HC ↑NO3 in sALS,
↑GSSG/GSH ratio in
sALS
Bonnefont-
Rousselot et al,
2000 (83)
167 62 HC ↑TBARS in ALS, ↑ SOD1
activity in ALS, no other
significant differences
Ryberg et al,
2004 (84)
14 19 HC, 17 AD No increasing in ALS
Sohmiya et al,
2005 (85)
20 sALS 20 HC ↑ oxidized form of coQ10
in sALS and correlation
with disease duration, no
others significant
differences.
Yoshino et al,
2006a (74)
20 / ↓ CSF 3NT levels at the
end of antioxidant
therapy
Siciliano et al,
2007 (86)
44 sALS,
4 fALS
8 OND ↓ FRA in ALS CSF
↑ AOPP in CSF and
plasma ALS
Murata et al,
2008 (87)
30 sALS 17 HC ↑ percentage of oxidized
CoQ10 in CSF sALS,
correlation oxidized
CoQ10 and duration of
sALS, not correlation with
the ALS score
Keizman et al,
2009 (88)
86 86 HC ↓ ALS uric acid level;
correlation serum uric
acid levels and rate of
progression ALSFRS-R
Cova et al, 2010
(89)
88 sALS 50 HC ↓ all assayed enzymes
and correlation with
progression rate in sALS,
plasma ROS significantly
lower in sALS
Mendonça et al,
2011 (90)
10 6 HC No increased levels in
ALS

AD = Alzheimer disease, sALS = sporadic amyotrophic lateral sclerosis, fALS = familial amyotrophic lateral sclerosis, ALSFRS-R = Amyotrophic Lateral Sclerosis Functional Rating Scale- revised, AOPP = advanced oxidation protein products, CAT = catalase, CoQ10 = coenzyme Q10, CSF = cerebrospinal fluid, FRA = ferric reducing ability, GPX = glutathione peroxidase, GR = glutathione reductase, GSH = glutathione, GSSG = Glutathione disulfide, GST = glutathione transferase, G6PD = glucose-6-phosphate dehydrogenase, HC = healthy controls, HNE = 4-hydroxy-2,3-nonenal, F2-IsoPs = F2-isoprostanes, ND = neurodegenerative diseases, NND = non-neurodegenerative disease, ROS = reactive oxygen species, SOD = superoxide dismutase, T = tyrosine, TAS = serum total antioxidant status, TBARS = thiobarbituric acid-reactive substances, 3-NT = 3-Nitrotyrosine, 8-oxodG = 8-oxo-deoxyguanosine

a

Phase II clinical trial: 20 subjects with ALS received either 30 mg (5 subjects) or 60 mg (15 subjects) of edaravone (free radical scavenger) via intravenous drip once per day. Two weeks of administration was followed by a two-week observation period, for six times. In almost all patients in the 60 mg group, the level of CSF 3NT, was reduced at the end of the sixth cycle of administration.

In summary, biomarkers of oxidative stress and the methods used to measure them vary among sALS studies, making comparison of study findings difficult. Furthermore, whether oxidative stress is an inciting or a downstream event during the already ongoing degenerative process remains to be determined. Yet, it is clear that oxidative stress is likely to be a major player in the damage that occurs to DNA and pathologic changes in lipid and protein metabolism (19) .

ENVIRONMENTAL AND LIFESTYLE RISK FACTORS ASSOCIATED WITH sALS

In this section, we review environmental exposures and life style factors that have been associated with ALS that also may be mechanistically involved with underlying oxidative stress. Table 2 summarizes potential relationships among environmental and lifestyle factors and sALS and oxidative stress.

Table 2.

Environmental and Lifestyle Factors: Relationship to ALS and Mechanisms of OS

Factor Strength of the
Association
Mechanisms of Oxidative Stress Toxicity
Agricultural
chemicals
Uncertain (9199, 104, 193) Apoptosis and ROS-induced DNA damage (114)

PON1 genotype (263); protein carbonyl
production (116) mitochondrial damage (122)
Lead, other heavy
metals
Uncertain (92, 94, 104, 127, 128, 130, 353) ↓ cellular GSH (133); ↓ copper homeostasis
(134); combined toxicity with other metals
(136, 137)
Military service Uncertain (144146,
148151, 153)
Head trauma and ApoE4 genotype (154)
Trauma Uncertain (101, 154
164, 169)
Possibly via free radical production (ROS)
(172)
Strenuous physical
activity
Uncertain (189) ↓ GSH and ↑ apoptosis (186); ↑ free radicals
(187, 354)
Professional football Suspected (165
168)
Physical activity per se may not be the cause
(166, 190)
Diet (high in fat,
glutamate)
Uncertain (193195) ↑OS, inflammation, and NF-κB activation in
the cerebral cortex (304, 306)
Smoking Suspected (154, 156, 200209) Protein and DNA damage, cell death (213),
↑ heavy metal levels (216)
Malnutrition/Cachexia Clinically observed
(301, 303)
↑ OS (300)
Hypoxia Clinically observed
(307)
ROS production via mitochondrial dysfunction
(308);
↑ DNA, lipid, protein oxidation (312, 313)
Psychological stress Clinically observed
(315, 316)
OS and shorten telomere length (317);
induction of cytokines, apoptosis, and OS
(319)

ApoE = Apoliprotein E, GSH = glutathione, OS = oxidative stress, PON1 = Paraoxonase 1, ROS = reactive oxygen species.

Agricultural chemicals

Organophosphate pesticides, until their recent ban, were among the most widely used synthetic chemicals for controlling domestic and agricultural pests. Several epidemiological studies suggest that exposure to organophosphate pesticides is associated with sALS (91102). In 2003, a new evidence-based medicine method to evaluate the quality of ALS epidemiological studies was proposed (103). It includes assessment of 1) general methodological criteria (selection of control group, high response rate, blinding, recall bias, quantification of exposure, accounting for confounding and bias, and appropriate analytical approach); 2) the diagnostic certainty of ALS based on established criteria; and 3) temporal precedence of exposure and disease onset (103). Based on these criteria, Armon identified two population-based, one case-control, and one mortality surveillance study (91, 98, 101, 102). Further stringent criteria and a new method to qualify exposure accuracy and reproducibility have subsequently been developed based on 1) self-reported exposures, 2) assigned exposure by an expert or computer-based assessment of exposure levels, and 3) evidence of external exposure, internal exposure (e.g., for metals) or a biomarker of exposure (usually a metabolite) of suspected agents (e.g., for pesticides) (104). When one includes these critical assessment techniques, only two studies, both population-based, meet these criteria (91, 94). Both demonstrate that the risk for sALS is associated with pesticides exposure.

One of these two investigations (91) is a population-based case-control study of 174 cases and 348 controls in Washington state, matched according to sex and age, during 1990 through 1994, when the state had a population of approximately 2.5 million (105) . After adjustment for age and education, exposure to agricultural chemicals was found to be associated with sALS (OR = 2.0, 95%CI = 1.1 – 3.5); this association was observed for men (OR = 2.4, 95%CI = 1.2 – 4.8) but not for women (OR = 0.9, 95%CI = 0.2 – 3.8) (91). Park et al (94) conducted a mortality surveillance in a population over 2 million in 22 states during 1992–1998 and analyzed the association of mortality with neurodegenerative disease, including motor neuron disease (MND), in occupational groups previously described (99) and occupations with probable pesticide exposure: 1) all farming occupations, 2) farming occupations with likely pesticide exposure (farm workers, horticultural specialty workers etc.), and 3) farmers alone (99). Risk for MND was strongest for farmers (OR = 1.23, 95% CI = 1.03 – 1.46). However, in other occupations where pesticide use might be expected (e.g., farm workers, horticultural specialty workers), significant elevations were not observed (94).

Although not fulfilling the strict criteria described by Sutedja et al (104), two case-control studies (93, 95), originating from Europe (93) and Oceania (95), showed that pesticide exposure was positively associated with ALS. The first one showed that the number of incident cases of ALS in which the patient's usual occupation was in agricultural work exceeded the expected number (observed ALS cases = 22, 95% Poisson CI 13.8–32.3, expected ALS cases = 6) (93). A case-control study performed in Oceania showed a higher risk of developing sALS with exposure to industrial herbicides, pesticides, or both (OR = 4.18, 95% CI = 1.79–9.74, P = 0.006) but not for farm-based herbicides or pesticides (95).

A population-based case-control study (97) and a case-control study (96), both European and published after the critical assessment techniques described above were developed, showed very similar ORs for pesticide exposure (OR = 3.6; 95% = CI 1.2–10.5, and OR = 3.04; 95% CI = 1.1–7.7, respectively). Moreover, the American Cancer Society's recent prospective Cancer Prevention Study II, with nearly 1.2 million participants (106), evaluated the relationship between regular exposure to 11 different chemical classes or × rays and sALS mortality (92). Follow-up from 1989 through 2004 identified 617 deaths from ALS among men and 539 among women but found no statistically increased risk of ALS mortality by self-reported exposure to pesticides, herbicides, or both (OR = 1.07; 95%CI = 0.79 – 1.44). Regarding the duration of exposure, ORs for those reporting < 4 years’ exposure were 0.62 (95%CI = 0.09 – 4.45), for 4 to 10 years, 1.92 (95% CI = 0.71 – 5.19), and for >10 years of exposure, 1.48 (95% CI = 0.82 – 2.67) (92).

Therefore, epidemiological evidence of an association between exposure to agricultural chemicals and sALS is present but modest and thus is, at best, suggestive. If the association between agricultural pesticides and sALS is small to modest and if a specific genotype modifies the association, then even large-scale case-control and cohort studies may not be effective in finding consistent associations between organophosphate-induced oxidative stress and sALS.

Most agricultural organophosphates can be clearly linked to oxidative stress (107110). Among these, chlorpyrifos, parathion, and malathion have been shown to impair GSH homeostasis by decreasing GSH levels (111). GSH is a tripeptide (L-γ-glutamyl-L-cysteinyl-glycine) with multiple functions in living organisms (112). It can be directly oxidized by hydroxyl radical (HO•) and peroxynitrite (ONOO−) or indirectly during GSH-dependent peroxidase-catalyzed reactions (113). Increased oxidative stress biomarkers (urinary 8-oxodG, malondialdehyde), reduced GSH levels and DNA damage (COMET assay) are observed in organophosphate-treated lymphocyte cell cultures (109, 114).

Although the molecular mechanism underlying organophosphate induction of oxidative stress remains to be fully clarified (115), active metabolites (various oxon compounds), that are enzymatically converted from organophosphates and able to delete intrinsic antioxidants, have been identified in the human liver, lung, and brain tissues (116118). Paraquat, a common herbicide, can induce GSH depletion in the CNS (119). In a neuron-microglia culture system, paraquat was toxic to neurons even at low concentrations (120). In this system, paraquat activates microglial NADPH oxidase generating superoxide, which is well studied in the microglia (120, 121). Moreover, organophosphate uptake into the mitochondria across the mitochondrial inner membrane leads to reduction by complex I, resulting in redox cycling and mitochondrial toxicity (122). Paraquat not only weakly inhibits complex I but impairs complex III function as well, and the respiratory chain disruption generates more oxidants (121, 123).

Lead exposure

Exposure to metals is an intriguing potential suspect in the cause of sALS, with many case reports linking different metals to sALS (124). Furthermore, some investigators have found higher levels of certain metals in the blood, bone, CSF, urine, or spinal cords of patients with sALS compared to controls [reviewed in (125)]. The role of metals exposure has also been investigated in epidemiological studies (92, 94, 126130). Despite many investigations, the role, if any, of these metals in the pathogenesis of sALS remains unclear [reviewed in (104)]. A study by Kamel et al (128) is particularly noteworthy, however, because they longitudinally assessed lead exposure in sALS. In a case-control study, they recruited 109 ALS cases from two major referral ALS centers in New England from 1993 to 1996 along with 256 controls (matched to cases by age, sex, and region of residence), identified by random-digit dialing. They collected information on occupational, residential, and recreational exposure to lead using a structured interview. In addition, they measured blood and bone lead concentrations (blood was collected from 107 cases and 39 controls; bone lead was measured at two sites, the mid-tibial shaft and the patella, in 104 cases and 41 controls). The risk of ALS was associated with self-reported occupational exposure to lead (OR = 1.9; 95% CI = 1.1 – 3.3). Moreover, risk of ALS was associated with elevations in both blood and bone lead levels: the OR was 1.9 (95% CI = 1.4 – 2.6) for each µg/dL increase in blood lead, 3.6 (95% CI = 0.6 – 20.6) for each unit (µg /g) increase in log-transformed patella lead, and 2.3 (95% CI = 0.4 – 14.5) for each unit (µg /g) increase in log-transformed tibia lead.

Genotyping for known SNPs in the δ-aminolevulinic acid dehydratase (ALAD) gene (129) revealed that the ALAD 2 allele (177G to C; K59N) was associated with decreased lead concentration in both patella and tibia, although not in blood; it showed a trend toward an increased ALS risk (OR = 1.9; 95% CI = 0.60 – 6.3), although the confidence interval was wide, and the association was not statistically significant. Further, a previously unreported ALAD SNP at an Msp1 site in intron 2 (IVS2+299G>A) was associated with decreased bone lead concentration and a non-significant trend toward a decreased ALS risk (OR = 0.35; 95% CI = 0.10 – 1.2). The ALAD enzyme is the principal lead-binding site in erythrocytes, and the ALAD 2 protein binds lead more tightly than the ALAD 1 protein (131). This change in intron 2 alters the lead toxicokinetics and may modify the risk associated with lead exposure (132).

More recently, Kamel et al. (130) obtained follow-up data for 91% of the patients who participated in the original case-control study (128). Surprisingly, lead exposure appeared to directly correlate with longer survival time among ALS cases (OR = 0.5; 95% CI = 0.2 – 1.0).

Environmental lead exposure markedly reduces GSH levels and induces caspase 3 and prostaglandin E2 activation in neuroblastoma cells (133). In astroglia, lead also causes neuronal oxidative injury by interfering with copper homeostasis (134) and by inducing glial heme oxygenase synthesis (135). Collectively, this evidence suggests that lead alone or in combination with other biochemical stressors leads to oxidative injury (136, 137). Further laboratory studies have shown that astrocytes can sequester and buffer lead in primary co-culture with neurons, modulating its diffusion (138). In particular, astrocytes induce neuronal cytoprotective and antioxidant gene expression in response to lead exposure (139). In immortalized human fetal astrocytes, this expression results in increased vascular endothelial growth factor (VEGF) (135) and suggests that VEGF protects motor neurons in culture and in organotypic culture (140142). However, further studies are clearly needed. Recent evidence has shown that lead exposure can cause epigenetic changes, which can lead to alterations in the transcription and subsequent translation rates of genes and proteins throughout the genome (143).

Military deployment

Although controversial, two studies suggest an approximately 2-fold increase in the risk of sALS among veterans of the 1991 Gulf War (144, 145). A study involving only Gulf War veterans under the age of 45 also found an elevated risk of sALS in this population (146).

Subsequently, a secondary analysis of these data showed that the increased risk of ALS was limited to the decade following the war and also to deployed military personnel only, suggesting that exposure to potential neurotoxic agents occurred over a relatively brief period of time (147). However, previous case-control (148) and prospective (149) studies found no association between deployment and ALS among Gulf War veterans.

In 2005, Weisskopf et al (150) prospectively assessed the relation between military deployment and sALS mortality among participants in the American Cancer Society Prevention Study II, a cohort that included more than 500,000 men (106). They compared cause-specific mortality between 281,874 deployed military personnel, excluding Gulf War veterans (military service was ascertained in 1982), and 126,414 men who did not serve in the military. Of 280 deaths due to ALS, 217 occurred in the military group and 63 in the group who had not served, indicating that those who served in the military had an increased death rate from ALS (OR = 1.53; 95% CI = 1.12 – 2.09). Furthermore, when the relation between ALS mortality and the different branches of military service were analyzed, mortality was greater among those who served in the Army or National Guard (OR = 1.54; 95% CI = 1.09 – 2.17), the Navy (OR = 1.87; 95% CI = 1.28 – 2.74), Air Force (OR = 1.54; 95% CI = 0.99 – 2.39), and Coast Guard (OR = 2.24; 95% CI = 0.70 – 7.18), not in the Marines. The increased risk of ALS mortality was similar among those who served in World War II (OR = 1.60; 95% CI = 1.12 – 2.30), Korea (OR = 1.54; 95% CI = 0.92 – 2.60), or Vietnam (OR = 1.44; 95% CI = 0.47 – 4.47) compared with those who had never served.

In a recent study, various factors (demographics, medical history, major traumatic injury, smoking status) and aspects of military service [(cumulative time of military service, primary branch of service (Air Force, Army, Coast Guard, Marines, or Navy), and deployment (in Korea, Vietnam, or the Persian Gulf regions)] were investigated to evaluate their influence on the survival in a group of more than 1000 military veterans from across the US enrolled in the National Registry of Veterans with ALS (151). In this group, 14% reported past deployment to Korea, 21% to Vietnam, and 5% to the Persian Gulf; 4% reported deployment to more than one location. Past deployment in Vietnam compared to other locations was associated with shorter survival in ALS (OR = 1.73; 95%CI = 1.36 – 2.19), suggesting the possibility that patients who were deployed in Vietnam shared some common exposure (e.g., virus, toxin) that worsened the effects of ALS once it developed. One of the biases of this study is that only veterans alive in 2003 and beyond are in the ALS registry.

Obviously a single individual may be exposed to multiple different potential risk factors. A 2012 case-control study of 184 veterans with ALS and 194 control subjects from the National Registry of US Veterans (152) found that blood lead concentration was higher among ALS cases compared with controls (P < 0.0001, adjusted for age) and a doubling of blood lead was associated with a 1.9-fold increased risk of ALS (95% CI = 1.3 – 2.7) (153). However, it is possible that the wasting associated with ALS leads to increased bone turnover and lead leaching from bone into the blood. Thus, higher blood lead levels may be possible in ALS without being causal.

A recent cohort study of more than 2,000 medical records of US veterans from April 2003-September 2007 (152) included data on 241 incident ALS cases and 597 controls. The study authors found that veterans who had experienced head injuries during the 15 years before the reference date (the date of the ALS diagnosis for cases and the date of the interview for controls) had an adjusted (age, sex, race/ethnicity and education) OR of 2.33 (95% CI = 1.18 – 4.61) (154). Moreover, a gene-environment interaction analysis of 221 cases and 476 controls that included cigarette smoking (never vs. ever), head injury (reported by 79 cases and 183 controls at least one time), and apolipoprotein E (ApoE) genotype (homozygous ApoE-3 carriers vs. ApoE-2 carriers vs. ApoE-4 carriers) reported weak evidence for the interaction; the association between ALS and head injury was stronger in ApoE-4 carriers (likelihood ratio test of 2.7 1 df, P = 0.10) than in ApoE-2 carriers (0.2; 1 df, P = 0.68) (154). However, the very small sample size pertaining to the ApoE analysis is a limitation.

Trauma

Although an association between head trauma and sALS is tenuous (101, 155164), the report in US veterans discussed above suggests that the potential relationship to brain injury may deserve further exploration. The finding of a very high incidence of sALS among Italian professional footballers (165167) and American football players (168) has led to speculation over whether head injury caused by collision is a relevant cause. A rigorous population-based study of sALS in western Washington state found that sALS was not associated with previous fractures, head injuries, or hospitalizations (101). In a recent cohort study, the adjusted rate ratio for sALS after head injury was 1.5 (95% CI = 1.1 – 2.1), but this elevated risk was found only within the first year after injury, possibly suggesting that very early subclinical weakness can increase the risk of head injury due to falls and fractures in sALS (164). A recent review evaluated the epidemiological literature regarding the association between head trauma and ALS published between 1980 and October 2010 (169), using the American Academy of Neurology evidence-based classification of evidence for inferring causality and assigning level of conclusion (170). Twelve articles (one with Class II evidence, three with Class III evidence, and eight with Class IV evidence) met the inclusion criteria. Analysis showed that a causal relationship between a single instance of head trauma or head injury and occurrence of ALS is unproven or unsupported (Level U conclusion) (169).

During a traumatic brain injury, the brain and spinal cord undergo shear deformation, which transiently elongates axons. Traumatic axonal injury also perturbs the cytoskeleton, causing microtubule and neurofilament dissolution and pathologic reorganization of neurofilament proteins (171). Trauma to the CNS triggers stress responses that include oxidative stress due to ROS generation (172). Necrotic events due to cellular disintegration resulting from an acute injury are associated with release of cellular contents, eliciting an inflammatory response and oxygen radical production.

Repetitive head injury is associated with the development of chronic traumatic encephalopathy (CTE), a tauopathy characterized by neurofibrillary tangles throughout the brain in the relative absence of β-amyloid deposits. McKee et al. (173) described 12 cases of CTE, 10 of which had widespread TAR DNA-binding protein-43 (TDP-43) inclusions (TDP-43 proteinopathy). Among these cases, 3 were athletes who developed progressive MND that clinically resembled an ALS-like syndrome. TDP-43 seems to be critical in mediating the response of the neuronal cytoskeleton to axonal injury (174). It is intrinsically prone to aggregation, and TDP-43 expression is upregulated after experimental axotomy in mouse spinal motor neurons (175). Conceivably, traumatic axonal injury may also accelerate TDP-43 accumulation, aggregation, and translocation to the cytoplasm and thereby enhance its neurotoxicity. Even though the number of cases was small, the coexistence of any MND phenotype and CTE neuropathology has raised interest in the ALS community.

The importance of TDP-43 in ALS quickly became evident when researchers discovered TDP-43 inclusions in ALS and frontotemporal dementia (FTD) (176, 177). TDP-43 inclusions are now known to be present in the majority of ALS cases, including ALS-FTD, sALS, non-SOD1 fALS, and the ALS-Parkinson disease (PD) complex in Guam (178181). Recent studies suggest that oxidative stress may have a close relationship to TDP-43 and its pathologic alteration. Cohen et al. (182) demonstrated that oxidative stress promotes TDP-43 cross-linking via cysteine oxidation and disulphide bond formation, leading to decreased TDP-43 solubility. Moreover, prostaglandin metabolites, which are major players in oxidative stress and inflammation, can affect TDP-43 proteolysis, solubility, and subcellular localization (183). Chronic exposure of cultured neuron-like (SH-SY5Y and NSC34) cells to ROS and RNS via paraquat and ethacrynic acid administration revealed TDP-43 C-terminal phosphorylation, insolubility, C-terminal fragmentation, and translocation of nuclear TDP-43 to the cytosol (184, 185). Therefore, it appears that oxidative stress may play an important role in TDP-43 proteinopathy, further suggesting an intriguing relationship to sALS disease mechanisms.

Strenuous exercise and physical exertion

Strenuous exercise diminishes lymphocyte GSH content, resulting in oxidative stress-induced apoptosis, and increases urinary excretion of 8-oxodG, whereas moderate exercise attenuates lymphocyte apoptosis, possibly by improving antioxidative capacity (186). Strenuous exercise generates a host of complex biological changes; however, evidence is sufficient to suggest that it increases free radical production that leads to oxidative stress (187).

A study of aerobic exercise (incremental workload on a cycloergometer) in 10 patients with ALS, 8 of whom had sALS, assessed oxidative stress by measuring blood lipoperoxides (188). Values were significantly higher in patients with ALS (P < 0.05) than in control subjects (n = 5 with various chronic neuropathies) at rest, during exercise, and 30 minutes afterward. Moreover, the increase in lipoperoxide concentration during exercise strongly and positively correlated with lactate accumulation (r = 0.94, P < 0.01), a finding suggesting impaired mitochondrial function (188).

Epidemiological studies investigating an association between strenuous physical activity and sALS suggest that more evidence is needed to establish a possible causative role. In a case-control study of 279 patients with ALS and 152 with other neurologic diseases, a greater number of subjects with ALS reported they had been varsity athletes (OR = 1.7, 95%CI = 1.04 – 2.76) and had always been slim (OR = 2.21, 95%CI = 1.40 – 3.47) compared to disease controls (189). A recent matched case-control study has been performed in a large cohort of more than 680,000 Swedish male subjects born between 1951 and 1965 (190). Data on body weight and height, physical fitness, resting heart rate, and isometric strength measured at military conscription (age 18–19 years) were collected. The population was followed through December 2006, and 85 men died from ALS during this period. The authors found that not physical fitness per se but rather weight-adjusted physical fitness, one of the strength measures, was a risk factor for ALS (OR = 1.98, 95%CI = 1.32 – 2.97). They raised the possibility that unknown genetic factors that are involved in muscle function and modulate human strength and endurance could be the link between weight-adjusted physical fitness and ALS (190). In Italian professional soccer players, a higher risk for sALS was found among midfielders (standardized morbidity ratio of 12.2, 95%CI = 3.3 – 31.2), a role characterized by great aerobic effort and a higher proportion of lean body mass (165). But an absence of sALS cases in professional road cyclists and basketball players, who also have a high level of aerobic fitness and higher proportion of lean body mass (191), suggests that physical exertion or fitness may not explain a causal relationship with sALS. Further studies are clearly needed (166, 192).

Diet

A population-based, case-control study in western Washington state from 1990 to 1994 showed that increased dietary fat intake was associated with a greater risk of ALS (OR = 2.7, 95%Cl = 0.9 – 8.0) as was glutamate intake (OR = 3.2, 95%Cl = 1.2 – 8.0), whereas dietary fiber intake was associated with a decreased risk (OR = 0.3, 95%Cl = 0.1 – 0.7) (193). In 2008, Morozova et al (194) examined the relation between diet and risk of ALS among participants in the American Cancer Society Prevention Study II cohort (a population of nearly 1.2 million subjects) (106). No association was found. In a more recent case-control study, 153 sALS and 306 gender- and age-matched controls randomly selected from the general population in the Tokai area of Japan completed a self-administered food frequency questionnaire to estimate pre-illness intake of food groups and nutrients. A high intake of carbohydrates was significantly associated with an increased risk of ALS (OR = 2.14, 95%CI = 1.05 – 4.36) (195).

One micronutrient, vitamin E, deserves brief discussion as an extrinsic anti-oxidant. It is a fat-soluble hydrocarbon and an important component of cell membranes and lipoproteins. (196). Its antioxidant properties are attributed to the inhibition of lipid peroxidation and free-radical formation, because vitamin E can attack the peroxyl radical more rapidly than can polyunsaturated fatty acids, donating its phenolic hydrogen atom to the radical and converting it to a more stable product (197). Vitamin E might have a potential protective role in preventing the development of ALS (198) while also slowing disease progression (199). A mortality surveillance study conducted in a well-defined population [participants in the American Cancer Society Prevention Study II (106)] indicated that regular use of vitamin E supplements for 10 years or more is associated with a lower risk of dying of ALS (relative risk 0.38, 95% CI 0.16–0.92, based on trend analysis, P=0.004) (198). Further, a recent pooled analysis of mortality data from 5 different prospective cohort studies (among 1,055,546 participants, 805 developed ALS) revealed a positive trend (P=0.01) for a decline in ALS progression within patients (231 cases) with longer duration of vitamin E supplementation but found no overall protective role for vitamin E (199).

Smoking

Several studies, population-based and case-controlled, have established a probable relationship between sALS and cigarette smoking, as shown in Table 3 (154, 200209), although some results have been inconsistent (210, 211). Methodological differences and heterogeneity in studied populations could account in part for these disparities (103, 212).

Table 3.

Evidence-based Reviews of Smoking and ALSa

Author
year of
publication
Study
Population
Study
Design
Number of
Subjects
(case/controls)
Case
ascertainment
Adjustment
variables
OR
(95%
CI)
Kamel et
al, 1999
(202)
Two-
centers
study
Case-
control
110/256 ALS Clinic age,
gender,
education,
region
1.70
(1.00

2.80)
Nelson et
al, 2000
(201)
Population
basedb
Case-
control
161/321 Multiple case
ascertainment
sources
age,
gender,
education,
alcohol
2.00
(1.30

3.20)
Weisskopf
et al, 2004
(200)
Mortality
surveillance
c
Cohort 291/638,849
(women)
National
Death Index
age,
alcohol,
education
1.67
(1.24

2.24)
Weisskopf
et al, 2004
(200)
Mortality
surveillance
c
Cohort 330/459,360
(men)
National
Death Index
age,
alcohol,
education
0.69
(0.49

0.99)
Fang et al,
2006 (208)
Constructio
n workersd
Cohort 160/280,558 Registry age, area of
residence
0.80
(0.60

1.10)
Sutedja et
al, 2007
(204)
Single
center
study
Case-
control
364/392 ALS clinic education,
occupation,
age
1.6
(1.00

2.50)
Gallo et al,
2009 (203)
Multicentere Cohort 118/517,890 Death
certificates
age,
gender,
education,
center of
recruitment
1.89
(1.14

3.14)
Okamoto
et al, 2009
(207)
Multicenter Case-
control
153/306 ALS clinic age, gender 1.00
(0.60

1.30)
Schmidt et
al, 2010
(154)
Military
veterans
Case-
control
241/597 Registry age,
gender,
race,
education
0.90
(0.56

1.46)
Alonso et
al, 2010
(206)
Cohortf Case-
control
1143/11,371 Death
certificates
age at
diagnosis,
gender,
diagnostic
classificatio
n
1.04
(0.80

1.34)
Wang et al,
2011 (205)
Cohortg Case-
control
832/1,119,080 Death
certificates,
registries
age,
gender,
physical
activity,
education,
body mass
index
1.42
(1.07

1.88)
de Jong et
al, 2012
(209)
Population
based
incidence
cohort
Case-
control
494/1,599 Multiple
sources (ALS
clinics,
rehabilitation
centers,
patient
support
associations)
age,
gender,
education
1.38
(1.02

1.88)
a

Studies of more than 100 ALS patients unless they are population-based

b

Three counties of western Washington state (King, Pierce, and Snohomish) in the US

c

Participants were recruited by American Cancer Society in 50 States; families with at least one member over the age of 45 and other family members over the age of 30 were invited to participate (Cancer Prevention Study II cohort). Vital status of the study participants has been determined by automated linkage with the National Death Index through December 31, 1998.

d

Swedish Construction Workers Cohort

e

Subjects (aged 35–70) recruited from the general population residing in a given geographical area from 1991 to 2001, in 23 centers across 10 European countries (European Prospective Investigation into Cancer and Nutrition cohort)

f

General Practice Research Database, a computerized clinical database in the United Kingdom

g

Study population comprised of participants in the following: the Nurses’ Health Study, the Health Professionals Follow-up Study, the

Cancer Prevention Study -II Nutrition Cohort, the Multiethnic Cohort, and the National Institutes of Health American Association of

Retired Persons Diet and Health Study (NIH-AARP)

Cigarette smoking is associated with oxidative stress, inducing protein oxidation, DNA damage, and cell death (213, 214). It is hypothesized that cigarette smoking causes lipid peroxidation via formaldehyde formation (215) and increases cadmium and lead levels in seminal fluid, plasma and blood; all of which may increase oxidative damage (216).

A recent pooled analysis prospectively examined the relation between smoking and ALS in 5 well-established large cohorts with study-specific follow-up, ranging from 7 to 28 years (205). Among 562,804 men and 556,276 women, 832 ALS cases were documented. Smokers had a higher risk of ALS than those who never smoked, with age- and sex-adjusted relative risks of 1.44 (95% Cl = 1.23 – 1.68) for former smokers and 1.42 (95% Cl = 1.07 – 1.88) for current smokers (205).

Combined Effects of Multiple Stressors

Undoubtedly, concurrent exposure to environmental and lifestyle factors occurs. In experimental models, combined exposure to glutamate and lead as compared to either agent alone increased neuronal cell death via mechanisms involving an elevation in the production of oxygen radicals and a decrease in intracellular GSH defenses against oxidative stress (217). Exposure of mouse embryonic (day 12) spinal cord and dorsal root ganglion cells to low levels of paraquat, exogenous glutamate, and thermal stress, both singly and in combination (218), revealed that paraquat administered alone was associated with cell death in a dose-dependent manner, an effect that was multiplied by heat shock. This model is particularly intriguing because a combination of low-level oxidative stress factors was associated with motor neuron vulnerability to environmental stressors. Such low-level cumulative stress may, in part, explain the mid- to late-life symptom onset in both familial and sALS patients. Other investigations (219) found various combinations of stressors differentially affected enzymatic and non-enzymatic antioxidant defense systems, protein oxidation, and lipid peroxidation in the brain.

These experiments are important because they attempt to mimic the human situation in which multiple stressors are likely to be present, resulting in additive or multiplicative interactions that contribute to abnormal oxidative stress. Thus, multidisciplinary epidemiological studies are needed that consider joint associations between demographic, biologic and genetic risk factors to better understand the disease mechanisms and eventually identify markers of susceptibility in patients with sALS.

INTRINSIC ANTIOXIDANTS

Little is known about the role of intrinsic antioxidants in sALS. Serum total antioxidant status, a measure of peroxyl adduct-scavenging capacity, has been reported to be significantly (P < 0.05) higher in ALS patients (n = 28) compared with that of healthy controls (n = 20) but did not correlate with ALS onset phenotype, disease duration, or clinical state (220). More recently, the TBARS concentration and antioxidants such as SOD1, catalase, GSH peroxidase, GSH reductase, and glucose-6-phosphate dehydrogenase were assessed in the erythrocytes of 20 sALS patients and 20 controls (221). In sALS, the TBARS concentration was significantly increased with respect to that of controls (P <0.001); moreover, TBARS started to increase and antioxidant molecule concentration began to decrease as sALS progressed over 6 to 24 months, suggesting a correlation between these variables and duration of disease (221). In a second study, anti-oxidative stress markers were examined in 31 patients with ALS, 24 patients with PD, and 30 healthy subjects. SOD activity was significantly decreased in ALS (P = 0.001), whereas GSH peroxidase activity was decreased in PD (P = 0.001), suggesting that the affected antioxidant system might differ between ALS and PD (222).

SOD1

SOD1, or Cu/Zn-SOD, was the first identified and more comprehensively studied antioxidant enzyme of the SOD family; mitochondrial Mn-SOD2 and extracellular Cu/Zn-SOD3 also exist [for review see (25, 223)]. SOD1 catalyzes dismutation of O2 - to H2O2 and O2 in a two-step redox reaction involving the reduction and reoxidization of the copper ion in the active site of the dimeric protein.

The role of SOD1 in ALS pathogenesis has received great attention since the first description of mutations in the gene encoding SOD1 (224). Since then, more than 160 mutations have been reported. They account for approximately 15% – 20% of fALS cases, and thus SOD1 mutations are the second most frequent known cause of fALS after C9ORF72 (8, 9, 225). How mutant SOD1 leads to the death of motor neurons has not been established, although a number of hypotheses have been presented (25, 226229). Importantly, the cause of motoneuron degeneration in SOD1 mutation carriers is not the result of a loss of anti-oxidative function of the SOD1 enzyme, but instead, a gain of function appears to be pathogenic. We refer here to several excellent papers since this topic is beyond the scope of this review (25, 229, 230).

Although SOD1 mutations can cause fALS, wild-type SOD1 protein might also have a role in sALS pathogenesis. First, oxidized wild-type SOD1 occurs in lymphocytes of sALS patients, resulting in mitochondrial dysfunction (231). Moreover, misfolded SOD1 is found not only in sALS motor neurons (232, 233), but also in the motor neurons of FUS- or TDP43-linked fALS cases - these are cases with no associated SOD1 mutation(234). The mechanisms underlying how wild-type SOD1 is misfolded in these ALS cases remain to be clarified (229, 235). On the other hand, if this process is a frequent phenomenon in sALS or other types of fALS, SOD1 misfolding might explain a common molecular event between fALS and sALS (234).

Paraoxonase

Paraoaonase (PON) is a potential intrinsic antioxidative enzyme (236, 237). PON hydrolyzes paraoxon, which is a metabolite of parathion – a highly toxic organophosphate; exposure can lead to acute cholinergic crisis and subacute or chronic neurotoxicity. PON is a family of 3 independent enzymes: PON1, PON2, and PON3. The genes are located on chromosome 7 (238). PON1 is synthesized in the liver, and a portion is secreted into the plasma as a component of HDL (239).

Despite its name, PON1 is not very efficient in hydrolyzing paraoxon, which accounts in part for its toxicity. The mechanisms of organophosphate hydrolysis by PON1 have been well studied in laboratory animals, and oxidative stress occurs because organophosphate detoxification consumes GSH (240). On the other hand, oxidative stress may reduce PON1 activity (241). In fact PON1 is highly susceptible to hydroxyl radicals produced from metal (Cu2+ or Fe2+) -catalyzed oxidation, and the modification of some histidine residues results in the decrease of its antioxidant activity (241).

PON1 has been extensively studied in cardiovascular disease because of its ability to protect against atherosclerosis (242). Certain SNPs in PON1 have been associated with differential susceptibility to organophosphate exposure (243): two SNPs, Q192R (rs662) and L55M (rs854560) (244) have the greatest paraoxon hydrolytic activity but less effectively protect against low-density lipoprotein (LDL) oxidation (245). Decreased PON1 activity is associated with an increased risk of coronary heart disease (246) and organophosphate toxicity (247). In laboratory animals, PON1 decreases oxidation of LDL by hydrolyzing hydroperoxides into less reactive hydroxides (242). Moreover, PON1 has hydrolytic activities on aromatic and long-chain aliphatic lactones, including arachidonate and lipid mediators generated from the oxidation of polyunsaturated fatty acids and exogenous lactone-containing drugs, such as prulifloxacin. Statin lactones (simvastatin, lovastatin) and the diuretic spironolactone, previously reported to be hydrolyzed by PON1, are metabolized by PON3 (248). PON2 and PON3 lack the ability to hydrolyze organophosphates but have lactonase or peroxidase-like activity or both (249, 250). Like PON1, PON3 is also found in HDL and serum. PON2 is expressed in a number of tissues, including the brain (251, 252). A human cell line overexpressing PON2 had significantly less intracellular oxidative stress (as measured by a nonspecific fluorometric assay for oxidative stress) following treatment with H2O2 (251). Serum PON1 activity in a given human population can vary by 40-fold. Although a portion of this variation is explained by extensive genetic variation in the PON1 coding regions (more than 160 SNPs have been described), activity may be largely influenced by environmental and perhaps even epigenetic factors (253). Age plays the most relevant role, at least early in life, as PON1 activity is very low before birth and gradually increases during the 1 to 2 years after birth in humans (254). PON1 activity may also decline with aging, possibly because oxidative stress increases with age (255). A sex effect has also been proposed, as females have higher PON1 activity (253). A variety of other factors (environmental chemicals, drugs, smoking, alcohol, diet, disease conditions) are suspected to reduce PON1 activity (253). On the other hand, statins increase PON1 expression and activity in vitro studies (256, 257). PON1 activity increased by ~13% in patients who took low-dose acetylsalicylic acid (258).

PON1 SNPs have been reported in ALS (259, 260), but subsequent results have been conflicting (261268). A meta-analysis of 10 published studies and 1 unpublished study on PON SNPs found no significant association with sALS (269). A recent case-control study involving 1,160 sALS patients and 1,240 control subjects of Dutch descent that examined PON1 and PON3 genetic variants showed that the SNP frequencies did not differ between the two groups (268). Thus, work to date indicates that PON1 SNPs contributing to ALS either do not exist or, if they do, seem to not be easily detected. If SNP variation is a factor, it may only have an effect in a susceptible minority of patients. A different genetic and epidemiological approach may be required to detect such abnormalities (270).

MODIFIER GENES ASSOCIATED WITH sALS AND OXIDATIVE STRESS

In addition to PON1, other possible genetic factors modifying sALS disease expression have been reported: ApoE, survival motor neuron (SMN), inducers of angiogenesis [such as VEGF and angiogenin (ANG)], and other genes involved in the regulation of many cellular processes, including some of the known RNA processes (e.g., transcription and post-transcriptional and translational regulation).

The role of ApoE in sALS is uncertain. It was suggested that the ε4 allele has a deleterious effect on ALS survival (271, 272) and association with younger age at ALS onset (273), but these findings were not subsequently confirmed (274). ApoE is a widely distributed, well-characterized cholesterol transport protein that circulates in the plasma after being synthesized by the liver, spleen, and kidneys (275). It is the major apolipoprotein in the CNS, where it is synthesized by glia, macrophages, and neurons (276). Animal studies showed that ApoE might have a role in promoting neuron general health and survival (277). How the ApoE-4 genotype might confer vulnerability to a wide variety of neurodegenerative processes is not clear. It is hypothesized that this effect may result from impaired neuronal remyelination and axonal regeneration in ApoE-4 carriers. A possible link to oxidative stress could be due to the fact that ApoE itself has antioxidant activity, which varies depending on the isoform -E-2 has the highest antioxidant activity and E-4 the lowest (278).

Other genes, such as SMN, VEGF, and ANG, have been associated with sALS as potential disease modifiers, but the associations and underlying mechanisms remain to be established (279281). Duplications of SMN1 were shown to be a risk factor for sALS (280). Yet, to date how SMN can modify neuronal survival is not fully understood (282). Similarly, VEGF is known disease modifier for sALS, but its contribution is at best unsettled (279). Another angiogenesis factor, ANG, has just been reported as a susceptibility gene for sALS (283). ANG is a secreted ribonuclease that cleaves tRNA to initiate a stress-response program in mammalian cells; it possesses properties that are both angiogenic and cytoprotective against apoptotic inducers, such as oxidative stress, although the mechanisms have yet to be fully elucidated. Secreted ANG enters cells via receptor-mediated endocytosis and translocates to the nucleus to promote ribosomal RNA transcription and cellular proliferation (284, 285).

Dysregulation of iron homeostasis may also be involved in sALS motor neuron degeneration. As iron is a transition element, the ionic form is prone to participate in one-electron transfer reactions, making it a source of oxidative stress (286). Because iron pathway dysfunction has been identified in sALS (increased stored iron, increased levels of serum ferritin, and similar pathologies), some studies have examined the genes involved in iron metabolism, like HFE, the gene mutated in hereditary hemochromatosis. An association of the HFE alleles H63D and C282Y with ALS has emerged in several investigations [reviewed in (287)]. A recent case-control study analyzed the possible role of an intronic SNP in SLC11A2 (288), a gene that encodes an iron transporter in cerebral endosomal compartments. Comparing 579 ALS to 517 controls showed that the SLC11A2 genotype did not correlate with disease presence but that the C allele was associated with shorter disease duration in patients who also had lower limb onset (OR = 1.5, 95% CI = 1.1–2.1) (288). This finding suggests that SLC11A2 could be a genetic modifier in sALS. The metallothionein (MT) protein family is involved in pathways that detoxify heavy metals through intracellular sequestration of free toxic metals (289). A study examining a possible silencing of MT-Ia and MT-IIa promoters failed to show any modifications, however, in patients with ALS (290).

A recently discovered gene expressed in mitochondria, oxidation resistance 1 (OXR1) is induced by oxidative stress and can prevent oxidative damage to DNA in neuronal cells, although the full mechanism is not known (291). A recent study showed that Oxr1 is overexpressed in ALS spinal cord samples compared to age-matched controls. Oxr1 is susceptible to oxidation by peroxides, resulting in a loss of function that underscores how oxidative stress can be a vicious, self-sustaining process (292).

Gene mutations in fALS other than SOD1

Most gene mutations found in fALS cause mutated protein accumulation, aggregation, or both, that is likely to result in endoplasmic reticulum (ER) stress, a new area of ALS research (293). Particularly, vesicle-associated protein-associated protein B (VAPB), whose mutation causes of a rare form of fALS, is required for ER activation during ER stress and participates in intracellular vesicle transport. Other rare mutations occur in valosin-containing protein (VCP), essential for ubiquitin-dependent protein degradation (294, 295). The ER is a well-orchestrated protein-folding machine composed of protein chaperones, proteins that catalyze protein folding, and mechanisms to detect misfolded or unfolded proteins. Discrepancies between the demand and capacity of ER function lead to ER stress, leading to more misfolded proteins, which are also considered a potential cause of sALS [see in review (296)]. Although the mechanisms still remain to be clarified, persistent oxidative stress and protein misfolding may play a predominant role in the pathogenesis of these fALS cases (297). Further studies are clearly needed.

DISEASE PROGRESSION AND OXIDATIVE STRESS

The mean survival in sALS is approximately 40 months after symptom onset, but the duration of survival and functional prognosis vary widely, as approximately 10% of patients live beyond 10 years after symptom onset (298). The reasons for this variability are enigmatic. The most consistent finding is that the longer the duration between symptom onset and diagnosis, the longer the survival, a relationship that may be partly attributable to biological variability: the slower the disease progression, the longer it takes for the diagnosis to be established. Longer survival is associated with younger age of onset, pure upper neuron involvement (primary lateral sclerosis), or pure lower motor neuron involvement (progressive muscular atrophy) (298). It is possible that during the disease course, oxidative stress continues to accumulate and may hasten disease progression.

Malnutrition and cachexia

Cachexia is defined as a complex metabolic syndrome associated with underlying illness and characterized by loss of muscle with or without loss of fat mass and is associated with poor clinical outcome (299) . Classically, cachexia is associated with chronic infections and malignant conditions, such as advanced cancer, severe chronic obstructive pulmonary disease, kidney failure, and HIV infection, all of which have associated oxidative stress (300). Malnutrition and cachexia occur in sALS (301), and weight loss carries a 7.7-fold greater risk of death (302). Malnutrition is common among patients with sALS, and in some cases can be attributed to increased resting energy expenditure (303). Rats subjected to protein caloric restriction had greater oxidative stress (304). In humans, most data have come from studies performed in patients undergoing maintenance hemodialysis for kidney failure (305). Frequently, they have protein-energy malnutrition, and show elevated serum markers of oxidative stress may act synergistically with other factors, such as inflammatory processes, to involving the entire body in a vicious self-sustaining process (306).

Respiratory failure and hypoxia

Most patients with sALS develop progressive respiratory muscle weakness and die of respiratory failure. Noninvasive ventilation significantly prolongs survival and is advised when forced vital capacity (FVC) falls below 50% (307). Some patients in whom sleep disorders develop also experience intermittent hypoxia or possibly chronic hypoxia, both of which are associated with induction of oxidative stress (308).

In the CNS, O2 must cross the blood-brain barrier to enter the interstitial space and subsequently the intracellular space (309). Within the CNS, O2 distribution is heterogeneous (310). This heterogeneity is the result of differences in vascularization, tissue diffusion, and cell-specific oxygen consumption and indicates that CNS cells have a surprisingly high degree of flexibility and tolerance for variation in O2 concentration (311). Many studies in in vitro models of cerebral ischemia have shown that hypoxia induces oxidative stress in the brain (312). During hypoxia, mitochondria release superoxide anion (O2−). The mechanism for this is not well understood but probably involves respiratory chain dysfunction and a decrease in ATP production that leads to greater intracellular Ca2+, NOS activation, and thus, •NO production with a possible enhancement of RNS production (313).

Psychological stress and distress

Although major depressive illness is relatively uncommon in patients with sALS (314), the patients who do experience hopelessness, depression, and mental stress have shorter survival (315). A positive mood is associated with a 6-month longer survival time (316). In diseases other than ALS, depression and anxiety disorders, as well as perceived stress levels, are associated with increased oxidative stress, lower telomerase activity, and shorter telomere length (317). Trait anxiety and depressive symptoms in the absence of clinical disorders have been related to variations in gene expression and SNPs in healthy subjects (318). Stressor-induced alterations in proinflammatory cytokines (interleukin 1, TNF-α, interferon-γ) affect neuronal functioning through processes involving apoptosis, excitotoxicity, oxidative stress, and metabolic derangement (319). Overall, psychopathology and recent or current perceived stress may help to explain biological changes, including oxidative stress, that are associated with onset and perhaps progression of a wide range of medical conditions, including sALS. Although determining the temporal precedence of the association might be difficult, a longitudinal analysis of the temporal relationship between sALS progression and psychological stress may provide important clues to factors influencing the disease prognosis.

sALS AS A SYSTEMIC DISEASE

Although motor neurons are selectively affected in sALS, the possibility that sALS might be part of a systemic disease in which motor neurons are especially vulnerable has been postulated for some time (320, 321). A number of well-established abnormalities in sALS occur in the immune system, skin and skeletal muscle tissue, and lipid metabolism (28, 321325). Concentrations of proinflammatory cytokines, such as MCP-1 (79, 326) and IL-6 (327), are elevated in sALS CSF and serum, as is serum TNF-α (328). More recently, Zhang et al (324) found significantly numbers of CD4 T lymphocytes in sALS compared to normal controls (P < 0.05) but no differences in CD8 T-cell numbers and the CD4/CD8 ratio. The same group found significantly higher levels of plasma lipopolysaccharide and greater monocyte activation (directly related to level of lipopolysaccharide) in sALS compared to healthy individuals (P < 0.05) (323).

Abnormalities reported in lipid metabolism in patients with sALS raise highly intriguing questions concerning disease mechanisms and prognosis. In a cohort of 369 patients with ALS and 286 healthy controls, Dupuis et al (329) first reported that patients with ALS had high cholesterol levels, particularly a high LDL/HDL ratio compared to the controls. Within the ALS group, patients with a high LDL/HDL ratio (defined on the basis of the LDL/HDL ratio mean value) had a substantially longer (greater by ~12 months) median survival than those with a lower ratio. Subsequent studies regarding the role of lipid metabolism in ALS provided conflicting results. No independent association between lipid levels and survival was found in 658 Italian patients ALS as compared to 658 healthy controls, but patients with respiratory impairment had a significantly lower blood lipid concentration, particularly the LDL/HDL ratio (330). In contrast, in another study investigating 488 ALS patients, a triglyceride concentration above the median of 1.47 mmol/L was associated with a 14-month longer survival (median survival, 45 months, P = 0.006), compared to patients with a serum triglyceride level < 1.47 mmol/L (median survival 31 months). In addition, patients with hypercholesterolemia (serum cholesterol > 5.23 mmol/L) had a prolongation in life expectancy of 11 months (median survival, 42 months, P = 0.047), compared to patients with normal serum cholesterol (< 5.23 mmol/L, median survival = 31 months) (331). Two more recent studies failed to find that hyperlipidemia was significantly associated with survival in sALS patients (332, 333). Sutedja et al (334) conducted a case-control study of more than 300 ALS patients recruited at a tertiary referral clinic in the Netherlands and 500 controls recruited from participants enrolled previously in two prospective studies in the same area (335, 336). Compared to controls, patients with ALS used cholesterol-lowering agents less frequently (OR = 0.6, P =0.008), had a lower BMI (OR = 0.9, P = 0.001), a lower LDL/HDL ratio (women: OR=0.5, P <0.001; men: OR = 0.4, P <0.001), and a lower homocysteine concentration (women: OR=0.9, P = 0.02; men: OR = 0.9, P<0.001). The mean LDL and total cholesterol concentrations were significantly lower among patients with a lower percent predicted FVC. In univariate analysis, a higher LDL/HDL ratio correlated with increased survival (OR = 0.9, P = 0.04), but this association disappeared in the multivariate analysis, after adjusting for age, site at onset, and FVC. Because results vary depending on where the study was performed, geographic (such as diverse diet habits) or genetic variability might be present. Further studies are clearly needed.

Skeletal muscle mitochondria generate a large amount of oxidants and thus may affect oxidative stress in sALS (28, 321). A number of studies suggest skeletal myocyte mitochondria are abnormal in ALS (337341).

Various dermatologic abnormalities in patients with ALS have been reported, including a decrease in the amount and diameter of collagen fibrils, a marked increase in amorphous material in the ground substance, increased collagen solubility, changes in collagen cross-linking, decreased type IV collagen, increased hyaluronic acid, and increased expression of neurotropic factors such as ciliary neurotrophic factor, insulin-like growth factor, vascular endothelial growth factor, TDP-43, ubiquitin, and recently, the accumulation of valosin-containing protein, which is involved in the proteasome degradation pathway (325, 342346). Although the mechanisms of these cutaneous changes in ALS remain unknown, they all suggest systemic abnormalities are present outside of the CNS. Skin fibroblasts derived from sALS patients were more sensitive to oxidative stress induced by H2O2 and 3-morpholinosydnonimine hydrochloride than were fibroblasts from patients with fALS and the SOD1 mutation (347). We clearly need further studies to determine whether other cells, in addition to motor neurons, can be useful in studying the sALS disease process.

Other evidence suggesting that sALS is a systemic disease include ultrastructural abnormalities in hepatocyte mitochondria (348) and reduced GSH and GSH peroxidase activity in erythrocytes (89). Although oxidative stress biomarkers are most likely generated in the CNS because of its high oxygen consumption, some are undoubtedly produced outside of the CNS (42, 349). Particularly, lipid peroxidase products such as acrolein, HNE, and the IsoPs are highly biologically active, causing further cellular damage via apoptosis or nucleophilic action (58). These neurotoxic lipid peroxidase products can cause cellular damage far from their site of production because they can diffuse throughout the cell. In primary motor neuron cultures, ApoE protein isoforms differentially detoxified HNE, with ApoE-2 detoxifying more HNE than ApoE-4 (350). This suggests that lipid metabolism may be strongly associated with pro- and antioxidative processes. Following the oxidative stress profile throughout the disease course could give us much information about the adaptive changes of the individual and their ability to respond to oxidative stress. Oxidative stress biomarkers are not just an end product of oxidative stress but are bioactive and potentially neurotoxic themselves. Further studies are needed in this area.

WHAT IS THE NEXT STEP?

Although the evidence for oxidative damage in sALS pathogenesis is extensive, the ultimate trigger(s) that causes increased ROS levels is still unknown, leading to speculation as to whether oxidative stress is a primary cause of disease or merely a secondary consequence. Furthermore, it is always possible that there may be unrecognized alternative mechanisms that can, in part, explain the pathogenic changes described above. Prospective studies have shown that smoking, a cause of oxidative stress, is associated with sALS, whereas the evidence for other factors is, at best, equivocal. Smoking is relatively common in the general population, but other factors, such as agricultural or heavy metal exposure, are less common; thus, statistically significant relationships are more difficult to find. The results may depend heavily on the populations and locations studied. Many epidemiological studies have failed to demonstrate strong relationships, in part due to methodological shortcomings (104, 351, 352). Yet, the reality also is that it is difficult to find strong associations between rare events and a rare multifactorial disease such as sALS. Although a prospective epidemiological study to search for a positive causative factor in sALS is desirable, such a study would involve following a large population for an extended period of time. Such studies are expensive and not feasible.

We may be able to take an alternative approach to identifying common factors in patients that cause oxidative stress. Low levels of oxidative stress induced by various environmental or lifestyle factors may converge to produce a cumulative, measurable degree of oxidative stress, as discussed above. If sALS is indeed a multifactorial disease, oxidative stress may be an underlying biological basis for multiple, apparently diverse unrelated stressors. Oxidative stress can further accumulate throughout the disease course because of malnutrition, respiratory failure, and psychological stress. Moreover, if sALS is truly a systemic disease, oxidative stress not only affects the patient systemically but also affects motoneuron integrity. Lipid peroxidase products, such as HNE or IsoPs, which are highly neurotoxic, may be generated centrally and peripherally due to oxidative stress. However, they can affect the vulnerable motor neuron in the CNS because those lipid peroxidase products can damage the cerebral microvascular endothelial cells of the blood brain barrier, leading to their breakdown (57). Almost certainly, the real picture is far more complex than what we describe here, because individual genetic susceptibility and epigenetic mechanisms, which further modify gene-environment interactions, are likely to influence disease onset, disease progression, and even disease phenotypes. Furthermore, such studies are in their infancy. Nevertheless, we believe that a molecular epidemiological study in sALS, focusing on one biological process – namely, oxidative stress – may be an alternative and perhaps a more effective approach to identify potential factors in sALS.

Answering these simple but fundamental questions requires well-planned, well-designed epidemiological studies that recruit patients from multiple centers to achieve adequate sample sizes. Moreover, a multidisciplinary approach is essential, because both molecular and environmental risk factors are involved. Clinical investigators who define the disease, epidemiologists from different disciplines (general, nutritional, occupational, genetic epidemiology), laboratory scientists who are experts in oxidative stress biology, molecular geneticists, experts in epigenetics, clinical psychologists, and biostatisticians must participate in such studies, because their combined expertise is essential to uncover the etiology of a complex disease like sALS. In addition, the appropriate collection and processing of biospecimens is critical for success.

We believe that this approach will provide solid biological explanations for disease onset, phenotypic changes, and disease severity in sALS. If it is determined that oxidative stress is a key factor in the pathomechanism(s) of sALS, we will need to devise innovative clinical trials to successfully investigate oxidative stress and develop more effective treatments and prevention strategies. Although several past clinical trials that were intended to counteract presumed oxidative stress failed to demonstrate any clinical benefits in ALS (353357), these failures do not mean that the hypothesis of oxidative stress is wrong. These medications with anti-oxidative properties have limited ability to cross the blood brain barrier, and further, oxidative stress may be involved in different biological steps of cellular metabolism. With a more thorough understanding of these mechanisms of cellular oxidative stress, the treatment and management of sALS will be biologically based, and proactive and appropriate treatments can be developed that slow or even stop progression of this devastating neurodegenerative disease.

Highlights.

  • -

    Oxidative stress is a key pathogenic mechanism of motoneuronal degeneration in ALS.

  • -

    Oxidative stress biomarkers are increased in CSF, plasma and urine in ALS.

  • -

    Many environmental risk factors in ALS can cause systemic oxidative stress.

  • -

    Malnutrition and respiratory and psychological stress may cause oxidative stress.

  • -

    Increased systemic oxidative stress may quicken ALS disease progression.

Acknowledgement

Georgia Christodoulou, M.A., helped with the manuscript preparation and Cassandra Talerico, Ph.D., provided substantive editing.

Study Funding: We acknowledge grant support from NIEHS (1R01 ES016348 to Hiroshi Mitsumoto and P30 ES009089 to Regina Santella); also to HM, Muscular Dystrophy Association (number 4350), MDA Wings Over Wall Street, and The Judith and Jean Pape Adams Charitable Foundation, along with donations from the Spina Family, David Marren, the Senerchia family, the Drago family, and others to HM.

Glossary

Abbreviations

ALAD

δ-aminolevulinic acid dehydratase

ALS

Amyotrophic Lateral Sclerosis

fALS

familial Amyotrophic Lateral Sclerosis

sALS

sporadic Amyotrophic Lateral Sclerosis

ANG

angiogenin

ApoE

apolipoprotein E

CNS

central nervous system

CSF

cerebrospinal fluid

CTE

chronic traumatic encephalopathy

ER

endoplasmic reticulum

FVC

forced vital capacity

IsoPs

F2-isoprostanes

FTD

frontotemporal dementia

GSH

glutathione

HDL

high-density lipoprotein

HNE

4-hydroxy-2-nonenal

IsoPs

isoprostanes

LDL

low-density lipoprotein

MDA

malondialdehyde

MND

motor neuron disease

MT

metallothionein

NADPH

nicotinamide adenine dinucleotide phosphate

NOS

nitric oxide synthetase

3-NT

3-nitrotyrosine

8-oxoG

8-oxo-deoxyguanosine

OXR1

oxidation resistance 1

PD

Parkinson disease

PON

paraoxonase 1

RNS

reactive nitrogen species

ROS

reactive oxygen species

sALS

sporadic ALS

SMN

survival motor neuron

SNP

single nucleotide polymorphism

SOD1

superoxide dismutase 1

TBARS

thiobarbituric acid-reactive substances

TDP-43

TAR DNA-binding protein

VAPB

vesicle associated protein-associated protein B

VCP

valosin-containing protein

VEGF

vascular endothelial growth factor

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures (past 3 years): Hiroshi Mitsumoto, M.D. has received grant support as stated above.

Pam Factor-Litvak, Ph.D. has received grant support from NIEHS (1R01 ES016348, P30 ES009089, 1R01 ES017024, 1R01ES0122315, P42 ES010349).

Regina M. Santella, Ph.D. has received grant support as stated above, as well as R01 ES005116 and R03CA159427.

Emanuele D’Amico, M.D. has no conflict of interest.

The study sponsors had no involvement in the review development, collection, analysis, or interpretation of data, writing of the manuscript, or the decision to submit for publication.

Contributor Information

Emanuele D’Amico, Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, 710 West 168th Street (NI-9), New York, NY 10032, ed2550@columbia.edu; damico.ema@libero.it

Pam Factor-Litvak, Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, prf1@columbia.edu

Regina M. Santella, Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, rps1@columbia.edu

Hiroshi Mitsumoto, Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, 710 West 168th Street (NI-9), New York, NY 10032

References

  • 1.Miller RG, Jackson CE, Kasarskis EJ, England JD, Forshew D, Johnston W, Kalra S, Katz JS, Mitsumoto H, Rosenfeld J, Shoesmith C, Strong MJ, Woolley SC. Quality Standards Subcommittee of the American Academy of N Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: multidisciplinary care, symptom management, and cognitive/behavioral impairment (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2009;73(15):1227–1233. doi: 10.1212/WNL.0b013e3181bc01a4. Epub 2009/10/14; PubMed Central PMCID: PMC2764728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ludolph AC, Brettschneider J, Weishaupt JH. Amyotrophic lateral sclerosis. Current Opinion in Neurology. 2012;25(5):530–535. doi: 10.1097/WCO.0b013e328356d328. [DOI] [PubMed] [Google Scholar]
  • 3.Mitsumoto H, Rabkin JG. Palliative care for patients with amyotrophic lateral sclerosis -"Prepare for the worst and hope for the best". Jama-J Am Med Assoc. 2007;298(2):207–216. doi: 10.1001/jama.298.2.207. [DOI] [PubMed] [Google Scholar]
  • 4.Siddique T, Ajroud-Driss S. Familial amyotrophic lateral sclerosis, a historical perspective. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology / edited by the Gaetano Conte Academy for the study of striated muscle diseases. 2011;30(2):117–120. Epub 2011/11/24; PubMed Central PMCID: PMC3235825. [PMC free article] [PubMed] [Google Scholar]
  • 5.Williams KL, Solski JA, Nicholson GA, Blair IP. Mutation analysis of VCP in familial and sporadic amyotrophic lateral sclerosis. Neurobiol Aging. 2011 doi: 10.1016/j.neurobiolaging.2011.11.022. Epub 2011/12/27. [DOI] [PubMed] [Google Scholar]
  • 6.Andersen PM, Al-Chalabi A. Clinical genetics of amyotrophic lateral sclerosis: what do we really know? Nature Reviews Neurology. 2011;7(11):603–615. doi: 10.1038/nrneurol.2011.150. [DOI] [PubMed] [Google Scholar]
  • 7.Maruyama H, Morino H, Ito H, Izumi Y, Kato H, Watanabe Y, Kinoshita Y, Kamada M, Nodera H, Suzuki H, Komure O, Matsuura S, Kobatake K, Morimoto N, Abe K, Suzuki N, Aoki M, Kawata A, Hirai T, Kato T, Ogasawara K, Hirano A, Takumi T, Kusaka H, Hagiwara K, Kaji R, Kawakami H. Mutations of optineurin in amyotrophic lateral sclerosis. Nature. 2010;465(7295):223–236. doi: 10.1038/nature08971. Epub 2010/04/30. [DOI] [PubMed] [Google Scholar]
  • 8.DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, Kouri N, Wojtas A, Sengdy P, Hsiung GY, Karydas A, Seeley WW, Josephs KA, Coppola G, Geschwind DH, Wszolek ZK, Feldman H, Knopman DS, Petersen RC, Miller BL, Dickson DW, Boylan KB, Graff-Radford NR, Rademakers R. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72(2):245–256. doi: 10.1016/j.neuron.2011.09.011. Epub 2011/09/29; PubMed Central PMCID: PMC3202986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, Kalimo H, Paetau A, Abramzon Y, Remes AM, Kaganovich A, Scholz SW, Duckworth J, Ding JH, Harmer DW, Hernandez DG, Johnson JO, Mok K, Ryten M, Trabzuni D, Guerreiro RJ, Orrell RW, Neal J, Murray A, Pearson J, Jansen IE, Sondervan D, Seelaar H, Blake D, Young K, Halliwell N, Callister JB, Toulson G, Richardson A, Gerhard A, Snowden J, Mann D, Neary D, Nalls MA, Peuralinna T, Jansson L, Isoviita VM, Kaivorinne AL, Holtta-Vuori M, Ikonen E, Sulkava R, Benatar M, Wuu J, Chio A, Restagno G, Borghero G, Sabatelli M, Heckerman D, Rogaeva E, Zinman L, Rothstein JD, Sendtner M, Drepper C, Eichler EE, Alkan C, Abdullaev Z, Pack SD, Dutra A, Pak E, Hardy J, Singleton A, Williams NM, Heutink P, Pickering-Brown S, Morris HR, Tienari PJ, Traynor BJ, Consortium I. A Hexanucleotide Repeat Expansion in C9ORF72 Is the Cause of Chromosome 9p21-Linked ALS-FTD. Neuron. 2011;72(2):257–268. doi: 10.1016/j.neuron.2011.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rothstein JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol. 2009;65(Suppl 1):S3–S9. doi: 10.1002/ana.21543. Epub 2009/02/05. [DOI] [PubMed] [Google Scholar]
  • 11.Van Damme P, Robberecht W. Recent advances in motor neuron disease. Curr Opin Neurol. 2009;22(5):486–492. doi: 10.1097/WCO.0b013e32832ffbe3. Epub 2009/07/14. [DOI] [PubMed] [Google Scholar]
  • 12.RL . The cause of amyotrophic lateral sclerosis. In: Mitsumoto H, GP PS, editors. Amytrophic Lateral Sclerosis. New York: Taylor and Francis; 2009. pp. 81–98. [Google Scholar]
  • 13.Ischiropoulos H, Beckman JS. Oxidative stress and nitration in neurodegeneration: cause, effect, or association? J Clin Invest. 2003;111(2):163–169. doi: 10.1172/JCI17638. Epub 2003/01/18; PubMed Central PMCID: PMC151889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Circu ML, Aw TY. Reactive oxygen species, cellular redox systems, and apoptosis. Free Radical Bio Med. 2010;48(6):749–762. doi: 10.1016/j.freeradbiomed.2009.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sies H. Oxidative stress: Oxidants and antioxidants. Journal of Physiology-London. 1996;491P:S2–S2. doi: 10.1113/expphysiol.1997.sp004024. [DOI] [PubMed] [Google Scholar]
  • 16.Sies H. Oxidative Stress - from Basic Research to Clinical-Application. American Journal of Medicine. 1991;91:S31–S38. doi: 10.1016/0002-9343(91)90281-2. [DOI] [PubMed] [Google Scholar]
  • 17.Cosgrove JP, Church DF, Pryor WA. The kinetics of the autoxidation of polyunsaturated fatty acids. Lipids. 1987;22(5):299–304. doi: 10.1007/BF02533996. Epub 1987/05/01. [DOI] [PubMed] [Google Scholar]
  • 18.Contestabile A. Oxidative stress in neurodegeneration: mechanisms and therapeutic perspectives. Curr Top Med Chem. 2001;1(6):553–568. doi: 10.2174/1568026013394723. Epub 2002/03/16. [DOI] [PubMed] [Google Scholar]
  • 19.Barber SC, Shaw PJ. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic Biol Med. 2010;48(5):629–641. doi: 10.1016/j.freeradbiomed.2009.11.018. Epub 2009/12/09. [DOI] [PubMed] [Google Scholar]
  • 20.Agar J, Durham H. Relevance of oxidative injury in the pathogenesis of motor neuron diseases. Amyotroph Lateral Scler Other Motor Neuron Disord. 2003;4(4):232–242. doi: 10.1080/14660820310011278. Epub 2004/02/03. [DOI] [PubMed] [Google Scholar]
  • 21.Calingasan NY, Chen J, Kiaei M, Beal MF. Beta-amyloid 42 accumulation in the lumbar spinal cord motor neurons of amyotrophic lateral sclerosis patients. Neurobiol Dis. 2005;19(1–2):340–347. doi: 10.1016/j.nbd.2005.01.012. Epub 2005/04/20. [DOI] [PubMed] [Google Scholar]
  • 22.Henkel JS, Engelhardt JI, Siklos L, Simpson EP, Kim SH, Pan T, Goodman JC, Siddique T, Beers DR, Appel SH. Presence of dendritic cells, MCP-1, and activated microglia/macrophages in amyotrophic lateral sclerosis spinal cord tissue. Ann Neurol. 2004;55(2):221–235. doi: 10.1002/ana.10805. Epub 2004/02/03. [DOI] [PubMed] [Google Scholar]
  • 23.Kato S, Kato M, Abe Y, Matsumura T, Nishino T, Aoki M, Itoyama Y, Asayama K, Awaya A, Hirano A, Ohama E. Redox system expression in the motor neurons in amyotrophic lateral sclerosis (ALS): immunohistochemical studies on sporadic ALS, superoxide dismutase 1 (SOD1)-mutated familial ALS, and SOD1-mutated ALS animal models. Acta Neuropathol. 2005;110(2):101–112. doi: 10.1007/s00401-005-1019-3. Epub 2005/06/29. [DOI] [PubMed] [Google Scholar]
  • 24.Kim SH, Henkel JS, Beers DR, Sengun IS, Simpson EP, Goodman JC, Engelhardt JI, Siklos L, Appel SH. PARP expression is increased in astrocytes but decreased in motor neurons in the spinal cord of sporadic ALS patients. J Neuropath Exp Neur. 2003;62(1):88–103. doi: 10.1093/jnen/62.1.88. [DOI] [PubMed] [Google Scholar]
  • 25.Beckman JS, Estevez AG. Superoxide Dismutase Oxidative Stress and ALS. Am. 2004 In: h, editor. [Google Scholar]
  • 26.Shaw PJ. Molecular and cellular pathways of neurodegeneration in motor neurone disease. J Neurol Neurosurg Psychiatry. 2005;76(8):1046–1057. doi: 10.1136/jnnp.2004.048652. Epub 2005/07/19; PubMed Central PMCID: PMC1739758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Carri MT, Ferri A, Cozzolino M, Calabrese L, Rotilio G. Neurodegeneration in amyotrophic lateral sclerosis: the role of oxidative stress and altered homeostasis of metals. Brain Res Bull. 2003;61(4):365–374. doi: 10.1016/s0361-9230(03)00179-5. Epub 2003/08/12. [DOI] [PubMed] [Google Scholar]
  • 28.Dupuis L. Oxidative stress sensitivity in ALS muscle cells. Experimental Neurology. 2009;220(2):219–223. doi: 10.1016/j.expneurol.2009.08.023. [DOI] [PubMed] [Google Scholar]
  • 29.Barber SC, Mead RJ, Shaw PJ. Oxidative stress in ALS: a mechanism of neurodegeneration and a therapeutic target. Biochim Biophys Acta. 2006;1762(11–12):1051–1067. doi: 10.1016/j.bbadis.2006.03.008. Epub 2006/05/23. [DOI] [PubMed] [Google Scholar]
  • 30.Robberecht W. Oxidative stress in amyotrophic lateral sclerosis. Journal of Neurology. 2000;247:1–6. doi: 10.1007/s004150050551. [DOI] [PubMed] [Google Scholar]
  • 31.Simpson EP, Yen AA, Appel SH. Oxidative Stress: a common denominator in the pathogenesis of amyotrophic lateral sclerosis. Current Opinion in Rheumatology. 2003;15(6):730–736. doi: 10.1097/00002281-200311000-00008. [DOI] [PubMed] [Google Scholar]
  • 32.Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nature Neuroscience. 2008;11(3):251–253. doi: 10.1038/nn2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nagai M, Re DB, Nagata T, Chalazonitis A, Jessell TM, Wichterle H, Przedborski S. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat Neurosci. 2007;10(5):615–622. doi: 10.1038/nn1876. Epub 2007/04/17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Clement AM, Nguyen MD, Roberts EA, Garcia ML, Boillee S, Rule M, McMahon AP, Doucette W, Siwek D, Ferrante RJ, Brown RH, Jr, Julien JP, Goldstein LS, Cleveland DW. Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science. 2003;302(5642):113–117. doi: 10.1126/science.1086071. Epub 2003/10/04. [DOI] [PubMed] [Google Scholar]
  • 35.Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, Song S, Likhite S, Murtha MJ, Foust KD, Rao M, Eagle A, Kammesheidt A, Christensen A, Mendell JR, Burghes AH, Kaspar BK. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat Biotechnol. 2011;29(9):824–828. doi: 10.1038/nbt.1957. Epub 2011/08/13; PubMed Central PMCID: PMC3170425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, Song SW, Likhite S, Murtha MJ, Foust KD, Rao MH, Eagle A, Kammesheidt A, Christensen A, Mendell JR, Burghes AHM, Kaspar BK. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nature Biotechnology. 2011;29(9):824–U79. doi: 10.1038/nbt.1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Trotti D, Danbolt NC, Volterra A. Glutamate transporters are oxidant-vulnerable: a molecular link between oxidative and excitotoxic neurodegeneration? Trends in Pharmacological Sciences. 1998;19(8):328–334. doi: 10.1016/s0165-6147(98)01230-9. [DOI] [PubMed] [Google Scholar]
  • 38.Pehar M, Cassina P, Vargas MR, Castellanos R, Viera L, Beckman JS, Estevez AG, Barbeito L. Astrocytic production of nerve growth factor in motor neuron apoptosis: implications for amyotrophic lateral sclerosis. Journal of neurochemistry. 2004;89(2):464–473. doi: 10.1111/j.1471-4159.2004.02357.x. [DOI] [PubMed] [Google Scholar]
  • 39.Pellerin L, Pellegri G, Bittar PG, Charnay Y, Bouras C, Martin JL, Stella N, Magistretti PJ. Evidence supporting the existence of an activity-dependent astrocyte-neuron lactate shuttle. Dev Neurosci-Basel. 1998;20(4–5):291–299. doi: 10.1159/000017324. [DOI] [PubMed] [Google Scholar]
  • 40.Liddell JR, Zwingmann C, Schmidt MM, Thiessen A, Leibfritz D, Robinson SR, Dringen R. Sustained Hydrogen Peroxide Stress Decreases Lactate Production by Cultured Astrocytes. Journal of neuroscience research. 2009;87(12):2696–2708. doi: 10.1002/jnr.22093. [DOI] [PubMed] [Google Scholar]
  • 41.Thorburne SK, Juurlink BHJ. Low glutathione and high iron govern the susceptibility of oligodendroglial precursors to oxidative stress. Journal of neurochemistry. 1996;67(3):1014–1022. doi: 10.1046/j.1471-4159.1996.67031014.x. [DOI] [PubMed] [Google Scholar]
  • 42.Bogdanov M, Brown RH, Matson W, Smart R, Hayden D, O'Donnell H, Flint Beal M, Cudkowicz M. Increased oxidative damage to DNA in ALS patients. Free Radic Biol Med. 2000;29(7):652–628. doi: 10.1016/s0891-5849(00)00349-x. Epub 2000/10/18. [DOI] [PubMed] [Google Scholar]
  • 43.Mitsumoto H, Santella RM, Liu X, Bogdanov M, Zipprich J, Wu HC, Mahata J, Kilty M, Bednarz K, Bell D, Gordon PH, Hornig M, Mehrazin M, Naini A, Flint Beal M, Factor-Litvak P. Oxidative stress biomarkers in sporadic ALS. Amyotroph Lateral Scler. 2008;9(3):177–183. doi: 10.1080/17482960801933942. Epub 2008/06/25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Klaunig JE, Xu Y, Isenberg JS, Bachowski S, Kolaja KL, Jiang J, Stevenson DE, Walborg EF., Jr The role of oxidative stress in chemical carcinogenesis. Environ Health Perspect. 1998;106(Suppl 1):289–295. doi: 10.1289/ehp.98106s1289. Epub 1998/03/31; PubMed Central PMCID: PMC1533298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Halliwell B. Role of free radicals in the neurodegenerative diseases: therapeutic implications for antioxidant treatment. Drugs Aging. 2001;18(9):685–716. doi: 10.2165/00002512-200118090-00004. Epub 2001/10/16. [DOI] [PubMed] [Google Scholar]
  • 46.Blumberg J. Use of biomarkers of oxidative stress in research studies. Journal of Nutrition. 2004;134(11):3188s–3189s. doi: 10.1093/jn/134.11.3188S. [DOI] [PubMed] [Google Scholar]
  • 47.Halliwell B, Whiteman M. Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol. 2004;142(2):231–255. doi: 10.1038/sj.bjp.0705776. Epub 2004/05/25; PubMed Central PMCID: PMC1574951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lee YA, Yun BH, Kim SK, Margolin Y, Dedon PC, Geacintov NE, Shafirovich V. Mechanisms of oxidation of guanine in DNA by carbonate radical anion, a decomposition product of nitrosoperoxycarbonate. Chem-Eur J. 2007;13(16):4571–4581. doi: 10.1002/chem.200601434. [DOI] [PubMed] [Google Scholar]
  • 49.Steenken S, Jovanovic SV. How easily oxidizable is DNA? One-electron reduction potentials of adenosine and guanosine radicals in aqueous solution. Journal of the American Chemical Society. 1997;119(3):617–618. [Google Scholar]
  • 50.Radak Z, Boldogh I. 8-Oxo-7,8-dihydroguanine: Links to gene expression, aging, and defense against oxidative stress. Free Radical Bio Med. 2010;49(4):587–596. doi: 10.1016/j.freeradbiomed.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Radak Z, Zhao Z, Goto S, Koltai E. Age-associated neurodegeneration and oxidative damage to lipids, proteins and DNA. Mol Aspects Med. 2011;32(4–6):305–315. doi: 10.1016/j.mam.2011.10.010. Epub 2011/10/25. [DOI] [PubMed] [Google Scholar]
  • 52.Catala A. An overview of lipid peroxidation with emphasis in outer segments of photoreceptors and the chemiluminescence assay. Int J Biochem Cell Biol. 2006;38(9):1482–1495. doi: 10.1016/j.biocel.2006.02.010. Epub 2006/04/20. [DOI] [PubMed] [Google Scholar]
  • 53.Carini M, Aldini G, Facino RM. Mass spectrometry for detection of 4-hydroxy-trans-2-nonenal (HNE) adducts with peptides and proteins. Mass Spectrom Rev. 2004;23(4):281–305. doi: 10.1002/mas.10076. Epub 2004/05/11. [DOI] [PubMed] [Google Scholar]
  • 54.Uchida K. Histidine and lysine as targets of oxidative modification. Amino Acids. 2003;25(3–4):249–257. doi: 10.1007/s00726-003-0015-y. [DOI] [PubMed] [Google Scholar]
  • 55.Wilson R, Lyall K, Smyth L, Fernie CE, Riemersma RA. Dietary hydroxy fatty acids are absorbed in humans: implications for the measurement of 'oxidative stress' in vivo. Free Radic Biol Med. 2002;32(2):162–168. doi: 10.1016/s0891-5849(01)00780-8. Epub 2002/02/14. [DOI] [PubMed] [Google Scholar]
  • 56.Farooqui AA, Horrocks LA. Phospholipase A2-generated lipid mediators in the brain: the good, the bad, and the ugly. Neuroscientist. 2006;12(3):245–260. doi: 10.1177/1073858405285923. Epub 2006/05/11. [DOI] [PubMed] [Google Scholar]
  • 57.Eckl PM. Genotoxicity of HNE. Mol Aspects Med. 2003;24(4–5):161–165. doi: 10.1016/s0098-2997(03)00010-4. Epub 2003/08/02. [DOI] [PubMed] [Google Scholar]
  • 58.LoPachin RM, Gavin T, Petersen DR, Barber DS. Molecular mechanisms of 4-hydroxy-2-nonenal and acrolein toxicity: nucleophilic targets and adduct formation. Chem Res Toxicol. 2009;22(9):1499–1508. doi: 10.1021/tx900147g. Epub 2009/07/21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Milatovic D, Montine TJ, Aschner M. Measurement of isoprostanes as markers of oxidative stress. Methods Mol Biol. 2011;758:195–204. doi: 10.1007/978-1-61779-170-3_13. Epub 2011/08/05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Roberts LJ, 2nd, Morrow JD. Products of the isoprostane pathway: unique bioactive compounds and markers of lipid peroxidation. Cell Mol Life Sci. 2002;59(5):808–820. doi: 10.1007/s00018-002-8469-8. Epub 2002/06/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Basu S. Isoprostanes: novel bioactive products of lipid peroxidation. Free Radic Res. 2004;38(2):105–122. doi: 10.1080/10715760310001646895. Epub 2004/04/24. [DOI] [PubMed] [Google Scholar]
  • 62.Basu S, Helmersson J. Factors regulating isoprostane formation in vivo. Antioxid Redox Signal. 2005;7(1–2):221–235. doi: 10.1089/ars.2005.7.221. Epub 2005/01/15. [DOI] [PubMed] [Google Scholar]
  • 63.Montuschi P, Barnes PJ, Roberts LJ., 2nd Isoprostanes: markers and mediators of oxidative stress. FASEB J. 2004;18(15):1791–1800. doi: 10.1096/fj.04-2330rev. Epub 2004/12/04. [DOI] [PubMed] [Google Scholar]
  • 64.Morrow JD, Awad JA, Boss HJ, Blair IA, Roberts LJ. Non-Cyclooxygenase-Derived Prostanoids (F2-Isoprostanes) Are Formed Insitu on Phospholipids. P Natl Acad Sci USA. 1992;89(22):10721–10725. doi: 10.1073/pnas.89.22.10721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zeiger SLH, Musiek ES, Zanoni G, Vidari G, Morrow JD, Milne GJ, McLaughlin B. Neurotoxic lipid peroxidation species formed by ischemic stroke increase injury. Free Radical Bio Med. 2009;47(10):1422–1431. doi: 10.1016/j.freeradbiomed.2009.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Musiek ES, Milne GL, McLaughlin B, Morrow JD. Cyclopentenone eicosanoids as mediators of neurodegeneration: A pathogenic mechanism of oxidative stress-mediated and cyclooxygenase-mediated neurotoxicity. Brain Pathology. 2005;15(2):149–158. doi: 10.1111/j.1750-3639.2005.tb00512.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Berlett BS, Stadtman ER. Protein oxidation in aging, disease, and oxidative stress. J Biol Chem. 1997;272(33):20313–20316. doi: 10.1074/jbc.272.33.20313. Epub 1997/08/15. [DOI] [PubMed] [Google Scholar]
  • 68.Bruno M, Moore T, Nesnow S, Ge Y. Protein carbonyl formation in response to propiconazole-induced oxidative stress. J Proteome Res. 2009;8(4):2070–2078. doi: 10.1021/pr801061r. Epub 2009/08/29. [DOI] [PubMed] [Google Scholar]
  • 69.Pandey KB, Mehdi MM, Maurya PK, Rizvi SI. Plasma protein oxidation and its correlation with antioxidant potential during human aging. Dis Markers. 2010;29(1):31–36. doi: 10.3233/DMA-2010-0723. Epub 2010/09/10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Souza JM, Peluffo G, Radi R. Protein tyrosine nitration--functional alteration or just a biomarker? Free Radic Biol Med. 2008;45(4):357–366. doi: 10.1016/j.freeradbiomed.2008.04.010. Epub 2008/05/08. [DOI] [PubMed] [Google Scholar]
  • 71.Abe K, Pan LH, Watanabe M, Konno H, Kato T, Itoyama Y. Upregulation of protein-tyrosine nitration in the anterior horn cells of amyotrophic lateral sclerosis. Neurol Res. 1997;19(2):124–128. doi: 10.1080/01616412.1997.11740784. Epub 1997/04/01. [DOI] [PubMed] [Google Scholar]
  • 72.Casoni F, Basso M, Massignan T, Gianazza E, Cheroni C, Salmona M, Bendotti C, Bonetto V. Protein nitration in a mouse model of familial amyotrophic lateral sclerosis: possible multifunctional role in the pathogenesis. J Biol Chem. 2005;280(16):16295–16304. doi: 10.1074/jbc.M413111200. Epub 2005/02/09. [DOI] [PubMed] [Google Scholar]
  • 73.Bruijn LI, Beal MF, Becher MW, Schulz JB, Wong PC, Price DL, Cleveland DW. Elevated free nitrotyrosine levels, but not protein-bound nitrotyrosine or hydroxyl radicals, throughout amyotrophic lateral sclerosis (ALS)-like disease implicate tyrosine nitration as an aberrant in vivo property of one familial ALS-linked superoxide dismutase 1 mutant. P Natl Acad Sci USA. 1997;94(14):7606–7611. doi: 10.1073/pnas.94.14.7606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yoshino H, Kimura A. Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study) Amyotrophic lateral sclerosis : official publication of the World Federation of Neurology Research Group on Motor Neuron Diseases. 2006;7(4):241–245. doi: 10.1080/17482960600881870. Epub 2006/11/28. [DOI] [PubMed] [Google Scholar]
  • 75.Nardo G, Pozzi S, Pignataro M, Lauranzano E, Spano G, Garbelli S, Mantovani S, Marinou K, Papetti L, Monteforte M, Torri V, Paris L, Bazzoni G, Lunetta C, Corbo M, Mora G, Bendotti C, Bonetto V. Amyotrophic Lateral Sclerosis Multiprotein Biomarkers in Peripheral Blood Mononuclear Cells. PLoS One. 2011;6(10) doi: 10.1371/journal.pone.0025545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Beckman JS, Crow JP. Pathological implications of nitric oxide, superoxide and peroxynitrite formation. Biochem Soc Trans. 1993;21(2):330–334. doi: 10.1042/bst0210330. Epub 1993/05/01. [DOI] [PubMed] [Google Scholar]
  • 77.Beckman JS, Carson M, Smith CD, Koppenol WH. ALS, SOD and peroxynitrite. Nature. 1993;364(6438):584. doi: 10.1038/364584a0. Epub 1993/08/12. [DOI] [PubMed] [Google Scholar]
  • 78.Peluffo H, Shacka JJ, Ricart K, Bisig CG, Martinez-Palma L, Pritsch O, Kamaid A, Eiserich JP, Crow JP, Barbeito L, Estevez AG. Induction of motor neuron apoptosis by free 3-nitro-L-tyrosine. J Neurochem. 2004;89(3):602–612. doi: 10.1046/j.1471-4159.2004.02363.x. Epub 2004/04/17. [DOI] [PubMed] [Google Scholar]
  • 79.Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH. Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology. 2004;62(10):1758–1765. doi: 10.1212/wnl.62.10.1758. Epub 2004/05/26. [DOI] [PubMed] [Google Scholar]
  • 80.Montine TJ, Beal MF, Cudkowicz ME, O'Donnell H, Margolin RA, McFarland L, Bachrach AF, Zackert WE, Roberts LJ, Morrow JD. Increased CSF F2-isoprostane concentration in probable AD. Neurology. 1999;52(3):562–565. doi: 10.1212/wnl.52.3.562. Epub 1999/02/20. [DOI] [PubMed] [Google Scholar]
  • 81.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. Annals of Neurology. 1999;46(1):129–131. doi: 10.1002/1531-8249(199907)46:1<129::aid-ana21>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
  • 82.Tohgi H, Abe T, Yamazaki K, Murata T, Ishizaki E, Isobe C. Increase in oxidized NO products and reduction in oxidized glutathione in cerebrospinal fluid from patients with sporadic form of amyotrophic lateral sclerosis. Neuroscience Letters. 1999;260(3):204–206. doi: 10.1016/s0304-3940(98)00986-0. [DOI] [PubMed] [Google Scholar]
  • 83.Bonnefont-Rousselot D, Lacomblez L, Jaudon M, Lepage S, Salachas F, Bensimon G, Bizard C, Doppler V, Delattre J, Meininger V. Blood oxidative stress in amyotrophic lateral sclerosis. J Neurol Sci. 2000;178(1):57–62. doi: 10.1016/s0022-510x(00)00365-8. Epub 2000/10/06. [DOI] [PubMed] [Google Scholar]
  • 84.Ryberg H, Soderling AS, Davidsson P, Blennow K, Caidahl K, Persson LI. Cerebrospinal fluid levels of free 3-nitrotyrosine are not elevated in the majority of patients with amyotrophic lateral sclerosis or Alzheimer's disease. Neurochemistry International. 2004;45(1):57–62. doi: 10.1016/j.neuint.2003.12.012. [DOI] [PubMed] [Google Scholar]
  • 85.Sohmiya M, Tanaka M, Suzuki Y, Tanino Y, Okamoto K, Yamamoto Y. An increase of oxidized coenzyme Q-10 occurs in the plasma of sporadic ALS patients. Journal of the Neurological Sciences. 2005;228(1):49–53. doi: 10.1016/j.jns.2004.09.030. [DOI] [PubMed] [Google Scholar]
  • 86.Siciliano G, Piazza S, Carlesi C, Del Corona A, Franzini M, Pompella A, Malvaldi G, Mancuso M, Paolicchi A, Murri L. Antioxidant capacity and protein oxidation in cerebrospinal fluid of amyotrophic lateral sclerosis. Journal of Neurology. 2007;254(5):575–580. doi: 10.1007/s00415-006-0301-1. [DOI] [PubMed] [Google Scholar]
  • 87.Murata T, Ohtsuka C, Terayama Y. Increased mitochondrial oxidative damage in patients with sporadic amyotrophic lateral sclerosis. Journal of the Neurological Sciences. 2008;267(1–2):66–69. doi: 10.1016/j.jns.2007.09.038. [DOI] [PubMed] [Google Scholar]
  • 88.Keizman D, Ish-Shalom M, Berliner S, Maimon N, Vered Y, Artamonov I, Tsehori J, Nefussy B, Drory VE. Low uric acid levels in serum of patients with ALS: Further evidence for oxidative stress? Journal of the Neurological Sciences. 2009;285(1–2):95–99. doi: 10.1016/j.jns.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 89.Cova E, Bongioanni P, Cereda C, Metelli MR, Salvaneschi L, Bernuzzi S, Guareschi S, Rossi B, Ceroni M. Time course of oxidant markers and antioxidant defenses in subgroups of amyotrophic lateral sclerosis patients. Neurochemistry international. 2010;56(5):687–693. doi: 10.1016/j.neuint.2010.02.004. Epub 2010/02/16. [DOI] [PubMed] [Google Scholar]
  • 90.Mendonca DM, Martins SC, Higashi R, Muscara MN, Neto VM, Chimelli L, Martinez AM. Neurofilament heavy subunit in cerebrospinal fluid: a biomarker of amyotrophic lateral sclerosis? Amyotrophic lateral sclerosis : official publication of the World Federation of Neurology Research Group on Motor Neuron Diseases. 2011;12(2):144–147. doi: 10.3109/17482968.2010.542002. Epub 2011/01/05. [DOI] [PubMed] [Google Scholar]
  • 91.McGuire V, Longstreth WT, Jr, Nelson LM, Koepsell TD, Checkoway H, Morgan MS, van Belle G. Occupational exposures amyotrophic lateral sclerosis A population-based case-control study. American journal of epidemiology. 1997;145(12):1076–1088. doi: 10.1093/oxfordjournals.aje.a009070. Epub 1997/06/15. [DOI] [PubMed] [Google Scholar]
  • 92.Weisskopf MG, Morozova N, O'Reilly EJ, McCullough ML, Calle EE, Thun MJ, Ascherio A. Prospective study of chemical exposures and amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry. 2009;80(5):558–561. doi: 10.1136/jnnp.2008.156976. Epub 2009/04/18; PubMed Central PMCID: PMC2765376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Govoni V, Granieri E, Fallica E, Casetta I. Amyotrophic lateral sclerosis, rural environment and agricultural work in the Local Health District of Ferrara, Italy, in the years 1964–1998. J Neurol. 2005;252(11):1322–1327. doi: 10.1007/s00415-005-0859-z. Epub 2005/07/05. [DOI] [PubMed] [Google Scholar]
  • 94.Park RM, Schulte PA, Bowman JD, Walker JT, Bondy SC, Yost MG, Touchstone JA, Dosemeci M. Potential occupational risks for neurodegenerative diseases. Am J Ind Med. 2005;48(1):63–77. doi: 10.1002/ajim.20178. Epub 2005/06/09. [DOI] [PubMed] [Google Scholar]
  • 95.Morahan JM, Pamphlett R. Amyotrophic lateral sclerosis and exposure to environmental toxins: an Australian case-control study. Neuroepidemiology. 2006;27(3):130–105. doi: 10.1159/000095552. Epub 2006/09/02. [DOI] [PubMed] [Google Scholar]
  • 96.Furby A, Beauvais K, Kolev I, Rivain JG, Sebille V. Rural environment and risk factors of amyotrophic lateral sclerosis: a case-control study. Journal of neurology. 2010;257(5):792–798. doi: 10.1007/s00415-009-5419-5. Epub 2009/12/17. [DOI] [PubMed] [Google Scholar]
  • 97.Bonvicini F, Marcello N, Mandrioli J, Pietrini V, Vinceti M. Exposure to pesticides and risk of amyotrophic lateral sclerosis: a population-based case-control study. Annali dell'Istituto superiore di sanita. 2010;46(3):284–287. doi: 10.4415/ANN_10_03_10. Epub 2010/09/18. [DOI] [PubMed] [Google Scholar]
  • 98.Chancellor AM, Slattery JM, Fraser H, Warlow CP. Risk factors for motor neuron disease: a case-control study based on patients from the Scottish Motor Neuron Disease Register. J Neurol Neurosurg Psychiatry. 1993;56(11):1200–1206. doi: 10.1136/jnnp.56.11.1200. Epub 1993/11/01; PubMed Central PMCID: PMC489821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Schulte PA, Burnett CA, Boeniger MF. Neurodegenerative diseases: Occupational occurrence and potential risk factors, 1982 through 1991. Am J Public Health. 1996;86(9):1281–1288. doi: 10.2105/ajph.86.9.1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Bergomi M, Vinceti M, Nacci G, Pietrini V, Bratter P, Alber D, Ferrari A, Vescovi L, Guidetti D, Sola P, Malagu S, Aramini C, Vivoli G. Environmental exposure to trace elements and risk of amyotrophic lateral sclerosis: a population-based case-control study. Environ Res. 2002;89(2):116–123. doi: 10.1006/enrs.2002.4361. Epub 2002/07/19. [DOI] [PubMed] [Google Scholar]
  • 101.Cruz DC, Nelson LM, McGuire V, Longstreth WT., Jr Physical trauma and family history of neurodegenerative diseases in amyotrophic lateral sclerosis: a population-based case-control study. Neuroepidemiology. 1999;18(2):101–110. doi: 10.1159/000069413. Epub 1999/02/19. [DOI] [PubMed] [Google Scholar]
  • 102.Gunnarsson LG, Lindberg G, Soderfeldt B, Axelson O. Amyotrophic lateral sclerosis in Sweden in relation to occupation. Acta neurologica Scandinavica. 1991;83(6):394–398. doi: 10.1111/j.1600-0404.1991.tb03970.x. Epub 1991/06/01. [DOI] [PubMed] [Google Scholar]
  • 103.Armon C. An evidence-based medicine approach to the evaluation of the role of exogenous risk factors in sporadic amyotrophic lateral sclerosis. Neuroepidemiology. 2003;22(4):217–228. doi: 10.1159/000070562. [DOI] [PubMed] [Google Scholar]
  • 104.Sutedja NA, Veldink JH, Fischer K, Kromhout H, Heederik D, Huisman MH, Wokke JH, van den Berg LH. Exposure to chemicals and metals and risk of amyotrophic lateral sclerosis: a systematic review. Amyotrophic lateral sclerosis : official publication of the World Federation of Neurology Research Group on Motor Neuron Diseases. 2009;10(5–6):302–309. doi: 10.3109/17482960802455416. Epub 2009/11/20. [DOI] [PubMed] [Google Scholar]
  • 105.USGPO, editor. Bureau of the Census UDoC. 1990 census of population and housing. Summary: social, economic and housing characteristics. Washington: United States; p. DC1991. [Google Scholar]
  • 106.Thun MJ, Calle EE, Rodriguez C, Wingo PA. Epidemiological research at the American Cancer Society. Cancer Epidem Biomar. 2000;9(9):861–868. [PubMed] [Google Scholar]
  • 107.Shadnia S, Azizi E, Hosseini R, Khoei S, Fouladdel S, Pajoumand A, Jalali N, Abdollahi M. Evaluation of oxidative stress and genotoxicity in organophosphorus insecticide formulators. Hum Exp Toxicol. 2005;24(9):439–445. doi: 10.1191/0960327105ht549oa. Epub 2005/10/21. [DOI] [PubMed] [Google Scholar]
  • 108.Franco R, Sanchez-Olea R, Reyes-Reyes EM, Panayiotidis MI. Environmental toxicity, oxidative stress and apoptosis: menage a trois. Mutat Res. 2009;674(1–2):3–22. doi: 10.1016/j.mrgentox.2008.11.012. Epub 2008/12/31. [DOI] [PubMed] [Google Scholar]
  • 109.Soltaninejad K, Abdollahi M. Current opinion on the science of organophosphate pesticides and toxic stress: a systematic review. Med Sci Monit. 2009;15(3):RA75–RA90. Epub 2009/02/28. [PubMed] [Google Scholar]
  • 110.Schmuck G, Rohrdanz E, Tran-Thi QH, Kahl R, Schluter G. Oxidative stress in rat cortical neurons and astrocytes induced by paraquat in vitro. Neurotox Res. 2002;4(1):1–13. doi: 10.1080/10298420290007574. Epub 2003/06/27. [DOI] [PubMed] [Google Scholar]
  • 111.Costa LG. Current issues in organophosphate toxicology. Clin Chim Acta. 2006;366(1–2):1–13. doi: 10.1016/j.cca.2005.10.008. Epub 2005/12/13. [DOI] [PubMed] [Google Scholar]
  • 112.Lushchak VI. Glutathione homeostasis and functions: potential targets for medical interventions. J Amino Acids. 2012;2012:736837. doi: 10.1155/2012/736837. Epub 2012/04/14; PubMed Central PMCID: PMC3303626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Griffith OW, Meister A. Glutathione: interorgan translocation, turnover, and metabolism. Proc Natl Acad Sci U S A. 1979;76(11):5606–5610. doi: 10.1073/pnas.76.11.5606. Epub 1979/11/01; PubMed Central PMCID: PMC411698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Muniz JF, McCauley L, Scherer J, Lasarev M, Koshy M, Kow YW, Nazar-Stewart V, Kisby GE. Biomarkers of oxidative stress and DNA damage in agricultural workers: a pilot study. Toxicol Appl Pharmacol. 2008;227(1):97–107. doi: 10.1016/j.taap.2007.10.027. Epub 2007/12/19. [DOI] [PubMed] [Google Scholar]
  • 115.Franco R, Li S, Rodriguez-Rocha H, Burns M, Panayiotidis MI. Molecular mechanisms of pesticide-induced neurotoxicity: Relevance to Parkinson's disease. Chem Biol Interact. 2010;188(2):289–300. doi: 10.1016/j.cbi.2010.06.003. Epub 2010/06/15; PubMed Central PMCID: PMC2942983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Suntres ZE. Role of antioxidants in paraquat toxicity. Toxicology. 2002;180(1):65–77. doi: 10.1016/s0300-483x(02)00382-7. Epub 2002/09/27. [DOI] [PubMed] [Google Scholar]
  • 117.Dutheil F, Beaune P, Loriot MA. Xenobiotic metabolizing enzymes in the central nervous system: Contribution of cytochrome P450 enzymes in normal and pathological human brain. Biochimie. 2008;90(3):426–436. doi: 10.1016/j.biochi.2007.10.007. Epub 2007/11/14. [DOI] [PubMed] [Google Scholar]
  • 118.Day BJ, Patel M, Calavetta L, Chang LY, Stamler JS. A mechanism of paraquat toxicity involving nitric oxide synthase. Proc Natl Acad Sci U S A. 1999;96(22):12760–12765. doi: 10.1073/pnas.96.22.12760. Epub 1999/10/27; PubMed Central PMCID: PMC23088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Purisai MG, McCormack AL, Cumine S, Li J, Isla MZ, Di Monte DA. Microglial activation as a priming event leading to paraquat-induced dopaminergic cell degeneration. Neurobiology of Disease. 2007;25(2):392–400. doi: 10.1016/j.nbd.2006.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wu XF, Block ML, Zhang W, Qin L, Wilson B, Zhang WQ, Veronesi B, Hong JS. The role of microglia in paraquat-induced dopaminergic neurotoxicity. Antioxid Redox Signal. 2005;7(5–6):654–661. doi: 10.1089/ars.2005.7.654. Epub 2005/05/14. [DOI] [PubMed] [Google Scholar]
  • 121.Kim S, Hwang J, Lee WH, Hwang DY, Suk K. Role of protein kinase Cdelta in paraquat-induced glial cell death. J Neurosci Res. 2008;86(9):2062–2070. doi: 10.1002/jnr.21643. Epub 2008/03/13. [DOI] [PubMed] [Google Scholar]
  • 122.Cocheme HM, Murphy MP. Complex I is the major site of mitochondrial superoxide production by paraquat. Journal of Biological Chemistry. 2008;283(4):1786–1798. doi: 10.1074/jbc.M708597200. [DOI] [PubMed] [Google Scholar]
  • 123.Choi WS, Kruse SE, Palmiter RD, Xia ZG. Mitochondrial complex I inhibition is not required for dopaminergic neuron death induced by rotenone, MPP(+), or paraquat. P Natl Acad Sci USA. 2008;105(39):15136–15141. doi: 10.1073/pnas.0807581105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Mitsumoto H, Hanson MR, Chad DA. Amyotrophic Lateral Sclerosis - Recent Advances in Pathogenesis and Therapeutic Trials. Arch Neurol-Chicago. 1988;45(2):189–202. doi: 10.1001/archneur.1988.00520260077025. [DOI] [PubMed] [Google Scholar]
  • 125.Callaghan B, Feldman D, Gruis K, Feldman E. The association of exposure to lead, mercury, and selenium and the development of amyotrophic lateral sclerosis and the epigenetic implications. Neuro-degenerative diseases. 2011;8(1–2):1–8. doi: 10.1159/000315405. Epub 2010/08/07. [DOI] [PubMed] [Google Scholar]
  • 126.Vinceti M, Nacci G, Rocchi E, Cassinadri T, Vivoli R, Marchesi C, Bergomi M. Mortality in a population with long-term exposure to inorganic selenium via drinking water. J Clin Epidemiol. 2000;53(10):1062–1068. doi: 10.1016/s0895-4356(00)00233-x. Epub 2000/10/12. [DOI] [PubMed] [Google Scholar]
  • 127.Gait R, Maginnis C, Lewis S, Pickering N, Antoniak M, Hubbard R, Lawson I, Britton J. Occupational exposure to metals and solvents and the risk of motor neuron disease A case-control study. Neuroepidemiology. 2003;22(6):353–356. doi: 10.1159/000072925. Epub 2003/10/15. [DOI] [PubMed] [Google Scholar]
  • 128.Kamel F, Umbach DM, Munsat TL, Shefner JM, Hu H, Sandler DP. Lead exposure and amyotrophic lateral sclerosis. Epidemiology. 2002;13(3):311–319. doi: 10.1097/00001648-200205000-00012. Epub 2002/04/20. [DOI] [PubMed] [Google Scholar]
  • 129.Kamel F, Umbach DM, Lehman TA, Park LP, Munsat TL, Shefner JM, Sandler DP, Hu H, Taylor JA. Amyotrophic lateral sclerosis, lead, and genetic susceptibility: polymorphisms in the delta-aminolevulinic acid dehydratase and vitamin D receptor genes. Environ Health Perspect. 2003;111(10):1335–1339. doi: 10.1289/ehp.6109. Epub 2003/08/05; PubMed Central PMCID: PMC1241615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kamel F, Umbach DM, Stallone L, Richards M, Hu H, Sandler DP. Association of lead exposure with survival in amyotrophic lateral sclerosis. Environ Health Perspect. 2008;116(7):943–947. doi: 10.1289/ehp.11193. Epub 2008/07/17; PubMed Central PMCID: PMC2453164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Bergdahl IA, Gerhardsson L, Schutz A, Desnick RJ, Wetmur JG, Skerfving S. Delta-aminolevulinic acid dehydratase polymorphism: influence on lead levels and kidney function in humans. Arch Environ Health. 1997;52(2):91–96. doi: 10.1080/00039899709602870. Epub 1997/03/01. [DOI] [PubMed] [Google Scholar]
  • 132.Kelada SN, Shelton E, Kaufmann RB, Khoury MJ. Delta-aminolevulinic acid dehydratase genotype and lead toxicity: a HuGE review. Am J Epidemiol. 2001;154(1):1–13. doi: 10.1093/aje/154.1.1. Epub 2001/06/28. [DOI] [PubMed] [Google Scholar]
  • 133.Chetty CS, Vemuri MC, Campbell K, Suresh C. Lead-induced cell death of human neuroblastoma cells involves GSH deprivation. Cell Mol Biol Lett. 2005;10(3):413–423. Epub 2005/10/12. [PubMed] [Google Scholar]
  • 134.Qian Y, Zheng Y, Ramos KS, Tiffany-Castiglioni E. The involvement of copper transporter in lead-induced oxidative stress in astroglia. Neurochem Res. 2005;30(4):429–438. doi: 10.1007/s11064-005-2677-1. Epub 2005/08/04. [DOI] [PubMed] [Google Scholar]
  • 135.Cabell L, Ferguson C, Luginbill D, Kern M, Weingart A, Audesirk G. Differential induction of heme oxygenase and other stress proteins in cultured hippocampal astrocytes and neurons by inorganic lead. Toxicol Appl Pharmacol. 2004;198(1):49–60. doi: 10.1016/j.taap.2004.03.001. Epub 2004/06/23. [DOI] [PubMed] [Google Scholar]
  • 136.Fowler BA, Whittaker MH, Lipsky M, Wang G, Chen XQ. Oxidative stress induced by lead, cadmium and arsenic mixtures: 30-day, 90-day, and 180-day drinking water studies in rats: an overview. Biometals. 2004;17(5):567–568. doi: 10.1023/b:biom.0000045740.52182.9d. Epub 2005/02/04. [DOI] [PubMed] [Google Scholar]
  • 137.Prasanthi RPJ, Devi CB, Basha DC, Reddy NS, Reddy GR. Calcium and zinc supplementation protects lead (Pb)-induced perturbations in antioxidant enzymes and lipid peroxidation in developing mouse brain. International Journal of Developmental Neuroscience. 2010;28(2):161–167. doi: 10.1016/j.ijdevneu.2009.12.002. [DOI] [PubMed] [Google Scholar]
  • 138.Tiffany-Castiglioni E. Cell culture models for lead toxicity in neuronal and glial cells. Neurotoxicology. 1993;14(4):513–536. Epub 1993/01/01. [PubMed] [Google Scholar]
  • 139.Barbeito AG, Martinez-Palma L, Vargas MR, Pehar M, Manay N, Beckman JS, Barbeito L, Cassina P. Lead exposure stimulates VEGF expression in the spinal cord and extends survival in a mouse model of ALS. Neurobiol Dis. 2010;37(3):574–580. doi: 10.1016/j.nbd.2009.11.007. Epub 2009/11/17; PubMed Central PMCID: PMC2824063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bogaert E, Van Damme P, Poesen K, Dhondt J, Hersmus N, Kiraly D, Scheveneels W, Robberecht W, Van Den Bosch L. VEGF protects motor neurons against excitotoxicity by upregulation of GluR2. Neurobiol Aging. 2010;31(12):2185–2191. doi: 10.1016/j.neurobiolaging.2008.12.007. Epub 2009/02/03. [DOI] [PubMed] [Google Scholar]
  • 141.Tovar YRLB, Tapia R. VEGF protects spinal motor neurons against chronic excitotoxic degeneration in vivo by activation of PI3-K pathway and inhibition of p38MAPK. J Neurochem. 2010;115(5):1090–1101. doi: 10.1111/j.1471-4159.2010.06766.x. Epub 2010/05/12. [DOI] [PubMed] [Google Scholar]
  • 142.Lambrechts D, Storkebaum E, Morimoto M, Del-Favero J, Desmet F, Marklund SL, Wyns S, Thijs V, Andersson J, van Marion I, Al-Chalabi A, Bornes S, Musson R, Hansen V, Beckman L, Adolfsson R, Pall HS, Prats H, Vermeire S, Rutgeerts P, Katayama S, Awata T, Leigh N, Lang-Lazdunski L, Dewerchin M, Shaw C, Moons L, Vlietinck R, Morrison KE, Robberecht W, Van Broeckhoven C, Collen D, Andersen PM, Carmeliet P. VEGF is a modifier of amyotrophic lateral sclerosis in mice and humans and protects motoneurons against ischemic death. Nature Genetics. 2003;34(4):383–394. doi: 10.1038/ng1211. [DOI] [PubMed] [Google Scholar]
  • 143.Jasso-Pineda Y, Diaz-Barriga F, Calderon J, Yanez L, Carrizales L, Perez-Maldonado IN. DNA Damage and Decreased DNA Repair in Peripheral Blood Mononuclear Cells in Individuals Exposed to Arsenic and Lead in a Mining Site. Biol Trace Elem Res. 2011 doi: 10.1007/s12011-011-9237-0. Epub 2011/10/22. [DOI] [PubMed] [Google Scholar]
  • 144.Coffman CJ, Horner RD, Grambow SC, Lindquist J, Studi IVc. Estimating the occurrence of amyotrophic lateral sclerosis among Gulf War (1990–1991) Veterans using capture-recapture methods - An assessment of case ascertainment bias. Neuroepidemiology. 2005;24(3):141–150. doi: 10.1159/000083297. [DOI] [PubMed] [Google Scholar]
  • 145.Horner RD, Kamins KG, Feussner JR, Grambow SC, Hoff-Lindquist J, Harati Y, Mitsumoto H, Pascuzzi R, Spencer PS, Tim R, Howard D, Smith TC, Ryan MAK, Coffman CJ, Kasarskis EJ. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology. 2003;61(6):742–749. doi: 10.1212/01.wnl.0000069922.32557.ca. [DOI] [PubMed] [Google Scholar]
  • 146.Haley RW. Excess incidence of ALS in young Gulf War veterans. Neurology. 2003;61(6):750–756. doi: 10.1212/wnl.61.6.750. [DOI] [PubMed] [Google Scholar]
  • 147.Horner RD, Grambow SC, Coffman CJ, Lindquist JH, Oddone EZ, Allen KD, Kasarskis EJ. Amyotrophic lateral sclerosis among 1991 Gulf War veterans: evidence for a time-limited outbreak. Neuroepidemiology. 2008;31(1):28–32. doi: 10.1159/000136648. Epub 2008/06/07. [DOI] [PubMed] [Google Scholar]
  • 148.Sharief MK, Priddin J, Delamont RS, Unwin C, Rose MR, David A, Wessely S. Neurophysiologic analysis of neuromuscular symptoms in UK Gulf War veterans: a controlled study. Neurology. 2002;59(10):1518–1525. doi: 10.1212/01.wnl.0000032755.27372.fc. Epub 2002/11/27. [DOI] [PubMed] [Google Scholar]
  • 149.Kang HK, Bullman TA. Mortality among US veterans of the Persian Gulf War: 7-year follow-up. American Journal of Epidemiology. 2001;154(5):399–405. doi: 10.1093/aje/154.5.399. [DOI] [PubMed] [Google Scholar]
  • 150.Weisskopf MG, O'Reilly EJ, McCullough ML, Calle EE, Thun MJ, Cudkowicz M, Ascherio A. Prospective study of military service and mortality from ALS. Neurology. 2005;64(1):32–37. doi: 10.1212/01.WNL.0000148649.17706.D9. Epub 2005/01/12. [DOI] [PubMed] [Google Scholar]
  • 151.Pastula DM, Coffman CJ, Allen KD, Oddone EZ, Kasarskis EJ, Lindquist JH, Morgenlander JC, Norman BB, Rozear MP, Sams LA, Sabet A, Bedlack RS. Factors associated with survival in the National Registry of Veterans with ALS. Amyotroph Lateral Scler. 2009;10(5–6):332–338. doi: 10.3109/17482960802320545. Epub 2009/11/20. [DOI] [PubMed] [Google Scholar]
  • 152.Allen KD, Kasarskis EJ, Bedlack RS, Rozear MP, Morgenlander JC, Sabet A, Sams L, Lindquist JH, Harrelson ML, Coffman CJ, Oddone EZ. The National Registry of Veterans with Amyotrophic Lateral Sclerosis. Neuroepidemiology. 2008;30(3):180–190. doi: 10.1159/000126910. [DOI] [PubMed] [Google Scholar]
  • 153.Fang F, Kwee LC, Allen KD, Umbach DM, Ye WM, Watson M, Keller J, Oddone EZ, Sandler DP, Schmidt S, Kamel F. Association Between Blood Lead and the Risk of Amyotrophic Lateral Sclerosis. American Journal of Epidemiology. 2010;171(10):1126–1133. doi: 10.1093/aje/kwq063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Schmidt S, Kwee LC, Allen KD, Oddone EZ. Association of ALS with head injury, cigarette smoking and APOE genotypes. J Neurol Sci. 2010;291(1–2):22–29. doi: 10.1016/j.jns.2010.01.011. Epub 2010/02/05; PubMed Central PMCID: PMC2840700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kurtzke JF, Beebe GW. Epidemiology of amyotrophic lateral sclerosis: 1A case-control comparison based on ALS deaths. Neurology. 1980;30(5):453–462. doi: 10.1212/wnl.30.5.453. Epub 1980/05/01. [DOI] [PubMed] [Google Scholar]
  • 156.Kondo K, Tsubaki T. Case-control studies of motor neuron disease: association with mechanical injuries. Arch Neurol. 1981;38(4):220–226. doi: 10.1001/archneur.1981.00510040046007. Epub 1981/04/01. [DOI] [PubMed] [Google Scholar]
  • 157.Deapen DM, Henderson BE. A case-control study of amyotrophic lateral sclerosis. Am J Epidemiol. 1986;123(5):790–799. doi: 10.1093/oxfordjournals.aje.a114308. Epub 1986/05/01. [DOI] [PubMed] [Google Scholar]
  • 158.Granieri E, Carreras M, Tola R, Paolino E, Tralli G, Eleopra R, Serra G. Motor neuron disease in the province of Ferrara, Italy, in 1964–1982. Neurology. 1988;38(10):1604–1608. doi: 10.1212/wnl.38.10.1604. Epub 1988/10/01. [DOI] [PubMed] [Google Scholar]
  • 159.Chio A, Meineri P, Tribolo A, Schiffer D. Risk factors in motor neuron disease: a case-control study. Neuroepidemiology. 1991;10(4):174–184. doi: 10.1159/000110267. Epub 1991/01/01. [DOI] [PubMed] [Google Scholar]
  • 160.Gresham LS, Molgaard CA, Golbeck AL, Smith R. Amyotrophic lateral sclerosis and history of skeletal fracture: a case-control study. Neurology. 1987;37(4):717–719. doi: 10.1212/wnl.37.4.717. Epub 1987/04/01. [DOI] [PubMed] [Google Scholar]
  • 161.Gallagher JP, Sanders M. Trauma and amyotrophic lateral sclerosis: a report of 78 patients. Acta Neurol Scand. 1987;75(2):145–150. doi: 10.1111/j.1600-0404.1987.tb07909.x. Epub 1987/02/01. [DOI] [PubMed] [Google Scholar]
  • 162.Williams DB, Annegers JF, Kokmen E, O'Brien PC, Kurland LT. Brain injury and neurologic sequelae: a cohort study of dementia, parkinsonism, and amyotrophic lateral sclerosis. Neurology. 1991;41(10):1554–1557. doi: 10.1212/wnl.41.10.1554. Epub 1991/10/01. [DOI] [PubMed] [Google Scholar]
  • 163.Beghi E, Logroscino G, Chio A, Hardiman O, Millul A, Mitchell D, Swingler R, Traynor BJ. Amyotrophic lateral sclerosis, physical exercise, trauma and sports: results of a population-based pilot case-control study. Amyotrophic lateral sclerosis : official publication of the World Federation of Neurology Research Group on Motor Neuron Diseases. 2010;11(3):289–292. doi: 10.3109/17482960903384283. Epub 2010/05/04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Turner MR, Abisgold J, Yeates DGR, Talbot K, Goldacre MJ. Head and other physical trauma requiring hospitalisation is not a significant risk factor in the development of ALS. Journal of the Neurological Sciences. 2010;288(1–2):45–48. doi: 10.1016/j.jns.2009.10.010. [DOI] [PubMed] [Google Scholar]
  • 165.Chio A, Benzi G, Dossena M, Mutani R, Mora G. Severely increased risk of amyotrophic lateral sclerosis among Italian professional football players. Brain. 2005;128(Pt 3):472–476. doi: 10.1093/brain/awh373. Epub 2005/01/07. [DOI] [PubMed] [Google Scholar]
  • 166.Chio A, Calvo A, Dossena M, Ghiglione P, Mutani R, Mora G. ALS in Italian professional soccer players: the risk is still present and could be soccer-specific. Amyotrophic lateral sclerosis : official publication of the World Federation of Neurology Research Group on Motor Neuron Diseases. 2009;10(4):205–209. doi: 10.1080/17482960902721634. Epub 2009/03/10. [DOI] [PubMed] [Google Scholar]
  • 167.Belli S, Vanacore N. Proportionate mortality of Italian soccer players: is amyotrophic lateral sclerosis an occupational disease? Eur J Epidemiol. 2005;20(3):237–242. doi: 10.1007/s10654-004-6879-7. Epub 2005/06/01. [DOI] [PubMed] [Google Scholar]
  • 168.Abel EL. Football increases the risk for Lou Gehrig's disease, amyotrophic lateral sclerosis. Percept Mot Skills. 2007;104(3 Pt 2):1251–1254. doi: 10.2466/pms.104.4.1251-1254. Epub 2007/09/21. [DOI] [PubMed] [Google Scholar]
  • 169.Armon C, Nelson LM. Is head trauma a risk factor for amyotrophic lateral sclerosis? An evidence based review. Amyotroph Lateral Sc. 0(0):1–6. doi: 10.3109/17482968.2012.660954. [DOI] [PubMed] [Google Scholar]
  • 170.Harden CL, Meador KJ, Pennell PB, Hauser WA, Gronseth GS, French JA, Wiebe S, Thurman D, Koppel BS, Kaplan PW, Robinson JN, Hopp J, Ting TY, Gidal B, Hovinga CA, Wilner AN, Vazquez B, Holmes L, Krumholz A, Finnell R, Hirtz D, Le Guen C. Practice parameter update: management issues for women with epilepsy--focus on pregnancy (an evidence-based review): teratogenesis and perinatal outcomes: report of the Quality Standards Subcommittee and Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology and American Epilepsy Society. Neurology. 2009;73(2):133–141. doi: 10.1212/WNL.0b013e3181a6b312. Epub 2009/04/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Serbest G, Burkhardt MF, Siman R, Raghupathi R, Saatman KE. Temporal profiles of cytoskeletal protein loss following traumatic axonal injury in mice. Neurochem Res. 2007;32(12):2006–2014. doi: 10.1007/s11064-007-9318-9. Epub 2007/04/03. [DOI] [PubMed] [Google Scholar]
  • 172.Cole K, Perez-Polo JR. Neuronal trauma model: in search of Thanatos. Int J Dev Neurosci. 2004;22(7):485–496. doi: 10.1016/j.ijdevneu.2004.07.015. Epub 2004/10/07. [DOI] [PubMed] [Google Scholar]
  • 173.McKee AC, Gavett BE, Stern RA, Nowinski CJ, Cantu RC, Kowall NW, Perl DP, Hedley-Whyte ET, Price B, Sullivan C, Morin P, Lee HS, Kubilus CA, Daneshvar DH, Wulff M, Budson AE. TDP-43 Proteinopathy and Motor Neuron Disease in Chronic Traumatic Encephalopathy. Journal of neuropathology and experimental neurology. 2010;69(9):918–929. doi: 10.1097/NEN.0b013e3181ee7d85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Strong MJ, Volkening K, Hammond R, Yang WC, Strong W, Leystra-Lantz C, Shoesmith C. TDP43 is a human low molecular weight neurofilament (hNFL) mRNA-binding protein. Mol Cell Neurosci. 2007;35(2):320–327. doi: 10.1016/j.mcn.2007.03.007. [DOI] [PubMed] [Google Scholar]
  • 175.Moisse K, Volkening K, Leystra-Lantz C, Welch I, Hill T, Strong MJ. Divergent patterns of cytosolic TDP-43 and neuronal progranulin expression following axotomy: Implications for TDP-43 in the physiological response to neuronal injury. Brain Research. 2009;1249:202–211. doi: 10.1016/j.brainres.2008.10.021. [DOI] [PubMed] [Google Scholar]
  • 176.Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y, Oda T. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Bioph Res Co. 2006;351(3):602–611. doi: 10.1016/j.bbrc.2006.10.093. [DOI] [PubMed] [Google Scholar]
  • 177.Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314(5796):130–133. doi: 10.1126/science.1134108. Epub 2006/10/07. [DOI] [PubMed] [Google Scholar]
  • 178.Ferrari R, Kapogiannis D, Huey ED, Momeni P. FTD and ALS: a tale of two diseases. Curr Alzheimer Res. 2011;8(3):273–294. doi: 10.2174/156720511795563700. Epub 2011/01/13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yang W, Strong MJ. Widespread neuronal and glial hyperphosphorylated tau deposition in ALS with cognitive impairment. Amyotroph Lateral Scler. 2012;13(2):178–193. doi: 10.3109/17482968.2011.622405. Epub 2012/01/05. [DOI] [PubMed] [Google Scholar]
  • 180.Maekawa S, Leigh PN, King A, Jones E, Steele JC, Bodi I, Shaw CE, Hortobagyi T, Al-Sarraj S. TDP-43 is consistently co-localized with ubiquitinated inclusions in sporadic and Guam amyotrophic lateral sclerosis but not in familial amyotrophic lateral sclerosis with and without SOD1 mutations. Neuropathology : official journal of the Japanese Society of Neuropathology. 2009;29(6):672–683. doi: 10.1111/j.1440-1789.2009.01029.x. Epub 2009/06/06. [DOI] [PubMed] [Google Scholar]
  • 181.Murray ME, DeJesus-Hernandez M, Rutherford NJ, Baker M, Duara R, Graff-Radford NR, Wszolek ZK, Ferman TJ, Josephs KA, Boylan KB, Rademakers R, Dickson DW. Clinical and neuropathologic heterogeneity of c9FTD/ALS associated with hexanucleotide repeat expansion in C9ORF72. Acta Neuropathologica. 2011;122(6):673–690. doi: 10.1007/s00401-011-0907-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Cohen TJ, Hwang AW, Unger T, Trojanowski JQ, Lee VM. Redox signalling directly regulates TDP-43 via cysteine oxidation and disulphide cross-linking. Embo J. 2011 doi: 10.1038/emboj.2011.471. Epub 2011/12/24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zhang HX, Tanji K, Yoshida H, Hayakari M, Shibata T, Mori F, Uchida K, Wakabayashi K. Alteration of biochemical and pathological properties of TDP-43 protein by a lipid mediator, 15-deoxy-Delta(12,14)-prostaglandin J(2) Exp Neurol. 2010;222(2):296–303. doi: 10.1016/j.expneurol.2010.01.007. Epub 2010/01/20. [DOI] [PubMed] [Google Scholar]
  • 184.Meyerowitz J, Parker SJ, Vella LJ, Ng D, Price KA, Liddell JR, Caragounis A, Li QX, Masters CL, Nonaka T, Hasegawa M, Bogoyevitch MA, Kanninen KM, Crouch PJ, White AR. C-Jun N-terminal kinase controls TDP-43 accumulation in stress granules induced by oxidative stress. Mol Neurodegener. 2011;6:57. doi: 10.1186/1750-1326-6-57. Epub 2011/08/09; PubMed Central PMCID: PMC3162576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Iguchi Y, Katsuno M, Takagi S, Ishigaki S, Niwa J, Hasegawa M, Tanaka F, Sobue G. Oxidative stress induced by glutathione depletion reproduces pathological modifications of TDP-43 linked to TDP-43 proteinopathies. Neurobiology of Disease. 2012;45(3):862–870. doi: 10.1016/j.nbd.2011.12.002. [DOI] [PubMed] [Google Scholar]
  • 186.Wang JS, Huang YH. Effects of exercise intensity on lymphocyte apoptosis induced by oxidative stress in men. Eur J Appl Physiol. 2005;95(4):290–297. doi: 10.1007/s00421-005-0005-8. Epub 2005/08/13. [DOI] [PubMed] [Google Scholar]
  • 187.Reid MB. Free radicals and muscle fatigue: Of ROS, canaries, and the IOC. Free Radic Biol Med. 2008;44(2):169–179. doi: 10.1016/j.freeradbiomed.2007.03.002. Epub 2008/01/15. [DOI] [PubMed] [Google Scholar]
  • 188.Siciliano G, D'Avino C, Del Corona A, Barsacchi R, Kusmic C, Rocchi A, Pastorini E, Murri L. Impaired oxidative metabolism and lipid peroxidation in exercising muscle from ALS patients. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002;3(2):57–62. doi: 10.1080/146608202760196011. Epub 2002/09/07. [DOI] [PubMed] [Google Scholar]
  • 189.Scarmeas N, Shih T, Stern Y, Ottman R, Rowland LP. Premorbid weight, body mass, and varsity athletics in ALS. Neurology. 2002;59(5):773–775. doi: 10.1212/wnl.59.5.773. Epub 2002/09/11. [DOI] [PubMed] [Google Scholar]
  • 190.Mattsson P, Lonnstedt I, Nygren I, Askmark H. Physical fitness, but not muscle strength, is a risk factor for death in amyotrophic lateral sclerosis at an early age. J Neurol Neurosur Ps. 2012;83(4):390–394. doi: 10.1136/jnnp.2010.218982. [DOI] [PubMed] [Google Scholar]
  • 191.Kamble PDV, Baji PS. Study of anthropological parameters, body composition, strength & endurance in basketball players. International Journal of Biological & Medical Research. 2012;3(1):1404–1406. [Google Scholar]
  • 192.Chiò A, Mora G. Physical fitness and amyotrophic lateral sclerosis: dangerous liaisons or common genetic pathways? Journal of Neurology, Neurosurgery & Psychiatry. 2012;83(4):389. doi: 10.1136/jnnp-2012-302351. [DOI] [PubMed] [Google Scholar]
  • 193.Nelson LM, Matkin C, Longstreth WT, Jr, McGuire V. Population-based case-control study of amyotrophic lateral sclerosis in western Washington State II Diet. Am J Epidemiol. 2000;151(2):164–173. doi: 10.1093/oxfordjournals.aje.a010184. Epub 2000/01/25. [DOI] [PubMed] [Google Scholar]
  • 194.Morozova N, Weisskopf MG, McCullough ML, Munger KL, Calle EE, Thun MJ, Ascherio A. Diet and amyotrophic lateral sclerosis. Epidemiology. 2008;19(2):324–337. doi: 10.1097/EDE.0b013e3181632c5d. Epub 2008/02/28. [DOI] [PubMed] [Google Scholar]
  • 195.Okamoto K, Kihira T, Kondo T, Kobashi G, Washio M, Sasaki S, Yokoyama T, Miyake Y, Sakamoto N, Inaba Y, Nagai M. Nutritional status and risk of amyotrophic lateral sclerosis in Japan. Amyotroph Lateral Scler. 2007;8(5):300–304. doi: 10.1080/17482960701472249. Epub 2007/09/14. [DOI] [PubMed] [Google Scholar]
  • 196.Wang X, Quinn PJ. Vitamin E and its function in membranes. Prog Lipid Res. 1999;38(4):309–336. doi: 10.1016/s0163-7827(99)00008-9. Epub 2000/05/04. [DOI] [PubMed] [Google Scholar]
  • 197.Jialal I, Grundy SM. Effect of dietary supplementation with alpha-tocopherol on the oxidative modification of low density lipoprotein. J Lipid Res. 1992;33(6):899–906. Epub 1992/06/01. [PubMed] [Google Scholar]
  • 198.Ascherio A, Weisskopf MG, O'Reilly EJ, Jacobs EJ, McCullough ML, Calle EE, Cudkowicz M, Thun MJ. Vitamin E intake and risk of amyotrophic lateral sclerosis. Annals of neurology. 2005;57(1):104–110. doi: 10.1002/ana.20316. Epub 2004/11/06. [DOI] [PubMed] [Google Scholar]
  • 199.Wang H, O'Reilly EJ, Weisskopf MG, Logroscino G, McCullough ML, Schatzkin A, Kolonel LN, Ascherio A. Vitamin E intake and risk of amyotrophic lateral sclerosis: a pooled analysis of data from 5 prospective cohort studies. American journal of epidemiology. 2011;173(6):595–602. doi: 10.1093/aje/kwq416. Epub 2011/02/22; PubMed Central PMCID: PMC3105261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Weisskopf MG, McCullough ML, Calle EE, Thun MJ, Cudkowicz M, Ascherio A. Prospective study of cigarette smoking and amyotrophic lateral sclerosis. American Journal of Epidemiology. 2004;160(1):26–33. doi: 10.1093/aje/kwh179. [DOI] [PubMed] [Google Scholar]
  • 201.Nelson LM, McGuire V, Longstreth WT, Jr, Matkin C. Population-based case-control study of amyotrophic lateral sclerosis in western Washington State. I. Cigarette smoking and alcohol consumption. Am J Epidemiol. 2000;151(2):156–163. doi: 10.1093/oxfordjournals.aje.a010183. Epub 2000/01/25. [DOI] [PubMed] [Google Scholar]
  • 202.Kamel F, Umbach DM, Munsat TL, Shefner JM, Sandler DP. Association of cigarette smoking with amyotrophic lateral sclerosis. Neuroepidemiology. 1999;18(4):194–202. doi: 10.1159/000026211. [DOI] [PubMed] [Google Scholar]
  • 203.Gallo V, Bueno-De-Mesquita HB, Vermeulen R, Andersen PM, Kyrozis A, Linseisen J, Kaaks R, Allen NE, Roddam AW, Boshuizen HC, Peeters PH, Palli D, Mattiello A, Sieri S, Tumino R, Jimenez-Martin JM, Diaz MJT, Suarez LR, Trichopoulou A, Agudo A, Arriola L, Barricante-Gurrea A, Bingham S, Khaw KT, Manjer J, Lindkvist B, Overvad K, Bach FW, Tjonneland A, Olsen A, Bergmann MM, Boeing H, Clavel-Chapelon F, Lund E, Hallmans G, Middleton L, Vineis P, Riboli E. Smoking and Risk for Amyotrophic Lateral Sclerosis: Analysis of the EPIC Cohort. Annals of Neurology. 2009;65(4):378–385. doi: 10.1002/ana.21653. [DOI] [PubMed] [Google Scholar]
  • 204.Sutedja NA, Veldink JH, Fischer K, Kromhout H, Wokke JHJ, Huisman MHB, Heederik DJJ, Van den Berg LH. Lifetime occupation, education, smoking, and risk of ALS. Neurology. 2007;69(15):1508–1514. doi: 10.1212/01.wnl.0000277463.87361.8c. [DOI] [PubMed] [Google Scholar]
  • 205.Wang H, O'Reilly EJ, Weisskopf MG, Logroscino G, McCullough ML, Thun MJ, Schatzkin A, Kolonel LN, Ascherio A. Smoking and risk of amyotrophic lateral sclerosis: a pooled analysis of 5 prospective cohorts. Arch Neurol. 2011;68(2):207–213. doi: 10.1001/archneurol.2010.367. Epub 2011/02/16; PubMed Central PMCID: PMC3319086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Alonso A, Logroscino G, Jick SS, Hernan MA. Association of smoking with amyotrophic lateral sclerosis risk and survival in men and women: a prospective study. BMC neurology. 2010;10:6. doi: 10.1186/1471-2377-10-6. Epub 2010/01/16; PubMed Central PMCID: PMC2820482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Okamoto K, Kihira T, Kondo T, Kobashi G, Washio M, Sasaki S, Yokoyama T, Miyake Y, Sakamoto N, Inaba Y, Nagai M. Lifestyle Factors and Risk of Amyotrophic Lateral Sclerosis: A Case-Control Study in Japan. Annals of Epidemiology. 2009;19(6):359–364. doi: 10.1016/j.annepidem.2009.01.015. [DOI] [PubMed] [Google Scholar]
  • 208.Fang F, Bellocco R, Hernan MA, Ye W. Smoking, snuff dipping and the risk of amyotrophic lateral sclerosis--a prospective cohort study. Neuroepidemiology. 2006;27(4):217–221. doi: 10.1159/000096956. Epub 2006/11/16. [DOI] [PubMed] [Google Scholar]
  • 209.de Jong SW, Huisman MH, Sutedja NA, van der Kooi AJ, de Visser M, Schelhaas HJ, Fischer K, Veldink JH, van den Berg LH. Smoking, Alcohol Consumption, and the Risk of Amyotrophic Lateral Sclerosis: A Population-based Study. Am J Epidemiol. 2012 doi: 10.1093/aje/kws015. Epub 2012/07/14. [DOI] [PubMed] [Google Scholar]
  • 210.Alonso A, Logroscino G, Hernán MA. Smoking and the risk of amyotrophic lateral sclerosis: a systematic review and meta-analysis. Journal of Neurology, Neurosurgery & Psychiatry. 2010;81(11):1249–1252. doi: 10.1136/jnnp.2009.180232. [DOI] [PubMed] [Google Scholar]
  • 211.Pamphlett R, Cochran Ward E. Smoking Is Not a Risk Factor for Sporadic Amyotrophic Lateral Sclerosis in an Australian Population. Neuroepidemiology. 2012;38(2):106–113. doi: 10.1159/000336013. [DOI] [PubMed] [Google Scholar]
  • 212.Armon C. Smoking may be considered an established risk factor for sporadic ALS. Neurology. 2009;73(20):1693–1698. doi: 10.1212/WNL.0b013e3181c1df48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Vayssier M, Banzet N, Francois D, Bellmann K, Polla BS. Tobacco smoke induces both apoptosis and necrosis in mammalian cells: differential effects of HSP70. Am J Physiol-Lung C. 1998;275(4):L771–L779. doi: 10.1152/ajplung.1998.275.4.L771. [DOI] [PubMed] [Google Scholar]
  • 214.Bertram KM, Baglole CJ, Phipps RP, Libby RT. Molecular regulation of cigarette smoke induced-oxidative stress in human retinal pigment epithelial cells: implications for age-related macular degeneration. Am J Physiol Cell Physiol. 2009;297(5):C1200–C1210. doi: 10.1152/ajpcell.00126.2009. Epub 2009/09/18; PubMed Central PMCID: PMC2777395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Yanbaeva DG, Dentener MA, Creutzberg EC, Wesseling G, Wouters EF. Systemic effects of smoking. Chest. 2007;131(5):1557–1566. doi: 10.1378/chest.06-2179. Epub 2007/05/15. [DOI] [PubMed] [Google Scholar]
  • 216.Kiziler AR, Aydemir B, Onaran I, Alici B, Ozkara H, Gulyasar T, Akyolcu MC. High levels of cadmium and lead in seminal fluid and blood of smoking men are associated with high oxidative stress and damage in infertile subjects. Biol Trace Elem Res. 2007;120(1–3):82–91. doi: 10.1007/s12011-007-8020-8. Epub 2007/10/06. [DOI] [PubMed] [Google Scholar]
  • 217.Loikkanen J, Naarala J, Vahakangas KH, Savolainen KM. Glutamate increases toxicity of inorganic lead in GT1-7 neurons: partial protection induced by flunarizine. Arch Toxicol. 2003;77(12):663–671. doi: 10.1007/s00204-003-0498-z. Epub 2003/09/05. [DOI] [PubMed] [Google Scholar]
  • 218.Kriscenski-Perry E, Durham HD, Sheu SS, Figlewicz DA. Synergistic effects of low level stressors in an oxidative damage model of spinal motor neuron degeneration. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002;3(3):151–157. doi: 10.1080/146608202760834166. Epub 2002/12/24. [DOI] [PubMed] [Google Scholar]
  • 219.Sahin E, Gumuslu S. Alterations in brain antioxidant status, protein oxidation and lipid peroxidation in response to different stress models. Behav Brain Res. 2004;155(2):241–248. doi: 10.1016/j.bbr.2004.04.022. Epub 2004/09/15. [DOI] [PubMed] [Google Scholar]
  • 220.Ilzecka J. Total antioxidant status is increased in the serum of amyotrophic lateral sclerosis patients. Scand J Clin Lab Inv. 2003;63(4):297–302. doi: 10.1080/00365510310001771. [DOI] [PubMed] [Google Scholar]
  • 221.Babu GN, Kumar A, Chandra R, Puri SK, Singh RL, Kalita J, Misra UK. Oxidant-antioxidant imbalance in the erythrocytes of sporadic amyotrophic lateral sclerosis patients correlates with the progression of disease. Neurochem Int. 2008;52(6):1284–1289. doi: 10.1016/j.neuint.2008.01.009. Epub 2008/03/01. [DOI] [PubMed] [Google Scholar]
  • 222.Baillet A, Chanteperdrix V, Trocme C, Casez P, Garrel C, Besson G. The role of oxidative stress in amyotrophic lateral sclerosis and Parkinson's disease. Neurochemical research. 2010;35(10):1530–1537. doi: 10.1007/s11064-010-0212-5. Epub 2010/06/11. [DOI] [PubMed] [Google Scholar]
  • 223.Maier CM, Chan PH. Role of superoxide dismutases in oxidative damage and neurodegenerative disorders. Neuroscientist. 2002;8(4):323–334. doi: 10.1177/107385840200800408. Epub 2002/08/27. [DOI] [PubMed] [Google Scholar]
  • 224.Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O'Regan JP, Deng HX, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362(6415):59–62. doi: 10.1038/362059a0. Epub 1993/03/04. [DOI] [PubMed] [Google Scholar]
  • 225.Synofzik M, Ronchi D, Keskin I, Basak AN, Wilhelm C, Gobbi C, Birve A, Biskup S, Zecca C, Fernandez-Santiago R, Kaugesaar T, Schols L, Marklund SL, Andersen PM. Mutant superoxide dismutase-1 indistinguishable from wild-type causes ALS. Hum Mol Genet. 2012;21(16):3568–3574. doi: 10.1093/hmg/dds188. Epub 2012/05/19. [DOI] [PubMed] [Google Scholar]
  • 226.Shibata N, Nagai R, Miyata S, Jono T, Horiuchi S, Hirano A, Kato S, Sasaki S, Asayama K, Kobayashi M. Nonoxidative protein glycation is implicated in familial amyotrophic lateral sclerosis with superoxide dismutase-1 mutation. Acta Neuropathologica. 2000;100(3):275–284. doi: 10.1007/s004019900173. [DOI] [PubMed] [Google Scholar]
  • 227.Shibata N, Hirano A, Yamamoto T, Kato Y, Kobayashi M. Superoxide dismutase-1 mutation-related neurotoxicity in familial amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1(3):143–161. doi: 10.1080/14660820050515151. Epub 2001/07/24. [DOI] [PubMed] [Google Scholar]
  • 228.Crow JP, Sampson JB, Zhuang Y, Thompson JA, Beckman JS. Decreased zinc affinity of amyotrophic lateral sclerosis-associated superoxide dismutase mutants leads to enhanced catalysis of tyrosine nitration by peroxynitrite. J Neurochem. 1997;69(5):1936–1944. doi: 10.1046/j.1471-4159.1997.69051936.x. Epub 1998/02/12. [DOI] [PubMed] [Google Scholar]
  • 229.Kabashi E, Valdmanis PN, Dion P, Rouleau GA. Oxidized/misfolded superoxide dismutase-1: The cause of all amyotrophic lateral sclerosis? Annals of Neurology. 2007;62(6):553–559. doi: 10.1002/ana.21319. [DOI] [PubMed] [Google Scholar]
  • 230.Trumbull KA, Beckman JS. A role for copper in the toxicity of zinc-deficient superoxide dismutase to motor neurons in amyotrophic lateral sclerosis. Antioxid Redox Signal. 2009;11(7):1627–1639. doi: 10.1089/ars.2009.2574. Epub 2009/03/25; PubMed Central PMCID: PMC2842582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Guareschi S, Cova E, Cereda C, Ceroni M, Donetti E, Bosco DA, Trotti D, Pasinelli P. An over-oxidized form of superoxide dismutase found in sporadic amyotrophic lateral sclerosis with bulbar onset shares a toxic mechanism with mutant SOD1. P Natl Acad Sci USA. 2012;109(13):5074–5079. doi: 10.1073/pnas.1115402109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Bosco DA, Morfini G, Karabacak NM, Song Y, Gros-Louis F, Pasinelli P, Goolsby H, Fontaine BA, Lemay N, McKenna-Yasek D, Frosch MP, Agar JN, Julien JP, Brady ST, Brown RH., Jr Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nat Neurosci. 2010;13(11):1396–1403. doi: 10.1038/nn.2660. Epub 2010/10/19; PubMed Central PMCID: PMC2967729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Forsberg K, Jonsson PA, Andersen PM, Bergemalm D, Graffmo KS, Hultdin M, Jacobsson J, Rosquist R, Marklund SL, Brannstrom T. Novel antibodies reveal inclusions containing non-native SOD1 in sporadic ALS patients. PLoS One. 2010;5(7):e11552. doi: 10.1371/journal.pone.0011552. Epub 2010/07/21; PubMed Central PMCID: PMC2904380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Pokrishevsky E, Grad LI, Yousefi M, Wang J, Mackenzie IR, Cashman NR. Aberrant Localization of FUS and TDP43 Is Associated with Misfolding of SOD1 in Amyotrophic Lateral Sclerosis. PLoS One. 2012;7(4) doi: 10.1371/journal.pone.0035050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Liu HN, Sanelli T, Horne P, Pioro EP, Strong MJ, Rogaeva E, Bilbao J, Zinman L, Robertson J. Lack of evidence of monomer/misfolded superoxide dismutase-1 in sporadic amyotrophic lateral sclerosis. Annals of neurology. 2009;66(1):75–80. doi: 10.1002/ana.21704. Epub 2009/08/12. [DOI] [PubMed] [Google Scholar]
  • 236.Aviram M, Rosenblat M, Bisgaier CL, Newton RS, Primo-Parmo SL, La Du BN. Paraoxonase inhibits high-density lipoprotein oxidation and preserves its functions A possible peroxidative role for paraoxonase. J Clin Invest. 1998;101(8):1581–1590. doi: 10.1172/JCI1649. Epub 1998/05/16; PubMed Central PMCID: PMC508738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Costa LG, Cole TB, Furlong CE. Paraoxonase (PON1): from toxicology to cardiovascular medicine. Acta Biomed. 2005;76(Suppl 2):50–57. Epub 2005/12/16. [PubMed] [Google Scholar]
  • 238.Primo-Parmo SL, Sorenson RC, Teiber J, La Du BN. The human serum paraoxonase/arylesterase gene (PON1) is one member of a multigene family. Genomics. 1996;33(3):498–507. doi: 10.1006/geno.1996.0225. Epub 1996/05/01. [DOI] [PubMed] [Google Scholar]
  • 239.Gaidukov L, Tawfik DS. The development of human sera tests for HDL-bound serum PON1 and its lipolactonase activity. Journal of Lipid Research. 2007;48(7):1637–1646. doi: 10.1194/jlr.D600045-JLR200. [DOI] [PubMed] [Google Scholar]
  • 240.Chambers JE. PON1 multitasks to protect health. Proc Natl Acad Sci U S A. 2008;105(35):12639–12640. doi: 10.1073/pnas.0807062105. Epub 2008/08/30; PubMed Central PMCID: PMC2529123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Nguyen SD, Sok DE. Oxidative inactivation of paraoxonase1, an antioxidant protein and its effect on antioxidant action. Free Radic Res. 2003;37(12):1319–1330. Epub 2004/02/03. [PubMed] [Google Scholar]
  • 242.Watson AD, Berliner JA, Hama SY, La Du BN, Faull KF, Fogelman AM, Navab M. Protective effect of high density lipoprotein associated paraoxonase Inhibition of the biological activity of minimally oxidized low density lipoprotein. J Clin Invest. 1995;96(6):2882–2891. doi: 10.1172/JCI118359. Epub 1995/12/01; PubMed Central PMCID: PMC185999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Furlong CE, Richter RJ, Seidel SL, Motulsky AG. Role of genetic polymorphism of human plasma paraoxonase/arylesterase in hydrolysis of the insecticide metabolites chlorpyrifos oxon and paraoxon. Am J Hum Genet. 1988;43(3):230–238. Epub 1988/09/01; PubMed Central PMCID: PMC1715392. [PMC free article] [PubMed] [Google Scholar]
  • 244.Li HL, Liu DP, Liang CC. Paraoxonase gene polymorphisms, oxidative stress, and diseases. J Mol Med (Berl) 2003;81(12):766–779. doi: 10.1007/s00109-003-0481-4. Epub 2003/10/11. [DOI] [PubMed] [Google Scholar]
  • 245.Watson CE, Draganov DI, Billecke SS, Bisgaier CL, La Du BN. Rabbits possess a serum paraoxonase polymorphism similar to the human Q192R. Pharmacogenetics. 2001;11(2):123–134. doi: 10.1097/00008571-200103000-00003. Epub 2001/03/27. [DOI] [PubMed] [Google Scholar]
  • 246.Zhao Y, Ma YS, Fang Y, Liu LL, Wu SD, Fu D, Wang XF. Association between PON1 activity and coronary heart disease risk: A meta-analysis based on 43 studies. Molecular Genetics and Metabolism. 2012;105(1):141–148. doi: 10.1016/j.ymgme.2011.09.018. [DOI] [PubMed] [Google Scholar]
  • 247.Singh S, Kumar V, Thakur S, Banerjee BD, Rautela RS, Grover SS, Rawat DS, Pasha ST, Jain SK, Ichhpujani RL, Rai A. Paraoxonase-1 genetic polymorphisms and susceptibility to DNA damage in workers occupationally exposed to organophosphate pesticides. Toxicol Appl Pharmacol. 2011;252(2):130–137. doi: 10.1016/j.taap.2011.01.014. Epub 2011/02/05. [DOI] [PubMed] [Google Scholar]
  • 248.Draganov DI, La Du BN. Pharmacogenetics of paraoxonases: a brief review. Naunyn Schmiedebergs Arch Pharmacol. 2004;369(1):78–88. doi: 10.1007/s00210-003-0833-1. Epub 2003/10/28. [DOI] [PubMed] [Google Scholar]
  • 249.Zurich MG, Honegger P, Schilter B, Costa LG, Monnet-Tschudi F. Involvement of glial cells in the neurotoxicity of parathion and chlorpyrifos. Toxicol Appl Pharmacol. 2004;201(2):97–104. doi: 10.1016/j.taap.2004.05.003. Epub 2004/11/16. [DOI] [PubMed] [Google Scholar]
  • 250.Precourt LP, Amre D, Denis MC, Lavoie JC, Delvin E, Seidman E, Levy E. The three-gene paraoxonase family: Physiologic roles, actions and regulation. Atherosclerosis. 2011;214(1):20–36. doi: 10.1016/j.atherosclerosis.2010.08.076. [DOI] [PubMed] [Google Scholar]
  • 251.Ng CJ, Wadleigh DJ, Gangopadhyay A, Hama S, Grijalva VR, Navab M, Fogelman AM, Reddy ST. Paraoxonase-2 is a ubiquitously expressed protein with antioxidant properties and is capable of preventing cell-mediated oxidative modification of low density lipoprotein. Journal of Biological Chemistry. 2001;276(48):44444–44449. doi: 10.1074/jbc.M105660200. [DOI] [PubMed] [Google Scholar]
  • 252.Reddy ST, Wadleigh DJ, Grijalva V, Ng C, Hama S, Gangopadhyay A, Shih DM, Lusis AJ, Navab M, Fogelman AM. Human paraoxonase-3 is an HDL-associated enzyme with biological activity similar to paraoxonase-1 protein but is not regulated by oxidized lipids. Arterioscl Throm Vas. 2001;21(4):542–547. doi: 10.1161/01.atv.21.4.542. [DOI] [PubMed] [Google Scholar]
  • 253.Costa LG, Vitalone A, Cole TB, Furlong CE. Modulation of paraoxonase (PON1) activity. Biochem Pharmacol. 2005;69(4):541–550. doi: 10.1016/j.bcp.2004.08.027. Epub 2005/01/27. [DOI] [PubMed] [Google Scholar]
  • 254.Richter RJ, Furlong CE. Determination of paraoxonase (PON1) status requires more than genotyping. Pharmacogenetics. 1999;9(6):745–753. [PubMed] [Google Scholar]
  • 255.Ferre N, Camps J, Fernandez-Ballart J, Arija V, Murphy MM, Ceruelo S, Biarnes E, Vilella E, Tous M, Joven J. Regulation of serum paraoxonase activity by genetic, nutritional, and lifestyle factors in the general population. Clin Chem. 2003;49(9):1491–1497. doi: 10.1373/49.9.1491. Epub 2003/08/21. [DOI] [PubMed] [Google Scholar]
  • 256.Aviram M, Rosenblat M, Bisgaier CL, Newton RS. Atorvastatin and gemfibrozil metabolites, but not the parent drugs, are potent antioxidants against lipoprotein oxidation. Atherosclerosis. 1998;138(2):271–280. doi: 10.1016/s0021-9150(98)00032-x. Epub 1998/08/05. [DOI] [PubMed] [Google Scholar]
  • 257.Deakin S, Guernier S, James RW. Pharmacogenetic interaction between paraoxonase-1 gene promoter polymorphism C-107T and statin. Pharmacogenet Genom. 2007;17(6):451–457. doi: 10.1097/FPC.0b013e3280925716. [DOI] [PubMed] [Google Scholar]
  • 258.Blatter-Garin MC, Kalix B, De Pree S, James RW. Aspirin use is associated with higher serum concentrations of the anti-oxidant enzyme, paraoxonase-1. Diabetologia. 2003;46(4):593–594. doi: 10.1007/s00125-003-1065-0. [DOI] [PubMed] [Google Scholar]
  • 259.Slowik A, Tomik B, Wolkow PP, Partyka D, Turaj W, Malecki MT, Pera J, Dziedzic T, Szczudlik A, Figlewicz DA. Paraoxonase gene polymorphisms and sporadic ALS. Neurology. 2006;67(5):766–770. doi: 10.1212/01.wnl.0000219565.32247.11. Epub 2006/07/11. [DOI] [PubMed] [Google Scholar]
  • 260.Saeed M, Siddique N, Hung WY, Usacheva E, Liu E, Sufit RL, Heller SL, Haines JL, Pericak-Vance M, Siddique T. Paraoxonase cluster polymorphisms are associated with sporadic ALS. Neurology. 2006;67(5):771–776. doi: 10.1212/01.wnl.0000227187.52002.88. Epub 2006/07/11. [DOI] [PubMed] [Google Scholar]
  • 261.Valdmanis PN, Kabashi E, Dyck A, Hince P, Lee J, Dion P, D'Amour M, Souchon F, Bouchard JP, Salachas F, Meininger V, Andersen PM, Camu W, Dupre N, Rouleau GA. Association of paraoxonase gene cluster polymorphisms with ALS in France, Quebec, and Sweden. Neurology. 2008;71(7):514–520. doi: 10.1212/01.wnl.0000324997.21272.0c. Epub 2008/08/13. [DOI] [PubMed] [Google Scholar]
  • 262.Cronin S, Greenway MJ, Prehn JH, Hardiman O. Paraoxonase promoter and intronic variants modify risk of sporadic amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry. 2007;78(9):984–986. doi: 10.1136/jnnp.2006.112581. Epub 2007/08/21; PubMed Central PMCID: PMC2117866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Morahan JM, Yu B, Trent RJ, Pamphlett R. A gene-environment study of the paraoxonase 1 gene and pesticides in amyotrophic lateral sclerosis. Neurotoxicology. 2007;28(3):532–540. doi: 10.1016/j.neuro.2006.11.007. Epub 2007/01/06. [DOI] [PubMed] [Google Scholar]
  • 264.Landers JE, Shi L, Cho TJ, Glass JD, Shaw CE, Leigh PN, Diekstra F, Polak M, Rodriguez-Leyva I, Niemann S, Traynor BJ, McKenna-Yasek D, Sapp PC, Al-Chalabi A, Wills AMA, Brown RH. A common haplotype within the PON1 promoter region is associated with sporadic ALS. Amyotroph Lateral Sc. 2008;9(5):306–314. doi: 10.1080/17482960802233177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Zawislak D, Ostrowska M, Golenia A, Marona M, Tomik B, Wolkow P, Gryz-Kurek E, Szczudlik A, Slowik A. The -A162G polymorphism of the PON1 gene and the risk of sporadic amyotrophic lateral sclerosis. Neurologia i neurochirurgia polska. 2010;44(3):246–250. doi: 10.1016/s0028-3843(14)60038-4. Epub 2010/07/14. [DOI] [PubMed] [Google Scholar]
  • 266.Ricci C, Battistini S, Cozzi L, Benigni M, Origone P, Verriello L, Lunetta C, Cereda C, Milani P, Greco G, Patrosso MC, Causarano R, Caponnetto C, Giannini F, Corbo M, Penco S. Lack of association of PON polymorphisms with sporadic ALS in an Italian population. Neurobiol Aging. 2011;32(3):552. doi: 10.1016/j.neurobiolaging.2010.02.010. e7–13 Epub 2010/04/13. [DOI] [PubMed] [Google Scholar]
  • 267.Ticozzi N, LeClerc AL, Keagle PJ, Glass JD, Wills AM, van Blitterswijk M, Bosco DA, Rodriguez-Leyva I, Gellera C, Ratti A, Taroni F, McKenna-Yasek D, Sapp PC, Silani V, Furlong CE, Brown RH, Jr, Landers JE. Paraoxonase gene mutations in amyotrophic lateral sclerosis. Annals of neurology. 2010;68(1):102–107. doi: 10.1002/ana.21993. Epub 2010/06/29; PubMed Central PMCID: PMC2945725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.van Blitterswijk M, Blokhuis A, van Es MA, van Vught PW, Rowicka PA, Schelhaas HJ, van der Kooi AJ, de Visser M, Veldink JH, van den Berg LH. Rare and common paraoxonase gene variants in amyotrophic lateral sclerosis patients. Neurobiol Aging. 2012 doi: 10.1016/j.neurobiolaging.2012.01.007. Epub 2012/02/15. [DOI] [PubMed] [Google Scholar]
  • 269.Wills AM, Cronin S, Slowik A, Kasperaviciute D, Van Es MA, Morahan JM, Valdmanis PN, Meininger V, Melki J, Shaw CE, Rouleau GA, Fisher EM, Shaw PJ, Morrison KE, Pamphlett R, Van den Berg LH, Figlewicz DA, Andersen PM, Al-Chalabi A, Hardiman O, Purcell S, Landers JE, Brown RH., Jr A large-scale international meta-analysis of paraoxonase gene polymorphisms in sporadic ALS. Neurology. 2009;73(1):16–24. doi: 10.1212/WNL.0b013e3181a18674. Epub 2009/03/27; PubMed Central PMCID: PMC2707108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Costa LG, Giordano G, Furlong CE. Pharmacological and dietary modulators of paraoxonase 1 (PON1) activity and expression: the hunt goes on. Biochem Pharmacol. 2011;81(3):337–344. doi: 10.1016/j.bcp.2010.11.008. Epub 2010/11/26; PubMed Central PMCID: PMC3077125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Drory VE, Birnbaum M, Korczyn AD, Chapman J. Association of APOE epsilon4 allele with survival in amyotrophic lateral sclerosis. J Neurol Sci. 2001;190(1–2):17–20. doi: 10.1016/s0022-510x(01)00569-x. Epub 2001/09/28. [DOI] [PubMed] [Google Scholar]
  • 272.Moulard B, Sefiani A, Laamri A, Malafosse A, Camu W. Apolipoprotein E genotyping in sporadic amyotrophic lateral sclerosis: evidence for a major influence on the clinical presentation and prognosis. J Neurol Sci. 1996;139(Suppl):34–37. doi: 10.1016/0022-510x(96)00085-8. Epub 1996/08/01. [DOI] [PubMed] [Google Scholar]
  • 273.Zetterberg H, Jacobsson J, Rosengren L, Blennow K, Andersen PM. Association of APOE with age at onset of sporadic amyotrophic lateral sclerosis. Journal of the Neurological Sciences. 2008;273(1–2):67–69. doi: 10.1016/j.jns.2008.06.025. [DOI] [PubMed] [Google Scholar]
  • 274.Jawaid A, Poon M, Strutt AM, Rice LK, McDowell EJ, Salamone AR, Qureshi SU, Simpson E, Appel SH, York MK, Schulz PE. Does apolipoprotein E genotype modify the clinical expression of ALS? European Journal of Neurology. 2011;18(4):618–624. doi: 10.1111/j.1468-1331.2010.03225.x. [DOI] [PubMed] [Google Scholar]
  • 275.Mahley RW, Rall SC. Apolipoprotein E: Far more than a lipid transport protein. Annu Rev Genom Hum G. 2000;1:507–537. doi: 10.1146/annurev.genom.1.1.507. [DOI] [PubMed] [Google Scholar]
  • 276.Bedlack RS, Strittmatter WJ, Morgenlander JC. Apolipoprotein E and neuromuscular disease: a critical review of the literature. Arch Neurol. 2000;57(11):1561–1465. doi: 10.1001/archneur.57.11.1561. Epub 2000/11/14. [DOI] [PubMed] [Google Scholar]
  • 277.Sabo T, Lomnitski L, Nyska A, Beni S, Maronpot RR, Shohami E, Roses AD, Michaelson DM. Susceptibility of transgenic mice expressing human apolipoprotein E to closed head injury: The allele E3 is neuroprotective whereas E4 increases fatalities. Neuroscience. 2000;101(4):879–884. doi: 10.1016/s0306-4522(00)00438-3. [DOI] [PubMed] [Google Scholar]
  • 278.Tayler H, Fraser T, Miners JS, Kehoe PG, Love S. Oxidative Balance in Alzheimer's Disease: Relationship to APOE, Braak Tangle Stage, and the Concentrations of Soluble and Insoluble Amyloid-beta. J Alzheimers Dis. 2010;22(4):1363–1373. doi: 10.3233/JAD-2010-101368. [DOI] [PubMed] [Google Scholar]
  • 279.Sathasivam S. VEGF and ALS. Neurosci Res. 2008;62(2):71–77. doi: 10.1016/j.neures.2008.06.008. Epub 2008/07/29. [DOI] [PubMed] [Google Scholar]
  • 280.Blauw HM, Barnes CP, van Vught PW, van Rheenen W, Verheul M, Cuppen E, Veldink JH, van den Berg LH. SMN1 gene duplications are associated with sporadic ALS. Neurology. 2012 doi: 10.1212/WNL.0b013e318249f697. Epub 2012/02/11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.van Es MA, Schelhaas HJ, van Vught PW, Ticozzi N, Andersen PM, Groen EJ, Schulte C, Blauw HM, Koppers M, Diekstra FP, Fumoto K, LeClerc AL, Keagle P, Bloem BR, Scheffer H, van Nuenen BF, van Blitterswijk M, van Rheenen W, Wills AM, Lowe PP, Hu GF, Yu W, Kishikawa H, Wu D, Folkerth RD, Mariani C, Goldwurm S, Pezzoli G, Van Damme P, Lemmens R, Dahlberg C, Birve A, Fernandez-Santiago R, Waibel S, Klein C, Weber M, van der Kooi AJ, de Visser M, Verbaan D, van Hilten JJ, Heutink P, Hennekam EA, Cuppen E, Berg D, Brown RH, Jr, Silani V, Gasser T, Ludolph AC, Robberecht W, Ophoff RA, Veldink JH, Pasterkamp RJ, de Bakker PI, Landers JE, van de Warrenburg BP, van den Berg LH. Angiogenin variants in Parkinson disease and amyotrophic lateral sclerosis. Annals of neurology. 2011;70(6):964–973. doi: 10.1002/ana.22611. Epub 2011/12/23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Zou T, Yang X, Pan D, Huang J, Sahin M, Zhou J. SMN deficiency reduces cellular ability to form stress granules, sensitizing cells to stress. Cell Mol Neurobiol. 2011;31(4):541–550. doi: 10.1007/s10571-011-9647-8. Epub 2011/01/15. [DOI] [PubMed] [Google Scholar]
  • 283.van Es MA, Schelhaas HJ, van Vught PWJ, Ticozzi N, Andersen PM, Groen EJN, Schulte C, Blauw HM, Koppers M, Diekstra FP, Fumoto K, LeClerc AL, Keagle P, Bloem BR, Scheffer H, van Nuenen BFL, van Blitterswijk M, van Rheenen W, Wills AM, Lowe PP, Hu GF, Yu WH, Kishikawa H, Wu D, Folkerth RD, Mariani C, Goldwurm S, Pezzoli G, Van Damme P, Lemmens R, Dahlberg C, Birve A, Fernandez-Santiago R, Waibel S, Klein C, Weber M, van der Kooi AJ, de Visser M, Verbaan D, van Hilten JJ, Heutink P, Hennekam EAM, Cuppen E, Berg D, Brown RH, Silani V, Gasser T, Ludolph AC, Robberecht W, Ophoff RA, Veldink JH, Pasterkamp RJ, de Bakker PIW, Landers JE, van de Warrenburg BP, van den Berg LH. Angiogenin variants in Parkinson disease and amyotrophic lateral sclerosis. Annals of Neurology. 2011;70(6):964–973. doi: 10.1002/ana.22611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Li S, Hu GF. Angiogenin-mediated rRNA transcription in cancer and neurodegeneration. Int J Biochem Mol Biol. 2010;1(1):26–35. Epub 2010/09/10; PubMed Central PMCID: PMC2933922. [PMC free article] [PubMed] [Google Scholar]
  • 285.Emara MM, Ivanov P, Hickman T, Dawra N, Tisdale S, Kedersha N, Hu GF, Anderson P. Angiogenin-induced tRNA-derived stress-induced RNAs promote stress-induced stress granule assembly. J Biol Chem. 2010;285(14):10959–10968. doi: 10.1074/jbc.M109.077560. Epub 2010/02/05; PubMed Central PMCID: PMC2856301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Jomova K, Valko M. Advances in metal-induced oxidative stress and human disease. Toxicology. 2011;283(2–3):65–87. doi: 10.1016/j.tox.2011.03.001. [DOI] [PubMed] [Google Scholar]
  • 287.Nandar W, Connor JR. HFE gene variants affect iron in the brain. J Nutr. 2011;141(4):729S–739S. doi: 10.3945/jn.110.130351. Epub 2011/02/25. [DOI] [PubMed] [Google Scholar]
  • 288.Blasco H, Vourc'h P, Nadjar Y, Ribourtout B, Gordon PH, Guettard YO, Camu W, Praline J, Meininger V, Andres CR, Corcia P, Grp FAS. Association between divalent metal transport 1 encoding gene (SLC11A2) and disease duration in amyotrophic lateral sclerosis. Journal of the Neurological Sciences. 2011;303(1–2):124–127. doi: 10.1016/j.jns.2010.12.018. [DOI] [PubMed] [Google Scholar]
  • 289.Kagi JH, Kojima Y. Chemistry and biochemistry of metallothionein. Experientia Suppl. 1987;52:25–61. doi: 10.1007/978-3-0348-6784-9_3. Epub 1987/01/01. [DOI] [PubMed] [Google Scholar]
  • 290.Morahan JM, Yu B, Trent RJ, Pamphlett R. Are metallothionein genes silenced in ALS? Toxicol Lett. 2007;168(1):83–87. doi: 10.1016/j.toxlet.2006.11.003. Epub 2006/12/13. [DOI] [PubMed] [Google Scholar]
  • 291.Elliott NA, Volkert MR. Stress induction and mitochondrial localization of Oxr1 proteins in yeast and humans. Mol Cell Biol. 2004;24(8):3180–3187. doi: 10.1128/MCB.24.8.3180-3187.2004. Epub 2004/04/03; PubMed Central PMCID: PMC381681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Oliver PL, Finelli MJ, Edwards B, Bitoun E, Butts DL, Becker EB, Cheeseman MT, Davies B, Davies KE. Oxr1 is essential for protection against oxidative stress-induced neurodegeneration. PLoS Genet. 2011;7(10):e1002338. doi: 10.1371/journal.pgen.1002338. Epub 2011/10/27; PubMed Central PMCID: PMC3197693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Atkin JD, Farg MA, Walker AK, McLean C, Tomas D, Horne MK. Endoplasmic reticulum stress and induction of the unfolded protein response in human sporadic amyotrophic lateral sclerosis. Neurobiology of Disease. 2008;30(3):400–407. doi: 10.1016/j.nbd.2008.02.009. [DOI] [PubMed] [Google Scholar]
  • 294.Nishimura AL, Mitne-Neto M, Silva HCA, Richieri-Costa A, Middleton S, Cascio D, Kok F, Oliveira JRM, Gillingwater T, Webb J, Skehel P, Zatz M. A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. American Journal of Human Genetics. 2004;75(5):822–831. doi: 10.1086/425287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding JH, McCluskey L, Martinez-Lage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein J, Landi F, Wang YD, Calvo A, Mora G, Sabatelli M, Monsurro MR, Battistini S, Salvi F, Spataro R, Sola P, Borghero G, Galassi G, Scholz SW, Taylor JP, Restagno G, Chio A, Traynor BJ, Consortium I. Exome Sequencing Reveals VCP Mutations as a Cause of Familial ALS. Neuron. 2010;68(5):857–864. doi: 10.1016/j.neuron.2010.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Ilieva EV, Ayala V, Jove M, Dalfo E, Cacabelos D, Povedano M, Bellmunt MJ, Ferrer I, Pamplona R, Portero-Otin M. Oxidative and endoplasmic reticulum stress interplay in sporadic amyotrophic lateral sclerosis. Brain. 2007;130:3111–3123. doi: 10.1093/brain/awm190. [DOI] [PubMed] [Google Scholar]
  • 297.Malhotra JD, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress: a vicious cycle or a double-edged sword? Antioxid Redox Signal. 2007;9(12):2277–2293. doi: 10.1089/ars.2007.1782. Epub 2007/11/06. [DOI] [PubMed] [Google Scholar]
  • 298.Murray B. Natural History and Prognosis in Amyotrophic Lateral Sclerosis. In: Mitsumoto HPS, Gordon PH, editors. Epidemiology of ALS. New York: Taylor Francis Group; 2006. pp. 227–255. [Google Scholar]
  • 299.Evans WJ, Morley JE, Argiles J, Bales C, Baracos V, Guttridge D, Jatoi A, Kalantar-Zadeh K, Lochs H, Mantovani G, Marks D, Mitch WE, Muscaritoli M, Najand A, Ponikowski P, Rossi Fanelli F, Schambelan M, Schols A, Schuster M, Thomas D, Wolfe R, Anker SD. Cachexia: a new definition. Clin Nutr. 2008;27(6):793–799. doi: 10.1016/j.clnu.2008.06.013. Epub 2008/08/23. [DOI] [PubMed] [Google Scholar]
  • 300.Laviano A, Meguid MM, Preziosa I, Rossi Fanelli F. Oxidative stress and wasting in cancer. Curr Opin Clin Nutr Metab Care. 2007;10(4):449–456. doi: 10.1097/MCO.0b013e328122db94. Epub 2007/06/15. [DOI] [PubMed] [Google Scholar]
  • 301.Norris FH, Denys EH, Sang K. ALS CACHEXIA. In: Tsubaki T, Toyokura Y, editors. Amyotroph Lateral Sc. BALTIMORE: UNIVERSITY PARK PRESS; 1979. pp. 17–19. [Google Scholar]
  • 302.Desport JC, Preux PM, Truong TC, Vallat JM, Sautereau D, Couratier P. Nutritional status is a prognostic factor for survival in ALS patients. Neurology. 1999;53(5):1059–1063. doi: 10.1212/wnl.53.5.1059. [DOI] [PubMed] [Google Scholar]
  • 303.Bouteloup C, Desport JC, Clavelou P, Guy N, Derumeaux-Burel H, Ferrier A, Couratier P. Hypermetabolism in ALS patients: an early and persistent phenomenon. J Neurol. 2009;256(8):1236–1242. doi: 10.1007/s00415-009-5100-z. Epub 2009/03/24. [DOI] [PubMed] [Google Scholar]
  • 304.Akinola FF, Oguntibeju OO, Alabi OO. Effects of severe malnutrition on oxidative stress in Wistar rats. Sci Res Essays. 2010;5(10):1145–1149. [Google Scholar]
  • 305.Kalantar-Zadeh K, Ikizler TA, Block G, Avram MM, Kopple JD. Malnutrition-inflammation complex syndrome in dialysis patients: causes and consequences. Am J Kidney Dis. 2003;42(5):864–881. doi: 10.1016/j.ajkd.2003.07.016. Epub 2003/10/29. [DOI] [PubMed] [Google Scholar]
  • 306.Morena M, Canaud B, Terrier N, Canaud L, Cristol J-P. Oxidative stress complex syndrome: The dark side of the malnutrition-inflammation complex syndrome. Hemodial Int. 2007;11:S32–S38. [Google Scholar]
  • 307.Miller RG, Jackson CE, Kasarskis EJ, England JD, Forshew D, Johnston W, Kalra S, Katz JS, Mitsumoto H, Rosenfeld J, Shoesmith C, Strong MJ, Woolley SC. Quality Standards Subcommittee of the American Academy of N Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: drug, nutritional, and respiratory therapies (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2009;73(15):1218–1226. doi: 10.1212/WNL.0b013e3181bc0141. Epub 2009/10/14; PubMed Central PMCID: PMC2764727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Pialoux V, Hanly PJ, Foster GE, Brugniaux JV, Beaudin AE, Hartmann SE, Pun M, Duggan CT, Poulin MJ. Effects of exposure to intermittent hypoxia on oxidative stress and acute hypoxic ventilatory response in humans. Am J Respir Crit Care Med. 2009;180(10):1002–1009. doi: 10.1164/rccm.200905-0671OC. Epub 2009/08/29. [DOI] [PubMed] [Google Scholar]
  • 309.Hornbein TF. The high-altitude brain. J Exp Biol. 2001;204(Pt 18):3129–3132. doi: 10.1242/jeb.204.18.3129. Epub 2001/10/03. [DOI] [PubMed] [Google Scholar]
  • 310.Acker T, Acker H. Cellular oxygen sensing need in CNS function: physiological and pathological implications. The Journal of experimental biology. 2004;207(Pt 18):3171–3188. doi: 10.1242/jeb.01075. Epub 2004/08/10. [DOI] [PubMed] [Google Scholar]
  • 311.Lenigerfollert E, Lubbers DW, Wrabetz W. Regulation of Local Tissue Po2 of Brain Cortex at Different Arterial O2 Pressures. Pflug Arch Eur J Phy. 1975;359(1–2):81–95. doi: 10.1007/BF00581279. [DOI] [PubMed] [Google Scholar]
  • 312.Schild L, Reiser G. Oxidative stress is involved in the permeabilization of the inner membrane of brain mitochondria exposed to hypoxia/reoxygenation and low micromolar Ca2+ FEBS J. 2005;272(14):3593–3601. doi: 10.1111/j.1742-4658.2005.04781.x. Epub 2005/07/13. [DOI] [PubMed] [Google Scholar]
  • 313.Bolanos JP, Almeida A. Roles of nitric oxide in brain hypoxia-ischemia. Bba-Bioenergetics. 1999;1411(2–3):415–436. doi: 10.1016/s0005-2728(99)00030-4. [DOI] [PubMed] [Google Scholar]
  • 314.McElhiney MC, Rabkin JG, Gordon PH, Goetz R, Mitsumoto H. Prevalence of fatigue and depression in ALS patients and change over time. J Neurol Neurosurg Psychiatry. 2009;80(10):1146–1149. doi: 10.1136/jnnp.2008.163246. Epub 2009/09/19. [DOI] [PubMed] [Google Scholar]
  • 315.McDonald ER, Wiedenfeld SA, Hillel A, Carpenter CL, Walter RA. Survival in amyotrophic lateral sclerosis The role of psychological factors. Arch Neurol. 1994;51(1):17–23. doi: 10.1001/archneur.1994.00540130027010. Epub 1994/01/01. [DOI] [PubMed] [Google Scholar]
  • 316.Johnston M, Earll L, Giles M, McClenahan R, Stevens D, Morrison V. Mood as a predictor of disability and survival in patients newly diagnosed with ALS MND. Brit J Health Psych. 1999;4:127–136. [Google Scholar]
  • 317.Epel ES, Blackburn EH, Lin J, Dhabhar FS, Adler NE, Morrow JD, Cawthon RM. Accelerated telomere shortening in response to life stress. Proc Natl Acad Sci U S A. 2004;101(49):17312–17315. doi: 10.1073/pnas.0407162101. Epub 2004/12/03; PubMed Central PMCID: PMC534658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Sklan EH, Lowenthal A, Korner M, Ritov Y, Landers DM, Rankinen T, Bouchard C, Leon AS, Rice T, Rao DC, Wilmore JH, Skinner JS, Soreq H. Acetylcholinesterase/paraoxonase genotype and expression predict anxiety scores in health, risk factors, exercise training, and genetics study. P Natl Acad Sci USA. 2004;101(15):5512–5517. doi: 10.1073/pnas.0307659101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Hayley S, Poulter MO, Merali Z, Anisman H. The pathogenesis of clinical depression: Stressor- and cytokine-induced alterations of neuroplasticity. Neuroscience. 2005;135(3):659–678. doi: 10.1016/j.neuroscience.2005.03.051. [DOI] [PubMed] [Google Scholar]
  • 320.Mitsumoto H, Hanson MR, Chad DA. Amyotrophic lateral sclerosis Recent advances in pathogenesis and therapeutic trials. Arch Neurol. 1988;45(2):189–202. doi: 10.1001/archneur.1988.00520260077025. Epub 1988/02/01. [DOI] [PubMed] [Google Scholar]
  • 321.Appel SH. Is ALS a systemic disorder? Evidence from muscle mitochondria. Experimental Neurology. 2006;198(1):1–3. doi: 10.1016/j.expneurol.2005.12.025. [DOI] [PubMed] [Google Scholar]
  • 322.Ono S. The skin in amyotrophic lateral sclerosis. Amyotrophic lateral sclerosis and other motor neuron disorders : official publication of the World Federation of Neurology, Research Group on Motor Neuron Diseases. 2000;1(3):191–199. doi: 10.1080/14660820050515188. Epub 2001/07/24. [DOI] [PubMed] [Google Scholar]
  • 323.Zhang R, Miller RG, Gascon R, Champion S, Katz J, Lancero M, Narvaez A, Honrada R, Ruvalcaba D, McGrath MS. Circulating endotoxin and systemic immune activation in sporadic amyotrophic lateral sclerosis (sALS) J Neuroimmunol. 2009;206(1–2):121–124. doi: 10.1016/j.jneuroim.2008.09.017. Epub 2008/11/18; PubMed Central PMCID: PMC2995297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Zhang RZ, Gascon R, Miller RG, Gelinas DF, Mass J, Hadlock K, Jin X, Reis J, Narvaez A, McGrath MS. Evidence for systemic immune system alterations in sporadic amyotrophic lateral sclerosis (sALS) Journal of Neuroimmunology. 2005;159(1–2):215–224. doi: 10.1016/j.jneuroim.2004.10.009. [DOI] [PubMed] [Google Scholar]
  • 325.Kolde G, Bachus R, Ludolph AC. Skin involvement in amyotrophic lateral sclerosis. Lancet. 1996;347(9010):1226–1227. doi: 10.1016/s0140-6736(96)90737-0. [DOI] [PubMed] [Google Scholar]
  • 326.Zhang RZ, Gascon R, Miller RG, Gelinas DF, Mass J, Lancero M, Narvaez A, McGrath MS. MCP-1 chemokine receptor CCR2 is decreased on circulating monocytes in sporadic amyotrophic lateral sclerosis (sALS) Journal of Neuroimmunology. 2006;179(1–2):87–93. doi: 10.1016/j.jneuroim.2006.06.008. [DOI] [PubMed] [Google Scholar]
  • 327.Ono S, Hu J, Shimizu N, Imai T, Nakagawa H. Increased interleukin-6 of skin and serum in amyotrophic lateral sclerosis. J Neurol Sci. 2001;187(1–2):27–34. doi: 10.1016/s0022-510x(01)00514-7. Epub 2001/07/07. [DOI] [PubMed] [Google Scholar]
  • 328.Babu GN, Kumar A, Chandra R, Puri SK, Kalita J, Misra UK. Elevated inflammatory markers in a group of amyotrophic lateral sclerosis patients from Northern India. Neurochemical Research. 2008;33(6):1145–1149. doi: 10.1007/s11064-007-9564-x. [DOI] [PubMed] [Google Scholar]
  • 329.Dupuis L, Corcia P, Fergani A, De Aguilar JLG, Bonnefont-Rousselot D, Bittar R, Seilhean D, Hauw JJ, Lacomblez L, Loeffler JP, Meininger V. Dyslipidemia is a protective factor in amyotrophic lateral sclerosis. Neurology. 2008;70(13):1004–1009. doi: 10.1212/01.wnl.0000285080.70324.27. [DOI] [PubMed] [Google Scholar]
  • 330.Chio A, Calvo A, Ilardi A, Cavallo E, Moglia C, Mutani R, Palmo A, Galletti R, Marinou K, Papetti L, Mora G. Lower serum lipid levels are related to respiratory impairment in patients with ALS. Neurology. 2009;73(20):1681–1685. doi: 10.1212/WNL.0b013e3181c1df1e. Epub 2009/11/18. [DOI] [PubMed] [Google Scholar]
  • 331.Dorst J, Kuhnlein P, Hendrich C, Kassubek J, Sperfeld AD, Ludolph AC. Patients with elevated triglyceride and cholesterol serum levels have a prolonged survival in amyotrophic lateral sclerosis. Journal of Neurology. 2011;258(4):613–617. doi: 10.1007/s00415-010-5805-z. [DOI] [PubMed] [Google Scholar]
  • 332.Paganoni S, Deng J, Jaffa M, Cudkowicz ME, Wills AM. Body Mass Index, Not Dyslipidemia, Is an Independent Predictor of Survival in Amyotrophic Lateral Sclerosis. Muscle & Nerve. 2011;44(1):20–24. doi: 10.1002/mus.22114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Dedic SIK, Stevic Z, Dedic V, Stojanovic VR, Milicev M, Lavrnic D. Is hyperlipidemia correlated with longer survival in patients with amyotrophic lateral sclerosis? Neurological Research. 2012;34(6):576–580. doi: 10.1179/1743132812Y.0000000049. [DOI] [PubMed] [Google Scholar]
  • 334.Sutedja NA, van der Schouw YT, Fischer K, Sizoo EM, Huisman MHB, Veldink JH, Van den Berg LH. Beneficial vascular risk profile is associated with amyotrophic lateral sclerosis. J Neurol Neurosur Ps. 2011;82(6):638–642. doi: 10.1136/jnnp.2010.236752. [DOI] [PubMed] [Google Scholar]
  • 335.Aleman A, Muller M, de Haan EH, van der Schouw YT. Vascular risk factors and cognitive function in a sample of independently living men. Neurobiol Aging. 2005;26(4):485–490. doi: 10.1016/j.neurobiolaging.2004.05.005. Epub 2005/01/18. [DOI] [PubMed] [Google Scholar]
  • 336.Boker LK, van Noord PA, van der Schouw YT, Koot NV, Bueno de Mesquita HB, Riboli E, Grobbee DE, Peeters PH. Prospect-EPIC Utrecht: study design characteristics of the cohort population European Prospective Investigation into Cancer and Nutrition. Eur J Epidemiol. 2001;17(11):1047–1053. doi: 10.1023/a:1020009325797. Epub 2002/10/17. [DOI] [PubMed] [Google Scholar]
  • 337.Krasnianski A, Deschauer M, Neudecker S, Gellerich FN, Muller T, Schoser BG, Krasnianski M, Zierz S. Mitochondrial changes in skeletal muscle in amyotrophic lateral sclerosis and other neurogenic atrophies. Brain. 2005;128(Pt 8):1870–1876. doi: 10.1093/brain/awh540. Epub 2005/05/20. [DOI] [PubMed] [Google Scholar]
  • 338.Crugnola V, Lamperti C, Lucchini V, Ronchi D, Peverelli L, Prelle A, Sciacco M, Bordoni A, Fassone E, Fortunato F, Corti S, Silani V, Bresolin N, Di Mauro S, Comi GP, Moggio M. Mitochondrial respiratory chain dysfunction in muscle from patients with amyotrophic lateral sclerosis. Archives of neurology. 2010;67(7):849–854. doi: 10.1001/archneurol.2010.128. Epub 2010/07/14. [DOI] [PubMed] [Google Scholar]
  • 339.Duffy LM, Chapman AL, Shaw PJ, Grierson AJ. Review: The role of mitochondria in the pathogenesis of amyotrophic lateral sclerosis. Neuropathology and applied neurobiology. 2011;37(4):336–352. doi: 10.1111/j.1365-2990.2011.01166.x. Epub 2011/02/09. [DOI] [PubMed] [Google Scholar]
  • 340.Dupuis L, Loeffler JP. Neuromuscular junction destruction during amyotrophic lateral sclerosis: insights from transgenic models. Curr Opin Pharmacol. 2009;9(3):341–346. doi: 10.1016/j.coph.2009.03.007. Epub 2009/04/24. [DOI] [PubMed] [Google Scholar]
  • 341.Muller FL, Song W, Jang YC, Liu Y, Sabia M, Richardson A, Van Remmen H. Denervation-induced skeletal muscle atrophy is associated with increased mitochondrial ROS production. Am J Physiol-Reg I. 2007;293(3):R1159–R1168. doi: 10.1152/ajpregu.00767.2006. [DOI] [PubMed] [Google Scholar]
  • 342.Watanabe T, Okeda Y, Yamano T, Ono S. An immunohistochemical study of ubiquitin in the skin of sporadic amyotrophic lateral sclerosis. J Neurol Sci. 2010;298(1–2):52–56. doi: 10.1016/j.jns.2010.08.026. [DOI] [PubMed] [Google Scholar]
  • 343.Yasui K, Oketa Y, Higashida K, Fukazawa H, Ono S. Increased progranulin in the skin of amyotrophic lateral sclerosis: an immunohistochemical study. J Neurol Sci. 2011;309(1–2):110–114. doi: 10.1016/j.jns.2011.07.003. Epub 2011/08/02. [DOI] [PubMed] [Google Scholar]
  • 344.Fang L, Huber-Abel F, Teuchert M, Hendrich C, Dorst J, Schattauer D, Zettlmeissel H, Wlaschek M, Scharffetter-Kochanek K, Tumani H, Ludolph AC, Brettschneider J. Linking neuron and skin: matrix metalloproteinases in amyotrophic lateral sclerosis (ALS) J Neurol Sci. 2009;285(1–2):62–66. doi: 10.1016/j.jns.2009.05.025. Epub 2009/06/16. [DOI] [PubMed] [Google Scholar]
  • 345.Arbesman H, Mackay-Wiggan JM, Liu XH, Andrews H, Arbesman M, Andrews J, Marcus R, Hoch A, Coppola C, Harrington-Money G, Feng M, Martinez F, Bickers D, Mitsumoto H. In Vivo Measurement of the Elastic Properties of the Skin as a Biomarker for Amyotrophic Lateral Sclerosis. Neurology. 2009;72(11):A417–A417. [Google Scholar]
  • 346.Ishikawa H, Yasui K, Oketa Y, Suzuki M, Ono S. Increased expression of valosin-containing protein in the skin of patients with amyotrophic lateral sclerosis. Journal of Clinical Neuroscience. 2012;19(4):522–526. doi: 10.1016/j.jocn.2011.05.044. [DOI] [PubMed] [Google Scholar]
  • 347.Aguirre T, Van Den Bosch L, Goetschalckx K, Tilkin P, Mathijs G, Cassiman JJ, Robberecht W. Increased sensitivity of fibroblasts from amyotrophic lateral sclerosis patients to oxidative stress. Ann Neurol. 1998;43(4):452–457. doi: 10.1002/ana.410430407. Epub 1998/04/18. [DOI] [PubMed] [Google Scholar]
  • 348.Nakano Y, Hirayama K, Terao K. Hepatic ultrastructural changes and liver dysfunction in amyotrophic lateral sclerosis. Arch Neurol. 1987;44(1):103–106. doi: 10.1001/archneur.1987.00520130079022. Epub 1987/01/01. [DOI] [PubMed] [Google Scholar]
  • 349.Mitsumoto H. Biochemical markers: con. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002;3(Suppl 1):S83. doi: 10.1080/146608202320374390. Epub 2002/10/25. [DOI] [PubMed] [Google Scholar]
  • 350.Pedersen WA, Chan SL, Mattson MP. A mechanism for the neuroprotective effect of apolipoprotein E: isoform-specific modification by the lipid peroxidation product 4-hydroxynonenal. J Neurochem. 2000;74(4):1426–1433. doi: 10.1046/j.1471-4159.2000.0741426.x. Epub 2000/03/29. [DOI] [PubMed] [Google Scholar]
  • 351.Chio A. Risk factors in the early diagnosis of ALS: European epidemiological studies. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1(Suppl 1):S13–S18. doi: 10.1080/14660820052415862. Epub 2001/07/24. [DOI] [PubMed] [Google Scholar]
  • 352.Armon C. Environmental risk factors for amyotrophic lateral sclerosis. Neuroepidemiology. 2001;20(1):2–6. doi: 10.1159/000054751. Epub 2001/02/15. [DOI] [PubMed] [Google Scholar]
  • 353.Louwerse ES, Weverling GJ, Bossuyt PM, Meyjes FE, de Jong JM. Randomized, double-blind, controlled trial of acetylcysteine in amyotrophic lateral sclerosis. Arch Neurol. 1995;52(6):559–564. doi: 10.1001/archneur.1995.00540300031009. Epub 1995/06/01. [DOI] [PubMed] [Google Scholar]
  • 354.Lange DJ, Murphy PL, Diamond B, Appel V, Lai EC, Younger DS, Appel SH. Selegiline is ineffective in a collaborative double-blind, placebo-controlled trial for treatment of amyotrophic lateral sclerosis. Arch Neurol-Chicago. 1998;55(1):93–96. doi: 10.1001/archneur.55.1.93. [DOI] [PubMed] [Google Scholar]
  • 355.Desnuelle C, Dib M, Garrel C, Favier A, Grp Ar-tS. A double-blind, placebo-controlled randomized clinical trial of alpha-tocopherol (vitamin E) in the treatment of amyotrophic lateral sclerosis. Amyotroph Lateral Sc. 2001;2(1):9–18. doi: 10.1080/146608201300079364. [DOI] [PubMed] [Google Scholar]
  • 356.Graf M, Ecker D, Horowski R, Kramer B, Riederer P, Gerlach M, Hager C, Ludolph AC, Grp GVE-AS. High dose vitamin E therapy in amyotrophic lateral sclerosis as add-on therapy to riluzole: results of a placebo-controlled double-blind study. J Neural Transm. 2005;112(5):649–660. doi: 10.1007/s00702-004-0220-1. [DOI] [PubMed] [Google Scholar]
  • 357.Kaufmann P, Thompson JLP, Levy G, Buchsbaum R, Shefner J, Krivickas LS, Katz J, Rollins Y, Barohn RJ, Jackson CE, Tiryaki E, Lomen-Hoerth C, Armon C, Tandan R, Rudnicki SA, Rezania K, Sufit R, Pestronk A, Novella SP, Heiman-Patterson T, Kasarskis EJ, Pioro EP, Montes J, Arbing R, Vecchio D, Barsdorf A, Mitsumoto H, Levin B, Grp QS. Phase II Trial of CoQ10 for ALS Finds Insufficient Evidence to Justify Phase III. Annals of neurology. 2009;66(2):235–244. doi: 10.1002/ana.21743. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES