Abstract
Multiple studies indicate that United States veterans have an increased risk of developing amyotrophic lateral sclerosis (ALS) compared to civilians. However, the responsible etiological factors are unknown. In the general population, specific occupational (e.g. truck drivers, airline pilots) and environmental exposures (e.g. metals, pesticides) are associated with an increased ALS risk. As such, the increased prevalence of ALS in veterans strongly suggests that there are exposures experienced by military personnel that are disproportionate to civilians. During service, veterans may encounter numerous neurotoxic exposures (e.g. burn pits, engine exhaust, firing ranges). So far, however, there is a paucity of studies investigating environmental factors contributing to ALS in veterans and even fewer assessing their exposure using biomarkers. Herein, we discuss ALS pathogenesis in relation to a series of persistent neurotoxicants (often emitted as mixtures) including: chemical elements, nanoparticles and lipophilic toxicants such as dioxins, polycyclic aromatic hydrocarbons and polychlorinated biphenyls. We propose these toxicants should be directly measured in veteran central nervous system tissue, where they may have accumulated for decades. Specific toxicants (or mixtures thereof) may accelerate ALS development following a multistep hypothesis or act synergistically with other service-linked exposures (e.g. head trauma/concussions). Such possibilities could explain the lower age of onset observed in veterans compared to civilians. Identifying high-risk exposures within vulnerable populations is key to understanding ALS etiopathogenesis and is urgently needed to act upon modifiable risk factors for military personnel who deserve enhanced protection during their years of service, not only for their short-term, but also long-term health.
Keywords: Amyotrophic lateral sclerosis, Veterans, Etiology, Neurotoxicant, Persistent, Exposure
Introduction
Amyotrophic lateral sclerosis (ALS) is a predominantly sporadic, paralytic, and fatal condition caused by motor neuron degeneration [1]. Despite the identification of disease-causing mutations for ~ 10% of cases [2–4], the pathogenic mechanism(s) of ALS remain elusive, hampering the development of effective preventive or therapeutic strategies. It is thought there are likely as many types of toxic exposures leading to sporadic ALS as there are genetic variants causing familial or inherited ALS (> 25 causative genes) [4]. Interestingly, not all carriers of ALS-causing mutations develop ALS, even in old age. For instance, most mutations in TDP-43 exhibit a reduced penetrance [5]. There is also a large disparity at what age familial ALS presents. These observations suggest that, even in familial ALS, there are environmental influences. While sporadic ALS occasionally results from oligogenic causes [6, 7], it is generally believed that most sporadic cases result from a combination of environmental factors and genetic susceptibilities. Currently, identifying rare genetic susceptibility factors that synergistically interact with a given environmental exposure requires a large case–control analysis, and often through the efforts of a global consortium.
The importance of environmental exposures in ALS etiology is supported by reports of high discordance of this disease in monozygotic twins [8, 9], conjugal ALS [10] and an increased risk of ALS for specific occupations [11], activities [12] and exposures (e.g. metals [13, 14], pesticides [15]). Relevant to veterans affected with ALS, disease risk increases, sometimes two-fold, with deployment [16] or total years of deployment [17]. Beard et al. also found ALS was positively associated with exposure to herbicides, pesticides, certain metals and burning agents [17]. Another study called genes and environmental exposures in veterans (GENEVA) used a retrospective exposure assessment to evaluate which industrial and occupational exposures, next to genetics, could drive the higher risk of ALS among military veterans [18]. They found that raters' confidence-modified exposure scores revealed potential higher exposure to hydrocarbon solvents, chlorinated solvents, and pesticides. Such evidence suggests that military personnel might be exposed to a variety of neurotoxicants and carcinogens whose adverse effects only emerge after sustained long-term exposure.
A recent study reported that the prevalence of El Escorial-defined ‘definite’ ALS cases is significantly higher among Air Force personnel, as well as among tactical operation officers and health care workers compared to other service branches and occupations [19]. These branch-specific differences are not reported by all studies [17, 20, 21]. Although a topic of debate, the suggested variation in rates among military branches may indicate that there are disproportionate exposures and/or factors experienced by certain military personnel. Despite such reports, there is no clear connection between a given environmental exposure and ALS veterans to date. One possible reason for this gap in understanding is that only a few studies (notably Fang et al., 2010 [22]) have evaluated biomarkers of physiological exposure to specific neurotoxicants, such as lead (Pb), in this population.
In this review, we assert that to reliably characterize the physiological burden of exposure in veterans, it is important to evaluate the potentially causal chemicals in disease-relevant tissues, such as the central nervous system (CNS). Importantly, the causative environmental exposure(s) likely occurred years prior to disease onset (e.g. 10 + years). Thus, due to changes of residence, retirement, etc., a given ALS patient is likely no longer actively exposed to the causal toxicants at the time of diagnosis. While epidemiological exposure assessments are therefore challenging, the direct measurement of persistent neurotoxicants from subject tissue remains feasible and offers many advantages. For instance, toxic metals, such as Pb, accumulate in bones where they have a half-life of up to 30 years [23]. As a result of aging and decreased activity, metals like Pb can be mobilized from bones, thus internally extending potential exposures that may have initially occurred years ago. Similarly, the CNS is also known to accumulate such metals with a longer half-life than most soft organs [24] as well as lipophilic toxicants with stable chemical structures due to the high lipid content of CNS tissue [25]. Although this approach may not capture all toxicants that contribute to ALS (e.g. those that are not persistent), persistent and non-persistent toxicants are often co-emitted, and thus tracking the persistent ones presents a feasible mechanism for the identification of fundamental sources of exposure that will provide an overall more informed understanding of ALS environmental etiology as a whole.
Together, there are several questions that could greatly increase our understanding of the particular vulnerability of military veterans to ALS:
What toxic environmental exposures/toxicants are associated with ALS?
What concentrations and/or (sub)cellular accumulations of such toxicants are relevant to disease?
Are there synergistic patterns between associated toxicants and predisposing/lifestyle factors (e.g. prior head trauma, spinal cord injury, smoking, occupation etc.)?
Similarities in disease development between ALS and cancer
In 2014, a groundbreaking epidemiological study by Al-Chalabi et al. described the development of ALS as a multistep process, similar to cancer [26]. The study was based on the premise that both ALS and cancer share characteristics such as the “onset being more common in later life, progression usually being rapid, the disease affecting a particular cell type, and showing complex inheritance” [26]. Using ALS registries from Ireland, the Netherlands, Italy, Scotland and England, Al-Chalabi and colleagues demonstrated a linear relationship between log incidence and log age in all five registries that was consistent with a six-step process [26], possibly derived from a combination of environmental exposures, genetic and other risk factors. Chiò et al. further demonstrated that patients with genetic mutations had fewer steps in their development of ALS, thereby supporting the idea of ALS as a multistep process [27]. Identifying exposures that serve as key steps or “hits” in ALS development will likley greatly increase our collective understanding of ALS etiology as well as the development of preventative and therapeutic strategies.
In this respect, military service members appear to represent a distinct subpopulation to which these principles could be applied. For instance, ALS incidence was found to be significantly higher during an approximate 10-year window following deployment for first Gulf War relative to those not deployed to the Gulf [28, 29]. Possibly following key exposures and/or interplay between toxicants and other risk factors, this defined window between service and disease onset provides a potential timeframe for the manifestation of ALS. Moreover, one study reports 85% of incident cases in Gulf War veterans within this window were < 45 years of age [30]. This decreased age of onset (typical ALS onset is ≥ 60 years in the general population) may also suggest that the exposures experienced by military personnel were more frequent and/or intense than that of persons developing ALS in the general population. We posit that service-linked exposures may represent one step (or several) in the proposed multistep process of ALS development [26].
Military service and ALS
There are numerous reports suggesting an increased risk of ALS associated with military service [16, 31, 32]. A representative report by the United States National Academy of Medicine (US NAM, previously the Institute of Medicine) shows that, regardless of what war, ALS rates were generally increased and often doubled compared to rates in non-military individuals [16]. Although evidence based on such studies has often been deemed limited but suggestive (e.g. due to small samples sizes, given the rarity of disease), ALS is currently defined as a service-connected disease and the Department of Veterans Affairs (VA) has provided additional financial assistance to Gulf War veterans suffering from ALS [33]. As of 2010, more than 2 million service members have been deployed in support of Operation Enduring Freedom and Operation Iraqi Freedom [34]. ALS prevalence among US veterans deployed (2002–2015) was 19.7/100,000 [19]. Thus, understanding risk factors in the military setting could provide valuable insight and future mitigation strategies for current and future military service members.
The Gulf War serves as a notable case study when examining the connection between military service and ALS. In addition to a doubled incidence of disease compared to general population [28, 29], Haley observed that 8 years postwar, 85% of the incident cases in Gulf War veterans were < 45 years of age [30]. The observed incidence in young veterans increased from 1 to 5 cases per year and was not explained by “a change in the interval from onset to diagnosis or by a change in the US population death rate of ALS in those aged < 45 years” [30]. To this end, NAM have considered relationships between Gulf War illnesses in general (including ALS) and broad exposures to environmental toxicants including insect repellents, such as N,N-diethyl-m-toluamide (DEET), oil well combustion products such as organic hydrocarbons (VOCs), diesel exhaust, and various polycyclic aromatic hydrocarbons (PAHs), or other substances likely to be encountered during service such as acid aerosols, hydrazines, mustard agents, sarin, pyridostigmine bromide, and depleted uranium [35]. However, NAM found insufficient evidence to demonstrate that subclinical exposures to these agents was associated with specific patterns of illness [36]. Previous studies probing this connection have been criticized in part for (1) a lack of evidence of a biologically credible environmental exposure that could result in such an increase in neurodegenerative illness (no reliable biomarker of CNS exposure) and (2) the statistical methodologies studies used [37–41]. In regards to the latter, however, subsequent investigations have confirmed the increased risk of ALS among deployed personnel using capture/recapture methodologies [42].
Although the etiology of ALS in veterans remains unclear, it is clear military service represents a unique circumstance that aggregates a special group of collective factors. For instance, military service members are exposed to multiple environmental hazards during deployment (e.g. pollutants from unregulated industry, particulate matter from desert environments, exhaust from military vehicles and aircraft, emissions from open-air burn pits and toxicants on military bases); among the myriad of toxicants present in these service-connected exposures, several were linked to diseases (e.g. lead, pesticides, dioxins, and even aerosolized cyanobacteria pertaining to Gulf Wars [43]). Soldiers also receive prophylactic treatment of cholinergic inhibitors, which have been linked with neurodegenerative disease and potentially ALS [17, 44], to protect them against nerve gas and insect pests. Beard et al. additionally identified numerous exposures associated with ALS and those with odds ratios > 3 included: treatment with nasopharyngeal radium, mixing or application of herbicides or burning agents as well as exposure to ground-level fumigation, nearby missile explosions, chemical agent-resistant compound paint or depleted uranium for munitions/armor [17]. Thus, while an effort has been made to link exposures to veteran service [32], no toxic exposures was demonstrated for its causal role in ALS.
In addition to environmental exposures, predisposing behavioral activities have also been considered in the context of military service and ALS. It has been postulated that the extreme physical exertion experienced by military personnel might also heighten ALS risk as well as smoking and alcohol consumption. However, variation in postwar ALS rates between military branches are not consistent with this idea. Altogether, previous attempts to establish a connection between military-linked environmental exposures and ALS based on historic records and questionnaires has proven difficult. Thus, evaluating the physiological dose of potential neurotoxicants in target tissues using quantitative and advanced molecular techniques would allow for stronger and improved correlations to be made between persistent toxicants and ALS. Although certain environmental toxicants have short half-lives and would not be measurable in postmortem CNS tissue, selecting toxicants that are (1) likely to contribute to disease based on evidence from the literature (see below) and (2) would be measurable as much as a decade or more after exposure will likely yield robust information. Potential toxicants include chemical elements, nanoparticles (NPs) and persistent, lipophilic toxicants such as dioxins, polycyclic aromatic hydrocarbons (PAHs) and polychlorinated biphenyls (PCBs). These environmental toxicants and their sources are described in more detail below.
ALS-relevant and military-linked environmental exposure sources
Diesel exhaust
Certain occupational studies have found increased ALS risk among persons in occupations with high exposure to diesel exhaust [45], such as truck drivers [46, 47] and construction workers [48], machine operators [49] and bus drivers [49]. Thus, diesel exhaust represents a proposed common denominator for ALS. Diesel engines of comparable power produce between 2 and 10 or up to 40-fold more particulate emissions than gasoline engines without or with a catalytic converter, respectively [50]. Moreover, inhaled diesel exhaust particles are ‘biopersistent’ and may carry additional chemical compounds on their surface [51]. Thus, the importance of diesel engine emissions to human health is not only a reflection of the toxic chemicals produced, but also the ability of particulate matter to ‘trap’ additional chemicals, thereby potentially extending the duration of exposure [51]. As a result, this may allow for the prolonged contact of passenger chemicals with the respiratory epithelium and increase their chance of uptake and subsequent initiation of oxidative stress, systemic inflammation and de novo mutations from resultant genomic damage [51, 52]. Diesel exhaust also contain PAHs such as benzo(a)pyrene and as well as additional toxicants and carcinogens (acetaldehyde, acrolein, benzene, 1,3-butadiene, ethyl benzene, formaldehyde, n-hexane, naphthalene, styrene, toluene, and xylene) identified by the US Environmental Protection Agency (EPA) [53]. Accordingly, one must consider diesel exhaust-derived toxicants as potential risk factors for neurodegeneration and, more specifically, ALS.
Living in close proximity to heavy traffic and air pollution is associated with a higher incidence of dementia [54] and risk of neurodegeneration. For example, fine particulate air pollution (particulate matter < 2.5 microns; PM2.5) has been linked to Alzheimer’s disease (AD) and Parkinson’s disease (PD) [55]. Specific to ALS, one recent study found suggestive evidence of increased ALS risk due to long term, high-concentration exposures to particulate matter < 10 microns (PM10) [56]. However, the statistical imprecision of the risk estimates due to the small sample size limited their capacity to definitively conclude an increase in risk [56]. In another study, Nunez et al. suggest that increased PM2.5 concentrations in New York state may contribute to the clinical aggravation of PD and ALS, especially for subjects > 70 years of age [57].
Jet exhaust
Detrimental health effects have been observed for those working or residing in close proximity to jet emissions, including certain cancers [58, 59]. As evidenced in vivo, exposure of mice to particles collected at two separate airports via intratracheal installation induced pulmonary acute phase response, inflammation and genotoxicity [60]. In connection with ALS, the literature suggests that civilian airline flight attendants, pilots and navigators likewise have higher rates of this disease [59, 61]. Given that ALS prevalence has been found higher for Air Force service members relative to other military branches by certain studies [19, 29], we speculate that exposure to jet exhaust may serve as a contributing factor.
Products from jet emissions include ultrafine combustion particles, lubrication oils (including organophosphate esters), PAHs, volatile organic compounds and chemical elements including: Pb, Copper (Cu), Chromium (Cr), Nickel (Ni), Iron (Fe), Zinc (Zn), and Aluminum (Al) among others [58]. Although we are aware of their production, the toxicity of aircraft emissions has not been extensively researched. Emitted particles are generally ultrafine (< 100 nm). Like particles derived from other combustion sources, they may deposit in respiratory airways when inhaled and potentially cause irreversible damage to lung tissue, as observed for NPs generally [62, 63]. Researchers have investigated the toxicity of NPs from the exhaust of a CFM56-7B turbofan, the most common aircraft turbine engine. Using an aerosol deposition chamber, Jonsdottir et al. observed varying amounts cell death and oxidative stress in cultured bronchial epithelial cells depending on the combination of turbine thrust level and fuel type [64]. This is of importance as different thrust settings and fuels (including jet propellant (JP)-8 jet fuel) reportedly emit varying particle amounts [65]. Similar to the commercial jet A-1 fuel, JP-8 is a kerosene-based fuel that is conventionally used in Air Force aircraft, military vehicles and generators, thus, is common chemical exposure for Air Force-associated personnel (e.g. flight and ground crew) [65, 66]. Although no direct connection has been made, it is of note that Pugh et al. identified a significant increase in chronic obstructive pulmonary disease and asthma in veterans from the Iraq and Afghanistan Wars [67], which could possibly be associated with military-linked, environmental exposures. Exposure to jet fuel itself has been linked to hearing loss in both animal models [68] and humans [69]. Akin to urban pollution (described above), such auditory defects may also be potentially connected to NP-associated, brainstem pathologies [70].
Together, current evidence suggests that exposure to toxicants derived from diesel and jet engine exhaust can cause detrimental health effects. However, there are differences between the two sources. One study found the size distribution of aircraft exhaust particles, including lead dibromide, is smaller than that produced by automobiles burning the same leaded fuel (13 vs. 35 nm mean diameter for all particle types, respectively) [71]. In a separate study, although 10–1000 nm particles were associated with both roadways and aircraft, a principle component analysis conducted by the authors revealed features that differentiated the two sources: larger particle size and higher black carbon concentrations were a feature of roadways, while smaller particles and lower amounts of black carbon were associated with aircraft [72]. Such differences may be relevant to our understanding of emission-linked exposures when considering their contribution to disease development, although it is possible the overall physiochemical properties of each are ultimately similar [60]. Overall, the neurotoxic mechanisms and potential for such environmental pollutants to drive the increased ALS risk observed in military service members, airline workers and truck drivers needs further study.
Brake and tire wear
In addition to the exhaust produced by combustion and other pollution sources associated with airports and air force bases, aircraft breaking and tire wear can also lead to the release of potential toxicants through the abrasion and subsequent production of smoke and ultrafine dust from these mechanical systems [65, 73]. For instance, experimental data collected at a major European airport have also reported high levels metals including barium (Ba), Zn, molybdenum (Mo), Cu and antimony (Sb) from runway smoke [74] and separate study also observed the production of trace elements generated by aircraft landing [75]. Although data are limited, one study estimates that the rubber lost from tires on varying aircraft can vary from tens of grams to ∼0.8 kg per landing [76].
Burn pits
Burn pits are open areas for burning solid waste that were widely used in combat zones before 2009. A report by the Institute of Medicine described burn pits in Iraq and Afghanistan as burning waste that “consisted generally of 5–6% plastics, 6–7% wood, 3–4% miscellaneous noncombustibles, 1–2% metals, and 81–84% combustible materials” [77]. JP-8 was typically used as the accelerant and, although there are no official inventories, the refuse reportedly burned (i.e. plastics, metal cans, rubber, paints, solvents, petroleum, munitions and wood waste) produced hazardous emissions containing harmful particles and toxicants [77]. Unsurprisingly, exposure to burn pit emissions has been a cause for concern in relation to respiratory illness, cancer and neurological effects [77–79].
Pollutants investigated and detected during the evaluation of the Joint Base Balad burn pit included: dioxins, particulate matter (including metals), PAHs, volatile organic compounds [77]. Concentrations of polychlorinated dibenzo-p-dioxins and dibenzo-p-furans were overall low, but generally increased relative to urban environments. Additionally, a large contribution of the detected particulate matter, PAHs and volatile organic compounds were concluded to have likely originated from traffic and jet emissions as well as regional sources (e.g. normal human activity, dust storms, etc.). Thus, in combination with the presence of burn pits, such a nexus of exposures could potentially cause additive or synergetic health effects for those exposed. A public law mandated by Congress in 2013 required the VA to establish a registry for veterans with potential burn pit exposure in Iraq or Afghanistan through which participants can complete a questionnaire detailing their deployment/occupational, health and exposure history [80]. Together, these data could be used to epidemiologically investigate the link between burn pit exposure, in potential combination with other service-linked exposures, and ALS.
Neurotoxic effects of toxicants
PAHs
PAHs are by-products of combustion [81] and enter the human body from a variety of sources including: gasoline and diesel-fueled engines (e.g. jet fuel) [82], coal, solid waste, and oil burners [81], grilled and smoked meats [83], use of indoor fireplaces and stoves [84], and smoke from cigarettes [83, 85]. PAH residues are found frequently in suspended fine or ultrafine particulate matter in the air and inhalation is a major route of exposure [86]. They are lipophilic, are stored in fat tissues including those of the breast [87], and have been shown to cause mammary cancer in rodents [88]. In humans, autopsied samples revealed that the highest accumulation of PAHs occurred in abdominal fat and the brain [89]. At the toxicological level, some PAH compounds are able to bind the aryl hydrocarbon receptor (AHR), which regulates xenobiotic-metabolizing enzymes including some of the cytochrome P450s, ultimately resulting in increased DNA mutations [90, 91]. PAHs also have been found to be directly genotoxic [92]. In the rodent brain, a gene expression analysis showed that exposure to PAHs in the form of diesel exhaust emissions activates several genes associated with antioxidant defenses and inflammation [93]. In addition, benzo[a]pyrene (B[a]P), an individual PAH compound, has been identified for its neurotoxic potential [94]. An in vitro study elegantly demonstrated that B[a]P is not directly toxic to neurons; it kills neurons via a non-cell autonomous mechanism by activating microglia [95], the resident immune cells of the brain that have been shown to play a key role in ALS [96]. Locomotor impairment after chronic exposure of B[a]P is significantly associated with development of neurodegenerative phenotypes typically affecting the dopaminergic system in zebrafish [97]. Histopathological observations in brain tissues showed a significant increase in pyknotic neuronal counts in the diencephalon and telencephalic region of zebrafish brain after B[a]P exposure and tyrosine hydroxylase, a marker of dopaminergic neurons, was reduced significantly in the exposure group. In addition to their neurotoxic effects, PAHs can be used to indicate exposure to incompletely combusted pollutants, such as soot. Soot particles can enter the brain and PAH compounds trapped inside the soot particles can be used to indicate the exposure from combustion [98].
Persistent organic pollutants (POPs): PCBs and dioxins
PCBs and dioxins are known as persistent organic pollutants (POPs) and have all been found at military bases and in conjunction with service-linked activities as previously described. In general, POPs are resistant to environmental degradation and can bioaccumulate, resulting in adverse impacts on human health such as an increased risk of ALS [99] and reduced ALS survival in association with increased plasma concentrations [100]. PCBs and dioxins are lipophilic and accumulate in fatty tissues [101], including the brain. Interestingly, accumulation of PCBs in the brain was clearly shown not to have any regionalization [102]. However, in adolescent and neonatal male rats, exposure to PCBs was shown to induce brain region-dependent exacerbation (e.g., in the hypothalamus) or silencing (e.g., the prefrontal cortex) of genes implicated in neuroimmune function such as those coding for factors of the nuclear factor kappa b (NF-κB) complex [103, 104]. This suggests that despite homogeneous brain accumulation, PCBs can have brain area-specific neurotoxic effects probably based on regional gene–environment interaction. These two studies also indicate that neuroimmune dysregulation may be a prominent pathway of PCB neurotoxicity specifically in males. A similar sexual dimorphism is observed in ALS which preferentially affects men with a male:female ratio that lessens (from ~ 2.5 to 1.4) with age [105]. PCB exposure has been associated with reduced cognition in older adults [106] and the specific PCB, PCB-151, has an increased odds ratio in relation to ALS [107]. It is of note that POPs, such as PCBs [108], are associated with multiple neurodegenerative diseases, such as PD [109–118]. Although diseases like ALS, PD and AD do not share the same neuropathology, it is possible that an overlapping, genetic susceptibility occurs through pleiotropy and lends to the existence of common environmental triggers [119–121].
Dioxins are another class of POPs and are a component of certain pesticides, such as Agent Orange, to which ALS has been positively associated [17]. Moreover, elevated levels of the dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), has been detected in breast milk from mothers living near the Vietnam Bien Hoa Air Base [122]. Exposure to TCDD from Agent Orange is believed to be an immunotoxin and the probable cause of several types of cancer in Vietnam veterans based on its clear carcinogenicity in experimental animal models [123]. One key mechanism proposed for TCDD carcinogenicity is epigenetic remodeling [124]. Supporting this view, exposure to dioxins or dioxin-like compounds has been associated with CNS developmental abnormalities in zebrafish and epigenetic modifications are well known to be particularly important during neuronal development [125]. Dioxins may also alter the expression of genes related to neuroimmune function [126], neurotransmission [127, 128], neurodevelopment [129, 130] and cytotoxicity [131, 132]. Overall, the molecular cascade by which the neurodegeneration-linked, chemical toxicants described thus far (dioxins, PCBs, and PAHs) contribute to disease is unclear, although one possibility is neuroimmune changes implicating in particular microglial cells potentially via the activation of the AHR. In vitro studies report agonists of the AHR, which interestingly was shown to induce a robust DNA demethylation of the Cyp1a1 promoter upon dioxin exposure [133], may induce up to a threefold increase in the ALS-linked TDP-43 protein in BE-M17 (human neuronal cell line), motor neuron differentiated iPSCs and the murine brain [134]. Although further research remains, such observations suggest the possibility that TDP-43 could be a potential molecular link between exposure to dioxins and ALS.
Neurotoxic metals and elements
Specific metals have been linked to neurotoxicity and ALS [135–138]. In fact, exposure to certain metals was one of the earliest environmental risk factors proposed for ALS, although the jury is still out regarding their pathogenic role [139]. For instance, depending on the biospecimens used, there are discrepancies in the findings reported and peripheral measurements rarely reflect the CNS metal load [140, 141]. As described above, exposure to toxic metals is likely a major concern in veterans and a number of metals including Pb, mercury (Hg), selenium (Se), Cu, manganese (Mn), Fe and arsenic (As) certainly deserve further investigation in ALS patient, CNS tissue. These metals were selected for their relevance to ALS in the literature and our own epidemiological studies [12, 142–144] and are described in more detail below:
Lead (Pb) was the first metal found to be elevated in cerebrospinal fluid (CSF), blood, and tibia from ALS patients [22, 145, 146]. Systematic reviews and meta-analyses cite Pb as an exposure with ‘convincing evidence’ of a causal link to ALS [147, 148]. Animal studies demonstrate the transport of Pb2+ ions across the blood–brain barrier [149] and the accumulation of insoluble TDP-43 in the cortex of exposed mice [150]. Based on the analysis of blood samples from occupationally exposed humans, there is evidence to suggest Pb-induced oxidative damage [151]. Relevant to military exposures, shooting at firing ranges results in the discharge of Pb dust, and elevated blood Pb levels that are associated with a variety of adverse health outcomes including essential tremor, cardiovascular morbidities and mortality, and decreased renal function [152]. In Denmark, occupational Pb exposure 10 years prior to diagnosis was associated with increased ALS risk (odds ratio 1.33, 95% confidence interval 1.03–1.72) [153]. Our own prior work linked activities involving Pb (e.g. casting lead bullets, making stained glass with lead joints and casting or using lead fishing sinkers) to ALS risk [144]. Moreover, Pb exposures 20 + years prior to diagnosis had larger effect sizes compared to those occurring more recently [144]. A further Australian study also linked petrol Pb emissions to ALS death rates both temporally and spatially [154].
Mercury (Hg) is potentially a risk factor for ALS [142, 143]. This metal was elevated in the brain of seven ALS patients compared to controls [155] and is associated with increased risk in several studies [139, 147, 156]. Case reports of Hg poisoning have shown convincing ALS-like, clinical symptoms and support a causal relationship [157–159]. Our own regional and nationwide US studies demonstrated higher toenail Hg levels in ALS patients compared to controls [142, 143]. In mutant SOD1 ALS mice, Hg accumulates in spinal neurons [160] and rats exposed to methylmercury (2 mg/kg/day) exhibited ALS-like neurological effects [161]. In vitro proteomics studies reveal that methylmercury exposure causes electron transport chain dysfunction, oxidative stress and ubiquitin proteasome system impairment [135], pathological mechanisms all linked to ALS. Methylmercury neurotoxicity may also involve glutamate dyshomeostasis and excitotoxicity [162], an ALS-linked mechanisms that could be of particular relevance to the multi-stage hypothesis of ALS [26].
Selenium (Se) was also linked to ALS [163–166]. Embedded shrapnel from explosive devices and retained bullet fragments also can increase serum levels of toxic metals including Se [167]. Higher risk of ALS was found in naturally seleniferous US regions [168, 169]. Veterinary and experimental animal evidence suggests that motor neurons are particularly vulnerable to Se [139, 164]. Despite Se being elevated in the spinal cord of ALS patients [163, 170], studies measuring Se levels in ALS-patient CSF and blood found both negative and positive correlations with ALS status [13, 166, 171]. Some of the discrepancies in these studies may be due to interactions among elements. For example, although not yet conclusively shown in humans, Se may counteract the absorption of methylmercury [172, 173].
Copper (Cu) has often been a focus of ALS research due to its role as a cofactor of SOD1 [174–176]. Moreover, the pesticide, copper sulfate can dissolve in blood after exposure and be carried throughout the body as well as transported across the blood brain barrier (BBB) as a free Cu ion [177]. Increased Cu levels were measured in the spinal cord of mutant TDP-43 ALS mice [175] and in the blood of veterans with ALS [13]. However, in various peripheral biospecimens, trends in Cu levels were inconsistent [141].
Excessive manganese (Mn) exposure is known to cause manganism, a neurological disorder resembling PD [178]. Moreover, Mn can cause ALS-like symptoms (such as muscle weakness) and ALS-like lesions of the corticospinal tract were reported in Mn-exposed workers [179]. In agreement, increased ALS rates have been reported in Mn miners and smelters worldwide [180, 181]. Elevated Mn levels were detected in the spinal cord of ALS patients and animal models [175, 182], whereas in patient body fluids, both negative and positive findings were reported [13, 174, 183, 184]. Roos et al. observed CSF Mn to be significantly increased compared to CSF of controls [184]. Notably, CSF Mn concentrations were higher than those in the plasma concentrations, suggesting transport of this element into the CNS.
Iron (Fe) accumulation in the CNS has been clearly demonstrated in ALS patients, animal models and in vitro cell models [185–187]. In contrast, most studies using peripheral biospecimens did not find sizable differences in Fe levels in blood, hair, nail, CSF, and urine of ALS patients and controls [140]. Yet, Fe likely has a role in ALS as chelation of this metal is beneficial in a SOD1 ALS mouse model [188].
Lastly, arsenic (As) exposure was shown to cause ALS-like motor neuropathy [189–191] and was suggested as a risk factor for ALS in association with folate deficiency [192]. A recent study reported lower As serum concentration in ALS patients, but As positively correlated with disease duration [136]. Prolonged As exposure in vitro triggers several features of ALS-linked TDP-43 pathology (e.g., mislocalization, aggregation) [193].
In addition to the ALS-linked elements described above, the following also possess evidence of an association with ALS: Using CSF (n = 17 cases, n = 10 controls), Roos et al. showed positive associations of ALS with Al, and cadmium (Cd), cobalt (Co), Zn, vanadium (V), and uranium (U) [137]. However, the Al finding was contradicted by a 1995 study of CSF that did not show elevation in ALS patients [185] as well as an additional study of temporal lobe tissue across neurological conditions did not find Al levels elevated in n = 16 ALS patients compared to age-matched controls [194]. Hozumi et al. found increased risk of ALS associated with Zn in the CSF (n = 52 cases, n = 15 controls). Zn was also elevated in CSF in a separate Greek cohort [195] as well as in the Roos 2013 CSF study [137], although this work was contradicted by Kapaki, who found no association with ALS and these elements in CSF (n = 28 cases, n = 36 controls) [196]. A further case study reported development of ALS in a battery worker exposed to high levels of Cd [197], although, elsewhere, Cd levels were lower in the CSF of ALS patients compared to controls [198]. Hozumi et al. found increased risk of ALS associated with elevated CSF magnesium (Mg) [183]. Using a cohort from the Danish National Patient Registry from 1982 to 2013 matched to controls, Dickerson et al. evaluated cumulative metal exposures estimated using job exposure matrices applied to occupational history. No statistically significant associations to ALS were discovered looking at Cr and Ni [199]. Lastly, based on studies of neurodegenerative illness in animals, Mo deficiency has been postulated to be a potential predisposing factor in ALS [200].
Several studies have directly assessed brain tissue for trace element levels. A review article of brain autopsy tissue sample measurements cites the detection of a large variety of the elements (see Table 3 of Grochowski et al. [201]), and provides quantitative reference levels for Cu, Zn, Mg, Fe, calcium (Ca), rubidium (Rb), Se, potassium (K), Mn, Al, silicon (Si), As, Ni, Pb, Cd, and Cr in various brain regions [201]. In studies of ALS, Cd and Zn were both significantly elevated in grey and white matter when Cd, Co, Cu, Fe, Mn, Rb, V, and Zn were measured in the brain tissue of n = 8 Guam ALS patients versus n = 5 controls [202]. By focusing on veterans, a population with increased ALS risk, and conducting a comprehensive assessment of elements in CNS tissue, one could potentially identify toxic elements that may not have been previously linked to ALS or examined in the general population. For example, uranium (U), Sb or tungsten (W; associated with munitions), rare earth and platinum group elements have not typically been measured in brain tissue.
The mechanisms of toxicity by which metals contribute to neurodegeneration in ALS remain to be clarified. However, in other neurodegenerative diseases like AD, multiple studies have validated that exposure to metals disrupts critical, immune-related pathways leading to chronic neuroinflammation and neuronal loss [203]. Metals have often been studied separately in terms of immunotoxicity and neurotoxicity, but one elegant study demonstrates that Pb, for instance, induces the production of autoantibodies against neural proteins, including myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) [204]. Therefore, Pb could contribute to the aggravation of neurodegenerative disease by exacerbating the immunogenicity of nervous system proteins. The consequences of metal dyshomeostasis in ALS has recently gained momentum and helped generate novel hypotheses, such as those linking mitochondrial dysfunction, intracellular calcium dyshomeostasis, pathological TDP-43 formation, pro-inflammatory microglia activation, and ultimately programmed neuronal cell death [205].
Nanoparticles (NPs)
NPs are particles ≤ 100 nm in all dimensions and exist in a variety of shapes, sizes and compositions (organic, inorganic and carbon based). The nanometer scale of these particles allows for their direct interaction with cells and/or passage through biological barriers. Molecules (including proteins) can coat the NP surface, forming a NP corona [206, 207] and effectively become particle passengers [208]. Leveraging this property, NPs have been used as a therapeutic delivery mechanism for a variety of chemotherapeutic compounds [209]. However, NP pharmacological characteristics are often quickly altered in vivo as NPs become coated with biological milieu following delivery. The interaction of NPs with cells is influenced by the proteins and other molecules attached to their surface, as demonstrated by one ex vivo study reporting that the binding specificity of targeted NPs can be lost in the presence of plasma proteins [210]. The complexity and diversity of protein interactions with NPs to form the corona has not been completely elucidated [207]. While NP size and ability to transport molecules contributes to their therapeutic promise, these same characteristics may also interfere with vital cellular processes, resulting in cellular toxicity as well as human health and environmental concerns [63, 209, 211, 212]. For instance, silver (Ag) NPs have been predominately used for the development of medicines, drug delivery systems and medical device coatings as a result of their antibacterial properties [213, 214], yet multiple studies have demonstrated the toxicity of Ag NPs in vitro and in vivo [209]. Thus, the soft duality of NP features may also pose a detriment to a number of organs and highlights the need for a clearer understanding of these particles in biomedicine, manufacturing [215, 216] as well as general regulations for use [217].
As an environmental toxicant, NPs likely had a large impact in the days of Charcot when he first described ALS in the late 1860’s (e.g. as product of combustion). Today, additional sources of NPs posing a potential health hazard are found in the form of powders, suspensions, or sprays, which are universally used in textiles, paints, cosmetics, water disinfectants, food packing and ubiquitous in polluted environments with combustion emissions [218, 219]. Given these sources for exposure, the respiratory and gastrointestinal tracts, mucosa and even skin represent entry routes for environmental NPs [220–223]. While large aerosolized particles tend to remain in the respiratory tract, NPs may cross the respiratory epithelium to enter blood vessels [224]. Once in the bloodstream, select NPs can directly cross the BBB and/or damage BBB integrity and increase its permeability [225–228]. Additionally, numerous studies demonstrate that NPs can also bypass the BBB entirely via the olfactory system (nasal olfactory epithelial → olfactory bulb → brain) [222, 229–232]. Furthermore, the substantia nigra and brainstem have been suggested as targets for NPs via access the gastrointestinal tract and neuroenteric system [221, 222, 224]. Although our understanding of NP entry routes and subsequent toxicity is evolving, toxic NPs typically appear to be inorganic in nature and water insoluble; toxicity also appears linked to dose and frequency [233–235].
Once NPs reach the brain, they can access neurons, oligodendrocytes and glia they may alter the structure or activity of the nervous system and induce glial activation [236]. The primary neurotoxic mechanism of NPs is the generation of free radicals and induction of oxidative stress, which can damage biological macromolecules, inducing de novo mutations of DNA. In addition, NPs may directly or indirectly trigger apoptosis, autophagy, immune-responses, neuroinflammation and subsequent BBB damage [209, 237–247]. For instance, Xue et al. demonstrated SiO2-, TiO2-, and magnetite (Fe3O4)-NPs treatment caused microglial activation and cytokine secretion, resulting in PC12 toxicity and altered dopamine production [248]. Similarly, 10 and 30 nm Fe3O4-NPs were found to reduce dopamine rat brains as well [249]. Multiple factors may influence NP neurotoxicity including: size, shape, surface coatings, dissolution rates of metals, and interactions with specific cells and proteins [250]. Of particular, importance is size. In one study, Ag NPs (20 nm) exhibited increased cytotoxicity and pro-inflammatory response in cultured cells compared with larger particles (i.e. 80 nm) [251]. Similarly, a separate in vitro study indicated increased toxicity and oxidative stress from 20 vs 40 nm Ag NPs [252]. Ultimately, the exact neural damage and resultant neuropathology may depend on genetic susceptibility, individual NP characteristics and the differential access to target tissues achieved via their respective entry routes [253, 254] (Table 1).
Table 1.
Nanoparticle | Mechanism and relevance to neurodegeneration |
---|---|
Iron oxides: magnetite (Fe3O4) iron oxide (Fe2O3), | Axonal transport and bypass the BBB via the nasal olfactory epithelium [222, 255, 256] |
Daily exposure affects synaptic transmission and nerve conduction, causing neural inflammation, apoptosis, induced neural antioxidant responses, and immune cell infiltration [257] | |
Disrupted Fe homeostasis [258, 259], release of free Fe ions to catalyze the production of reactive oxygen species (ROS) through the Fenton reaction [249, 260] as well as the promotion of amyloid-β toxicity, as shown in vitro [261] | |
Silicon dioxide (SiO2) | Increased oxidative stress and altered microglial function; deleterious effects on the striatum and dopaminergic neurons [262] |
Intranasal administration in a mouse model lead to cognitive dysfunction and impairment, synaptic changes as well as pathologies similar to neurodegeneration [263] | |
Induction of neuron depolarization in a cell culture model; no detected change in gene expression [264] | |
PD-like behavioral changes in SiO2 NP-exposed Zebrafish model [265] | |
Dose-dependent cytotoxicity and AD-like pathology in vitro [266] | |
Titanium oxide (TiO2) | Absorption and translocation into the brain by any portal of entry. Can further cross the placental barrier and accumulate in the fetal brain, causing impairments in the fetal brain development [267] |
Damage to BBB and induction of inflammatory response [268, 269] | |
Exposure precipitates the development of neuropathological findings of early PD, AD and ALS, some of which appear to be manifested symptomatically [221, 253, 270, 271] | |
Nickel (Ni) | Ni NPs increased (Aβ)40 and Aβ42 levels in murine brains [272] |
Given the need to better understand the neurotoxic potential of NPs, one important question is: what is the threshold of NP exposure for neurodegeneration or neurodegenerative pathology? In study of Mexico City subjects, an autopsy study showed hyperphosphorylated tau in the brainstem of an 11-month-old baby, who was found to have to 20 μg/m3 cumulative PM2.5 (calculated for age at death + pregnancy time), a fraction of the 2522 μg/m3 calculated for a 39 year old subject with AD neurofibrillary tangle advanced stages V-VI [270]. Calderón-Garcidueñas et al. additionally observed extensive structural organelle abnormalities in the substantia nigra involving mitochondria, endoplasmic reticulum and neuromelanin that were co-associated with the abundant presence of exogenous, Fe-, Al- and Ti-rich NPs in a population of young residents [221]. They also identified hyperphosphorylated tau, α-synuclein and TDP-43 in the brainstem of 182 Mexico City 27.29 ± 11.8y old Metropolitan Mexico City residents [221]. The co-existence of markers for two common neurodegenerative diseases (sporadic AD and PD), as well as the less common ALS/frontotemporal degeneration (FTD), suggests a common etiological denominator. Thus, NPs may act as catalysts for reactive oxygen species formation, altered cell signaling, protein misfolding, aggregation and fibril formation [221], hence, the co-clustering of such diseases (ALS, PD, dementia) in select geographical pockets [273]. Similarly, Fe-rich and TiO2-NPs may (even at low concentrations) may accelerate α-synuclein fibrillization [274], thereby representing a possible a pathomechanism that could potentially contribute to development of neurodegenerative-disease-linked pathology. Together, Calderón-Garcidueñas et al. have suggested that the properties of NPs, which result in cellular damage, potentially represent an additional pathomechanisms contributing to the development of neurodegeneration [221].
It is clear there is extensive and unregulated exposure to nanoparticles released in the environment and that emission sources are highly variable across military and civilian populations (Table 2). With the noteworthy progress of nanotechnology during the last decade, NP products will continue to be used increasingly in our everyday commercial products, industrial processes and medical applications. Thus, in addition to considering the courses of nanoparticle themselves, other variables such as high traffic locations, residential areas, indoor environments, personal exposures, smoker, non-smoker, wind, season, etc. must also be taken into account to fully understand their potential risk.
Table 2.
Expected NP detection profile | Shape and size | Expected cellular/anatomical location and properties in vitro and in vivo |
---|---|---|
Elements from fuel combustion (gasoline, diesel, alternative mixed biofuels) and industrial sources (e-waste, lubricating oils) [275–282]: Fe, Pb, Ni, Cu, Cd, Hg, Al, Bismuth (Bi), Titanium (Ti) | Shape and size determine NP toxicity and capacity to reach target cells | Mitochondria, Golgi apparatus, lysosomes, phagosomes, and nuclei [221] |
Need to define localization of NPs in the neurovascular unit, including the BBB at endothelial level (integrity of tight junctions) [221, 289] | ||
Technology-critical elements [283]:Gallium (Ga), Germanium (Ge), Indium (In), Tellurium (Te), Niobium (Nb), Tantalum (Ta), Thallium (Tl) | ||
Platinum Group Elements: Platinum (Pt), Palladium (Pd), Rhodium (Rh), Osmium (Os), Iridium (Ir), Ruthenium (Ru) | ||
Rare Earth Elements:Yttrium (Y), Lanthanum (La), Cerium (Ce), Praseodymium (Pr), Neodymium (Nd), Samarium (Sm), Europium (Eu), Gadolinium (Gd), Terbium (Tb), Dysprosium (Dy), Holmium (Ho), Erbium (Er), Ytterbium (Yb), Lutetium (Lu) [205, 282, 284–287] | Coexistence of multiple metal NPs alter the original toxicity of individual NP [285, 288] | |
Detection of metalloid, Si [290], which may be of relevance in the veterans from the Gulf Wars where desert dust (e.g. SiO2) and particulate matter are prevalent [291] | Si NPs 7 + nm evoke oxidative stress and mitochondrial dysfunction [292, 293] | Within neurons, microglia, oligodendrocytes, astrocytes. Greater vulnerability of astrocytes expected [294] |
Subcellular accumulation in: Mitochondria, axons [295] and Autophagosomes [221, 296] | ||
Detection of Ti nanorods (versus spherical shape) to determine its industrial origin | Ti nanorods are associated with e-waste, while spherical Ti NPs are associated with food sources [297] | Storage in autophagosomes [221] |
Membrane damage, cell cycle interference, reactive oxygen species formation [296] |
Conclusions
The pathogenic mechanisms of ALS remain elusive and has hampered the development of prevention strategies and effective therapeutics for this fatal disease. Although efforts have been made in the veteran population to understand the observed elevated ALS rates, no definitive factors have been implicated. Thus, measuring persistent toxicants of interest in ALS-patient CNS tissue and, particularly in evaluating potential pathogenic exposures in veteran cohorts, is warranted. Such a research approach could provide the basis for environmental exposure associations that are not unique to ALS, but potentially other neurodegenerative diseases with shared pleiotropy and to which veterans are also at higher risk [119–121, 298, 299]. To address this major challenge in ALS research, the field needs to further develop brain banks. In the traditional line of the Armed Forces Examiner System and Institute of Pathology [300], the Veterans Affairs Biorepository established a Brain Bank (VABBB) which provides a collection of carefully characterized, prepared and preserved CNS samples [301]. Moreover, linking these specimens to comprehensive demographic, lifestyle, residential, occupational, and clinical data are critical for the identification of novel associations between ALS and environmental toxicants. The Veterans Affairs Cooperative Studies Program Epidemiology Center in Durham, North Carolina (CSPEC-Durham) has assembled such a repository including extensive research data, genomic data, and study specimens (e.g., DNA, blood) for different content areas including ALS [302].
In summary, it will be crucial to evaluate service-linked toxicants such as PCBs, PAHs, dioxins, metals and NPs in veteran CNS tissue, none of which have been adequately evaluated in relation to neurodegenerative disease risk. These toxicant exposures in veterans appear to be persistent and cumulative, thereby, potentially allowing one to assess the link between ALS and past exposures. Based on the literature reviewed here, we hypothesize that the concentrations and/or distribution of proposed neurotoxicants will be increased and/or in the CNS tissue of ALS compared to controls. Advanced statistical techniques could be applied to clarify the ALS multistep hypothesis, evaluate toxicant synergy as well as anatomical and/or (sub)cellular locations. Finally, as brain banks often also collect genetic variant data, an evaluation of gene and environmental interactions could also be undertaken [121], thus enabling individualized risk assessments and exposure prevention strategies for susceptible individuals.
During their years of service, military personnel voluntarily expose themselves to short-term, life-threatening risks. Moreover, they may also often and unknowingly expose themselves to environmental factors as part of their duties, which can have dramatic consequences for their long-term health and lifespan. It is our responsibility as environmental health scientists to devise the best research strategies to clearly identify such factors, link them to disease and alert competent governmental agencies. The risk(s) associated with adverse exposures are certainly modifiable by prevention and mitigation strategies (e.g. personal protective equipment, exhaust emission control and reduction systems), which could be enabled by military authorities at lower costs than those associated with highly debilitating chronic diseases such as ALS (e.g., average total disease duration cost per patient for care and service in the U.S. is $1,433,992, excluding societal cost and family support cost [303]). We are hopeful that further research will address the urgent need to act upon modifiable risk factors for military personnel who deserve enhanced protection during their years of service for both their short- and long-term health.
Author contributions
EWS and DBR conceived the manuscript. EWS performed the primary literature search and wrote the initial draft. DBR, BY, LC-G, ASA provided content as well as reviewed and edited the manuscript. MT assisted in writing, organizing and editing the manuscript.
Funding
Not applicable.
Availability of data and materials
Not applicable.
Code availability
Not applicable.
Declarations
Conflicts of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Ethics approval
N/A as this is a review of the literature.
Consent to participate
N/A as this is a review of the literature.
Consent for publication
N/A as this is a review of the literature.
References
- 1.Rowland LP, Mitsumoto H, Przedborski S. Amyotrophic lateral sclerosis, progressive muscular atrophy, and primary lateral sclerosis. In: Rowland LP, Pedley TA, editors. Merritt's neurology. Lippincott: Williams & Wilkins; 2010. pp. 802–808. [Google Scholar]
- 2.Mejzini R, et al. ALS genetics, mechanisms, and therapeutics: where are we now? Front Neurosci. 2019;13:1310. doi: 10.3389/fnins.2019.01310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mathis S, et al. Genetics of amyotrophic lateral sclerosis: A review. J Neurol Sci. 2019;399:217–226. doi: 10.1016/j.jns.2019.02.030. [DOI] [PubMed] [Google Scholar]
- 4.Nguyen HP, et al. ALS genes in the genomic era and their implications for FTD. Trends Genet. 2018;34(6):404–423. doi: 10.1016/j.tig.2018.03.001. [DOI] [PubMed] [Google Scholar]
- 5.Harms MM et al (1993) TARDBP-related amyotrophic lateral sclerosis. In: Adam MP (eds) GeneReviews [Internet]. University of Washington, Seattle, WA
- 6.Leblond CS, et al. Dissection of genetic factors associated with amyotrophic lateral sclerosis. Exp Neurol. 2014;262(Pt B):91–101. doi: 10.1016/j.expneurol.2014.04.013. [DOI] [PubMed] [Google Scholar]
- 7.Cady J, et al. Amyotrophic lateral sclerosis onset is influenced by the burden of rare variants in known amyotrophic lateral sclerosis genes. Ann Neurol. 2015;77(1):100–113. doi: 10.1002/ana.24306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Al-Chalabi A, et al. An estimate of amyotrophic lateral sclerosis heritability using twin data. J Neurol Neurosurg Psychiatry. 2010;81(12):1324–1326. doi: 10.1136/jnnp.2010.207464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Meltz Steinberg K, et al. Whole genome analyses reveal no pathogenetic single nucleotide or structural differences between monozygotic twins discordant for amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2015;16(5–6):385–392. doi: 10.3109/21678421.2015.1040029. [DOI] [PubMed] [Google Scholar]
- 10.Camu W, et al. Conjugal amyotrophic lateral sclerosis: a report on two couples from Southern France. Neurology. 1994;44(3 Pt 1):547–548. doi: 10.1212/wnl.44.3_part_1.547. [DOI] [PubMed] [Google Scholar]
- 11.Gunnarsson LG, Bodin L. Amyotrophic lateral sclerosis and occupational exposures: a systematic literature review and meta-analyses. Int J Environ Res Public Health. 2018;15(11):2371. doi: 10.3390/ijerph15112371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Andrew AS, et al. Environmental and occupational exposures and amyotrophic lateral sclerosis in New England. Neurodegener Dis. 2017;17(2–3):110–116. doi: 10.1159/000453359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Peters TL, et al. Blood levels of trace metals and amyotrophic lateral sclerosis. Neurotoxicology. 2016;54:119–126. doi: 10.1016/j.neuro.2016.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Figueroa-Romero C, et al. Early life metal dysregulation in amyotrophic lateral sclerosis. Ann Clin Transl Neurol. 2020;7(6):872–882. doi: 10.1002/acn3.51006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kang H, et al. Amyotrophic lateral sclerosis and agricultural environments: a systematic review. J Korean Med Sci. 2014;29(12):1610–1617. doi: 10.3346/jkms.2014.29.12.1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Institute of Medicine . Amyotrophic lateral sclerosis in veterans: review of the scientific literature. Washington, DC: The National Academies Press; 2006. [Google Scholar]
- 17.Beard JD, et al. Military service, deployments, and exposures in relation to amyotrophic lateral sclerosis etiology. Environ Int. 2016;91:104–115. doi: 10.1016/j.envint.2016.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bello A, et al. Retrospective assessment of occupational exposures for the GENEVA study of ALS among military veterans. Ann Work Expo Health. 2017;61(3):299–310. doi: 10.1093/annweh/wxw028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sagiraju HKR, et al. Amyotrophic lateral sclerosis among veterans deployed in support of Post-9/11 U.S. conflicts. Mil Med. 2020;185(3–4):e501–e509. doi: 10.1093/milmed/usz350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Seals RM, et al. Amyotrophic lateral sclerosis and the military: a population-based study in the Danish Registries. Epidemiology. 2016;27(2):188–193. doi: 10.1097/EDE.0000000000000417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Weisskopf MG, et al. Prospective study of military service and mortality from ALS. Neurology. 2005;64(1):32–37. doi: 10.1212/01.WNL.0000148649.17706.D9. [DOI] [PubMed] [Google Scholar]
- 22.Fang F, et al. Association between blood lead and the risk of amyotrophic lateral sclerosis. Am J Epidemiol. 2010;171(10):1126–1133. doi: 10.1093/aje/kwq063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Mason LH, et al. Pb neurotoxicity: neuropsychological effects of lead toxicity. Biomed Res Int. 2014;2014:840547–840547. doi: 10.1155/2014/840547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zeliger HI. Exposure to lipophilic chemicals as a cause of neurological impairments, neurodevelopmental disorders and neurodegenerative diseases. Interdiscip Toxicol. 2013;6(3):103–110. doi: 10.2478/intox-2013-0018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Birnbaum LS. The role of structure in the disposition of halogenated aromatic xenobiotics. Environ Health Perspect. 1985;61:11–20. doi: 10.1289/ehp.856111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Al-Chalabi A, et al. Analysis of amyotrophic lateral sclerosis as a multistep process: a population-based modelling study. Lancet Neurol. 2014;13(11):1108–1113. doi: 10.1016/S1474-4422(14)70219-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chiò A, et al. The multistep hypothesis of ALS revisited: the role of genetic mutations. Neurology. 2018;91(7):e635–e642. doi: 10.1212/WNL.0000000000005996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Horner RD, et al. Amyotrophic lateral sclerosis among 1991 Gulf War veterans: evidence for a time-limited outbreak. Neuroepidemiology. 2008;31(1):28–32. doi: 10.1159/000136648. [DOI] [PubMed] [Google Scholar]
- 29.Horner RD, et al. 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]
- 30.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]
- 31.McKay KA, et al. Military service and related risk factors for amyotrophic lateral sclerosis. Acta Neurol Scand. 2021;143(1):39–50. doi: 10.1111/ane.13345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Beard JD, Kamel F. Military service, deployments, and exposures in relation to amyotrophic lateral sclerosis etiology and survival. Epidemiol Rev. 2015;37(1):55–70. doi: 10.1093/epirev/mxu001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Department of Veterans Affairs Presumption of service connection for amyotrophic lateral sclerosis. Final rule. Federal Regist. 2009;74(212):57072–57074. [PubMed] [Google Scholar]
- 34.Committee on the Assessment of the Readjustment Needs of Military Personnel, V.T., Families Board on the Health of Select, Populations Institute of Medicine (2013) In: Returning home from Iraq and Afghanistan: assessment of readjustment needs of veterans, service members, and their families. National Academies Press, Washington, DC [PubMed]
- 35.Institute of Medicine . Health consequences of service during the Persian Gulf War: initial findings and recommendations for immediate action. Washington, DC: The National Academies Press; 1995. [PubMed] [Google Scholar]
- 36.Hyams KC, et al. Resolving disputes about toxicological risks during military conflict: the US Gulf War experience. Toxicol Rev. 2005;24(3):167–180. doi: 10.2165/00139709-200524030-00009. [DOI] [PubMed] [Google Scholar]
- 37.Hotopf M, Wessely S. Can epidemiology clear the fog of war? Lessons from the 1990–91 Gulf War. Int J Epidemiol. 2005;34(4):791–800. doi: 10.1093/ije/dyi102. [DOI] [PubMed] [Google Scholar]
- 38.Kang HK, et al. Mortality among US and UK veterans of the Persian Gulf War: a review. Occup Environ Med. 2002;59(12):794–799. doi: 10.1136/oem.59.12.794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Rose MR, Brix KA. Neurological disorders in Gulf War veterans. Philos Trans R Soc Lond B Biol Sci. 2006;361(1468):605–618. doi: 10.1098/rstb.2006.1820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Armon C. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology. 2004;62(6):1027–1029. doi: 10.1212/wnl.62.6.1027. [DOI] [PubMed] [Google Scholar]
- 41.Armon C. Occurrence of amyotrophic lateral sclerosis among Gulf War veterans. Neurology. 2007;68(13):1083–1083. doi: 10.1212/01.wnl.0000260429.80237.c0. [DOI] [PubMed] [Google Scholar]
- 42.Coffman CJ, et al. Estimating the occurrence of amyotrophic lateral sclerosis among Gulf War (1990–1991) veterans using capture-recapture methods. Neuroepidemiology. 2005;24(3):141–150. doi: 10.1159/000083297. [DOI] [PubMed] [Google Scholar]
- 43.Cox PA, et al. Cyanobacteria and BMAA exposure from desert dust: a possible link to sporadic ALS among Gulf War veterans. Amyotroph Lateral Scler. 2009;10(Suppl 2):109–117. doi: 10.3109/17482960903286066. [DOI] [PubMed] [Google Scholar]
- 44.Merwin SJ, et al. Organophosphate neurotoxicity to the voluntary motor system on the trail of environment-caused amyotrophic lateral sclerosis: the known, the misknown, and the unknown. Arch Toxicol. 2017;91(8):2939–2952. doi: 10.1007/s00204-016-1926-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dickerson AS, et al. Amyotrophic lateral sclerosis and exposure to diesel exhaust in a Danish cohort. Am J Epidemiol. 2018;187(8):1613–1622. doi: 10.1093/aje/kwy069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kurtzke JF, Beebe GW. Epidemiology of amyotrophic lateral sclerosis: 1. A case–control comparison based on ALS deaths. Neurology. 1980;30(5):453–462. doi: 10.1212/wnl.30.5.453. [DOI] [PubMed] [Google Scholar]
- 47.Pamphlett R, Rikard-Bell A. Different occupations associated with amyotrophic lateral sclerosis: is diesel exhaust the link? PLoS ONE. 2013;8(11):e80993. doi: 10.1371/journal.pone.0080993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fang F, et al. Workplace exposures and the risk of amyotrophic lateral sclerosis. Environ Health Perspect. 2009;117(9):1387–1392. doi: 10.1289/ehp.0900580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Park RM, et al. Potential occupational risks for neurodegenerative diseases. Am J Ind Med. 2005;48(1):63–77. doi: 10.1002/ajim.20178. [DOI] [PubMed] [Google Scholar]
- 50.IARC (1989) Diesel and gasoline engine exhausts and some nitroarenes, IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, International Agency for Research on Cancer, Lyon, France [PMC free article] [PubMed]
- 51.Steiner S, et al. Diesel exhaust: current knowledge of adverse effects and underlying cellular mechanisms. Arch Toxicol. 2016;90(7):1541–1553. doi: 10.1007/s00204-016-1736-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ehsanifar M, et al. Exposure to nanoscale diesel exhaust particles: oxidative stress, neuroinflammation, anxiety and depression on adult male mice. Ecotoxicol Environ Saf. 2019;168:338–347. doi: 10.1016/j.ecoenv.2018.10.090. [DOI] [PubMed] [Google Scholar]
- 53.Korotney D. A comprehensive analysis of biodiesel impacts on exhaust emissions. Washington, DC: Draft report of the US Environmental Protection Agency; 2002. [Google Scholar]
- 54.Chen H, et al. Living near major roads and the incidence of dementia, Parkinson’s disease, and multiple sclerosis: a population-based cohort study. Lancet. 2017;389(10070):718–726. doi: 10.1016/S0140-6736(16)32399-6. [DOI] [PubMed] [Google Scholar]
- 55.Shi L, et al. Long-term effects of PM(2·5) on neurological disorders in the American Medicare population: a longitudinal cohort study. Lancet Planet Health. 2020;4(12):e557–e565. doi: 10.1016/S2542-5196(20)30227-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Filippini T, et al. Risk of amyotrophic lateral sclerosis and exposure to particulate matter from vehicular traffic: a case–control study. Int J Environ Res Public Health. 2021;18(3):973. doi: 10.3390/ijerph18030973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nunez Y, et al. Fine particle exposure and clinical aggravation in neurodegenerative diseases in New York State. Environ Health Perspect. 2021;129(2):27003. doi: 10.1289/EHP7425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bendtsen KM, et al. A review of health effects associated with exposure to jet engine emissions in and around airports. Environ Health. 2021;20(1):10. doi: 10.1186/s12940-020-00690-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Nicholas JS, et al. Mortality among US commercial pilots and navigators. J Occup Environ Med. 1998;40(11):980–985. doi: 10.1097/00043764-199811000-00008. [DOI] [PubMed] [Google Scholar]
- 60.Bendtsen KM, et al. Airport emission particles: exposure characterization and toxicity following intratracheal instillation in mice. Part Fibre Toxicol. 2019;16(1):23. doi: 10.1186/s12989-019-0305-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Pinkerton LE, et al. Mortality from neurodegenerative diseases in a cohort of US flight attendants. Am J Ind Med. 2016;59(7):532–537. doi: 10.1002/ajim.22608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bonner JC. Nanoparticles as a potential cause of pleural and interstitial lung disease. Proc Am Thorac Soc. 2010;7(2):138–141. doi: 10.1513/pats.200907-061RM. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lu X, et al. Right or left: the role of nanoparticles in pulmonary diseases. Int J Mol Sci. 2014;15(10):17577–17600. doi: 10.3390/ijms151017577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Jonsdottir HR, et al. Non-volatile particle emissions from aircraft turbine engines at ground-idle induce oxidative stress in bronchial cells. Commun Biol. 2019;2:90. doi: 10.1038/s42003-019-0332-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Masiol M, Harrison RM. Aircraft engine exhaust emissions and other airport-related contributions to ambient air pollution: a review. Atmos Environ. 2014;95:409–455. doi: 10.1016/j.atmosenv.2014.05.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pleil JD, et al. Personal exposure to JP-8 jet fuel vapors and exhaust at air force bases. Environ Health Perspect. 2000;108(3):183–192. doi: 10.1289/ehp.00108183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Pugh MJ, et al. Increasing prevalence of chronic lung disease in veterans of the wars in Iraq and Afghanistan. Mil Med. 2016;181(5):476–481. doi: 10.7205/MILMED-D-15-00035. [DOI] [PubMed] [Google Scholar]
- 68.Fechter LD, et al. Subchronic JP-8 jet fuel exposure enhances vulnerability to noise-induced hearing loss in rats. J Toxicol Environ Health A. 2012;75(5):299–317. doi: 10.1080/15287394.2012.652060. [DOI] [PubMed] [Google Scholar]
- 69.Kaufman LR, et al. Effects of concurrent noise and jet fuel exposure on hearing loss. J Occup Environ Med. 2005;47(3):212–218. doi: 10.1097/01.jom.0000155710.28289.0e. [DOI] [PubMed] [Google Scholar]
- 70.Calderón-Garcidueñas L, et al. Increased gain in the auditory pathway, Alzheimer’s Disease Continuum, and air pollution: peripheral and central auditory system dysfunction evolves across pediatric and adult urbanites. J Alzheimers Dis. 2019;70(4):1275–1286. doi: 10.3233/JAD-190405. [DOI] [PubMed] [Google Scholar]
- 71.Griffith JD. Electron microscopic characterization of exhaust particles containing lead dibromide beads expelled from aircraft burning leaded gasoline. Atmos Pollut Res. 2020;11(9):1481–1486. doi: 10.1016/j.apr.2020.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Austin E, et al. Distinct ultrafine particle profiles associated with aircraft and roadway traffic. Environ Sci Technol. 2021;55(5):2847–2858. doi: 10.1021/acs.est.0c05933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pant P, Harrison RM. Estimation of the contribution of road traffic emissions to particulate matter concentrations from field measurements: a review. Atmos Environ. 2013;77:78–97. [Google Scholar]
- 74.Amato F, et al. Concentrations, sources and geochemistry of airborne particulate matter at a major European airport. J Environ Monit. 2010;12(4):854–862. doi: 10.1039/b925439k. [DOI] [PubMed] [Google Scholar]
- 75.Bennett M, et al. Composition of smoke generated by landing aircraft. Environ Sci Technol. 2011;45(8):3533–3538. doi: 10.1021/es1027585. [DOI] [PubMed] [Google Scholar]
- 76.Morris K (2006) An estimation of the tyre material erosion from measurements of aircraft. British Airways Environmental Affairs, British Airways Technical Documents Relating to the Aircraft Operations Supporting the Project for the Sustainable Development of Heathrow, Document 7
- 77.Consensus Study Report Long-term health consequences of exposure to burn pits in Iraq and Afghanistan. Mil Med. 2015;180(6):601–613. doi: 10.7205/MILMED-D-15-00039. [DOI] [PubMed] [Google Scholar]
- 78.Sharkey JM, et al. Postdeployment respiratory health care encounters following deployment to Kabul, Afghanistan: a retrospective cohort study. Mil Med. 2016;181(3):265–271. doi: 10.7205/MILMED-D-14-00690. [DOI] [PubMed] [Google Scholar]
- 79.Liu J, et al. Burn Pit emissions exposure and respiratory and cardiovascular conditions among airborne hazards and open burn pit registry participants. J Occup Environ Med. 2016;58(7):e249–e255. doi: 10.1097/JOM.0000000000000776. [DOI] [PubMed] [Google Scholar]
- 80.National Academies of Sciences E et al (2017) In: Butler DA et al (eds) Assessment of the Department of Veterans Affairs Airborne Hazards and Open Burn Pit Registry. National Academies Press, Washington, DC [PubMed]
- 81.IARC (1984) Polynuclear aromatic hydrocarbons. Part 3, Industrial exposures in aluminum production, coal gasification, coke production, and iron and steel founding. In: International Agency for Research on Cancer Monographs on the Carcinogenic Risk of Chemicals to Humans.
- 82.Miguel A, et al. On-road emissions of particulate polycyclic aromatic hydrocarbons and black carbon from gasoline and diesel vehicles. Environ Sci Technol. 1998;32(4):450–455. [Google Scholar]
- 83.Phillips D. Polycyclic aromatic hydrocarbons in the diet. Mutation Res. 1999;443(1–2):139–147. doi: 10.1016/s1383-5742(99)00016-2. [DOI] [PubMed] [Google Scholar]
- 84.Pedersen M, et al. Increased micronuclei and bulky DNA adducts in cord blood after maternal exposures to traffic-related air pollution. Environ Res. 2009;109:1012–1020. doi: 10.1016/j.envres.2009.08.011. [DOI] [PubMed] [Google Scholar]
- 85.Lorenz DR, et al. Acrolein and other toxicant exposures in relation to cardiovascular disease among marijuana and tobacco smokers in a longitudinal cohort of HIV-positive and negative adults. EClinicalMedicine. 2021;31:100697. doi: 10.1016/j.eclinm.2020.100697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bonner M, et al. Breast cancer risk and exposure in early life to polycyclic aromatic hydrocarbons using total suspended particulates as a proxy measure. Cancer Epidemiol Biomarkers Prev. 2005;14(1):53–60. [PubMed] [Google Scholar]
- 87.Obana H, et al. Polycyclic aromatic hydrocarbons in human fat and liver. Bull Environ Contam Toxicol. 1981;27:23–27. doi: 10.1007/BF01610981. [DOI] [PubMed] [Google Scholar]
- 88.Ambrosone C, Shields P (1997) Molecular epidemiology of breast cancer. In: Aldaz C, et al (eds) Etiology of breast and gynecological cancers. Progress in Clinical and Biologic Research. Wiley-Liss Inc, New York, pp 93–99 [PubMed]
- 89.Pastor-Belda M, et al. Bioaccumulation of polycyclic aromatic hydrocarbons for forensic assessment using gas chromatography–mass spectrometry. Chem Res Toxicol. 2019;32(8):1680–1688. doi: 10.1021/acs.chemrestox.9b00213. [DOI] [PubMed] [Google Scholar]
- 90.Santodonato J. Review of the estrogenic and antiestrogenic activity of polycyclic aromatic hydrocarbons: relationship to carcinogenicity. Chemosphere. 1997;34(4):835–848. doi: 10.1016/s0045-6535(97)00012-x. [DOI] [PubMed] [Google Scholar]
- 91.Kemp M, et al. Induction of the transferrin receptor gene by benzo[a]pyrene in breast cancer MCF-7 cells: potential as a biomarker of PAH exposure. Environ Mol Mutagen. 2006;47(7):518–526. doi: 10.1002/em.20221. [DOI] [PubMed] [Google Scholar]
- 92.Ralston S, et al. Stereoselective activation of dibenzo[a, l]pyrene and its trans-11,12-dihydrodiol to fjord region 11,12-diol 13,14-epoxides in a human mammary carcinoma MCF-7 cell-mediated V79 cell mutation assay. Chem Res Toxicol. 1997;10:687–693. doi: 10.1021/tx9700275. [DOI] [PubMed] [Google Scholar]
- 93.Valand R, et al. Gene expression changes in rat brain regions after 7- and 28 days inhalation exposure to exhaust emissions from 1st and 2nd generation biodiesel fuels—the FuelHealth project. Inhal Toxicol. 2018;30(7–8):299–312. doi: 10.1080/08958378.2018.1520370. [DOI] [PubMed] [Google Scholar]
- 94.Saunders CR, et al. Benzo(a)pyrene-induced acute neurotoxicity in the F-344 rat: role of oxidative stress. J Appl Toxicol. 2006;26(5):427–438. doi: 10.1002/jat.1157. [DOI] [PubMed] [Google Scholar]
- 95.Dutta K, et al. A common carcinogen benzo[a]pyrene causes neuronal death in mouse via microglial activation. PLoS ONE. 2010;5(4):e9984. doi: 10.1371/journal.pone.0009984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Phani S, et al. The role of the innate immune system in ALS. Front Pharmacol. 2012;3:150. doi: 10.3389/fphar.2012.00150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Das SK, et al. Chronic waterborne exposure to benzo[a]pyrene induces locomotor dysfunction and development of neurodegenerative phenotypes in zebrafish. Neurosci Lett. 2020;716:134646. doi: 10.1016/j.neulet.2019.134646. [DOI] [PubMed] [Google Scholar]
- 98.Ohura T, et al. Polycyclic aromatic hydrocarbons in indoor and outdoor environments and factors affecting their concentrations. Environ Sci Technol. 2004;38(1):77–83. doi: 10.1021/es030512o. [DOI] [PubMed] [Google Scholar]
- 99.Kamel F, et al. Pesticide exposure and amyotrophic lateral sclerosis. Neurotoxicology. 2012;33(3):457–462. doi: 10.1016/j.neuro.2012.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Goutman SA, et al. High plasma concentrations of organic pollutants negatively impact survival in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry. 2019;90(8):907–912. doi: 10.1136/jnnp-2018-319785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Srogi K. Levels and congener distributions of PCDDs, PCDFs and dioxin-like PCBs in environmental and human samples: a review. Environ Chem Lett. 2008;6(1):1–28. [Google Scholar]
- 102.Ness DK, et al. PCB congeners in the rat brain: selective accumulation and lack of regionalization. J Toxicol Environ Health. 1994;43(4):453–468. doi: 10.1080/15287399409531934. [DOI] [PubMed] [Google Scholar]
- 103.Liberman DA, et al. Sex-specific effects of developmental exposure to polychlorinated biphenyls on neuroimmune and dopaminergic endpoints in adolescent rats. Neurotoxicol Teratol. 2020;79:106880. doi: 10.1016/j.ntt.2020.106880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Bell MR, et al. Sex differences in effects of gestational polychlorinated biphenyl exposure on hypothalamic neuroimmune and neuromodulator systems in neonatal rats. Toxicol Appl Pharmacol. 2018;353:55–66. doi: 10.1016/j.taap.2018.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Manjaly ZR, et al. The sex ratio in amyotrophic lateral sclerosis: a population based study. Amyotroph Lateral Scler. 2010;11(5):439–442. doi: 10.3109/17482961003610853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bouchard MF, et al. Polychlorinated biphenyl exposures and cognition in older US adults: NHANES (1999–2002) Environ Health Perspect. 2014;122(1):73–78. doi: 10.1289/ehp.1306532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Su FC, et al. Association of environmental toxins with amyotrophic lateral sclerosis. JAMA Neurol. 2016;73(7):803–811. doi: 10.1001/jamaneurol.2016.0594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ruder AM, et al. Mortality among 24,865 workers exposed to polychlorinated biphenyls (PCBs) in three electrical capacitor manufacturing plants: a ten-year update. Int J Hyg Environ Health. 2014;217(2–3):176–187. doi: 10.1016/j.ijheh.2013.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hatcher JM, et al. Parkinson's disease and pesticides: a toxicological perspective. Trends Pharmacol Sci. 2008;29(6):322–329. doi: 10.1016/j.tips.2008.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Weisskopf MG, et al. Persistent organochlorine pesticides in serum and risk of Parkinson disease. Neurology. 2010;74(13):1055–1061. doi: 10.1212/WNL.0b013e3181d76a93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Rhodes SL, et al. Pesticides that inhibit the ubiquitin-proteasome system: effect measure modification by genetic variation in SKP1 in Parkinson’s disease. Environ Res. 2013;126:1–8. doi: 10.1016/j.envres.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Steenland K, et al. Occupational pesticide exposure and screening tests for neurodegenerative disease among an elderly population in Costa Rica. Environ Res. 2013;120:96–101. doi: 10.1016/j.envres.2012.08.014. [DOI] [PubMed] [Google Scholar]
- 113.Richardson JR, et al. Elevated serum pesticide levels and risk of Parkinson disease. Arch Neurol. 2009;66(7):870–875. doi: 10.1001/archneurol.2009.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Richardson JR, et al. β-Hexachlorocyclohexane levels in serum and risk of Parkinson’s disease. Neurotoxicology. 2011;32(5):640–645. doi: 10.1016/j.neuro.2011.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Corrigan FM, et al. Organochlorine compounds in human brain. Hum Exp Toxicol. 1996;15(3):262–264. doi: 10.1177/096032719601500314. [DOI] [PubMed] [Google Scholar]
- 116.Corrigan FM, et al. Organochlorine insecticides in substantia nigra in Parkinson’s disease. J Toxicol Environ Health A. 2000;59(4):229–234. doi: 10.1080/009841000156907. [DOI] [PubMed] [Google Scholar]
- 117.Hatcher JM, et al. Dieldrin exposure induces oxidative damage in the mouse nigrostriatal dopamine system. Exp Neurol. 2007;204(2):619–630. doi: 10.1016/j.expneurol.2006.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hatcher JM, et al. Disruption of dopamine transport by DDT and its metabolites. Neurotoxicology. 2008;29(4):682–690. doi: 10.1016/j.neuro.2008.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Bellou E, et al. Polygenic risk and pleiotropy in neurodegenerative diseases. Neurobiol Dis. 2020;142:104953. doi: 10.1016/j.nbd.2020.104953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Pang SY, et al. The role of gene variants in the pathogenesis of neurodegenerative disorders as revealed by next generation sequencing studies: a review. Transl Neurodegener. 2017;6:27. doi: 10.1186/s40035-017-0098-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Bradley WG, et al. Gene–environment–-time interactions in neurodegenerative diseases: hypotheses and research approaches. Ann Neurosci. 2018;25(4):261–267. doi: 10.1159/000495321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Nghi TN, et al. Dioxins and Nonortho PCBs in breast milk of vietnamese mothers living in the largest hot spot of dioxin contamination. Environ Sci Technol. 2015;49(9):5732–5742. doi: 10.1021/es506211p. [DOI] [PubMed] [Google Scholar]
- 123.Khan K, et al. Wartime toxin exposure: recognising the silent killer. BMJ Case Rep. 2016 doi: 10.1136/bcr-2016-217438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Humphrey KM, et al. Establishing a role for environmental toxicant exposure induced epigenetic remodeling in malignant transformation. Semin Cancer Biol. 2019;57:86–94. doi: 10.1016/j.semcancer.2018.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hill A, et al. Neurodevelopmental defects in zebrafish (Danio rerio) at environmentally relevant dioxin (TCDD) concentrations. Toxicol Sci. 2003;76(2):392–399. doi: 10.1093/toxsci/kfg241. [DOI] [PubMed] [Google Scholar]
- 126.Xu G, et al. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced inflammatory activation is mediated by intracellular free calcium in microglial cells. Toxicology. 2013;308:158–167. doi: 10.1016/j.tox.2013.04.002. [DOI] [PubMed] [Google Scholar]
- 127.Xie HQ, et al. AhR-mediated effects of dioxin on neuronal acetylcholinesterase expression in vitro. Environ Health Perspect. 2013;121(5):613–618. doi: 10.1289/ehp.1206066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Del Pino Sans J, et al. Developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin may alter LH release patterns by abolishing sex differences in GABA/glutamate cell number and modifying the transcriptome of the male anteroventral periventricular nucleus. Neuroscience. 2016;329:239–253. doi: 10.1016/j.neuroscience.2016.04.051. [DOI] [PubMed] [Google Scholar]
- 129.Gohlke JM, et al. AhR-mediated gene expression in the developing mouse telencephalon. Reprod Toxicol. 2009;28(3):321–328. doi: 10.1016/j.reprotox.2009.05.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Kimura E, et al. In utero and lactational dioxin exposure induces Sema3b and Sema3g gene expression in the developing mouse brain. Biochem Biophys Res Commun. 2016;476(2):108–113. doi: 10.1016/j.bbrc.2016.05.048. [DOI] [PubMed] [Google Scholar]
- 131.Li Y, et al. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces microglial nitric oxide production and subsequent rat primary cortical neuron apoptosis through p38/JNK MAPK pathway. Toxicology. 2013;312:132–141. doi: 10.1016/j.tox.2013.08.008. [DOI] [PubMed] [Google Scholar]
- 132.Xu G, et al. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces expression of p27(kip1) and FoxO3a in female rat cerebral cortex and PC12 cells. Toxicol Lett. 2014;226(3):294–302. doi: 10.1016/j.toxlet.2014.02.019. [DOI] [PubMed] [Google Scholar]
- 133.Amenya HZ, et al. Dioxin induces Ahr-dependent robust DNA demethylation of the Cyp1a1 promoter via Tdg in the mouse liver. Sci Rep. 2016;6:34989. doi: 10.1038/srep34989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Ash PEA, et al. Dioxins and related environmental contaminants increase TDP-43 levels. Mol Neurodegener. 2017;12(1):35. doi: 10.1186/s13024-017-0177-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Karri V, et al. Differential protein expression of hippocampal cells associated with heavy metals (Pb, As, and MeHg) neurotoxicity: deepening into the molecular mechanism of neurodegenerative diseases. J Proteomics. 2018;187:106–125. doi: 10.1016/j.jprot.2018.06.020. [DOI] [PubMed] [Google Scholar]
- 136.De Benedetti S, et al. Blood trace metals in a sporadic amyotrophic lateral sclerosis geographical cluster. Biometals. 2017;30(3):355–365. doi: 10.1007/s10534-017-0011-4. [DOI] [PubMed] [Google Scholar]
- 137.Roos PM, et al. Metal concentrations in cerebrospinal fluid and blood plasma from patients with amyotrophic lateral sclerosis. Biol Trace Elem Res. 2013;151(2):159–170. doi: 10.1007/s12011-012-9547-x. [DOI] [PubMed] [Google Scholar]
- 138.Callaghan B, et al. The association of exposure to lead, mercury, and selenium and the development of amyotrophic lateral sclerosis and the epigenetic implications. Neurodegener Dis. 2011;8(1–2):1–8. doi: 10.1159/000315405. [DOI] [PubMed] [Google Scholar]
- 139.Trojsi F, et al. Exposure to environmental toxicants and pathogenesis of amyotrophic lateral sclerosis: state of the art and research perspectives. Int J Mol Sci. 2013;14(8):15286–15311. doi: 10.3390/ijms140815286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cicero CE, et al. Metals and neurodegenerative diseases. A systematic review. Environ Res. 2017;159:82–94. doi: 10.1016/j.envres.2017.07.048. [DOI] [PubMed] [Google Scholar]
- 141.Oggiano R, et al. Trace elements in ALS patients and their relationships with clinical severity. Chemosphere. 2018;197:457–466. doi: 10.1016/j.chemosphere.2018.01.076. [DOI] [PubMed] [Google Scholar]
- 142.Andrew AS, et al. Toenail mercury levels are associated with amyotrophic lateral sclerosis risk. Muscle Nerve. 2018 doi: 10.1002/mus.26055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Andrew AS, et al. Keratinous biomarker of mercury exposure associated with amyotrophic lateral sclerosis risk in a nationwide U.S. study. Amyotroph Lateral Scler Frontotemporal. 2020;21:421–427. doi: 10.1080/21678421.2020.1753777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Andrew AS, et al. Risk factors for amyotrophic lateral sclerosis: a regional United States case–control study. Muscle Nerve. 2021;63(1):52–59. doi: 10.1002/mus.27085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Kamel F, et al. Lead exposure as a risk factor for amyotrophic lateral sclerosis. Neurodegener Dis. 2005;2(3–4):195–201. doi: 10.1159/000089625. [DOI] [PubMed] [Google Scholar]
- 146.Ingre C, et al. Risk factors for amyotrophic lateral sclerosis. Clin Epidemiol. 2015;7:181–193. doi: 10.2147/CLEP.S37505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wang MD, et al. Identification of risk factors associated with onset and progression of amyotrophic lateral sclerosis using systematic review and meta-analysis. Neurotoxicology. 2017;61:101–130. doi: 10.1016/j.neuro.2016.06.015. [DOI] [PubMed] [Google Scholar]
- 148.Belbasis L, et al. Environmental risk factors and amyotrophic lateral sclerosis: an umbrella review and critical assessment of current evidence from systematic reviews and meta-analyses of observational studies. Neuroepidemiology. 2016;46(2):96–105. doi: 10.1159/000443146. [DOI] [PubMed] [Google Scholar]
- 149.Neal AP, Guilarte TR. Mechanisms of lead and manganese neurotoxicity. Toxicol Res (Camb) 2013;2(2):99–114. doi: 10.1039/C2TX20064C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Ash PEA, et al. Heavy metal neurotoxicants induce ALS-linked TDP-43 pathology. Toxicol Sci. 2019;167(1):105–115. doi: 10.1093/toxsci/kfy267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Kasperczyk S, et al. The effect of occupational lead exposure on lipid peroxidation, protein carbonylation, and plasma viscosity. Toxicol Ind Health. 2015;31(12):1165–1171. doi: 10.1177/0748233713491804. [DOI] [PubMed] [Google Scholar]
- 152.Laidlaw MAS, et al. Lead exposure at firing ranges—a review. Environ Health. 2017;16(1):34–34. doi: 10.1186/s12940-017-0246-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Dickerson AS, et al. Population-based study of amyotrophic lateral sclerosis and occupational lead exposure in Denmark. Occup Environ Med. 2019;76(4):208–214. doi: 10.1136/oemed-2018-105469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Zahran S, et al. Motor neuron disease mortality and lifetime petrol lead exposure: evidence from national age-specific and state-level age-standardized death rates in Australia. Environ Res. 2017;153:181–190. doi: 10.1016/j.envres.2016.11.023. [DOI] [PubMed] [Google Scholar]
- 155.Khare SS, et al. Trace element imbalances in amyotrophic lateral sclerosis. Neurotoxicology. 1990;11(3):521–532. [PubMed] [Google Scholar]
- 156.Zufiría M, et al. ALS: A bucket of genes, environment, metabolism and unknown ingredients. Prog Neurobiol. 2016;142:104–129. doi: 10.1016/j.pneurobio.2016.05.004. [DOI] [PubMed] [Google Scholar]
- 157.Adams CR, et al. Mercury intoxication simulating amyotrophic lateral sclerosis. JAMA. 1983;250(5):642–643. [PubMed] [Google Scholar]
- 158.Praline J, et al. ALS and mercury intoxication: a relationship? Clin Neurol Neurosurg. 2007;109(10):880–883. doi: 10.1016/j.clineuro.2007.07.008. [DOI] [PubMed] [Google Scholar]
- 159.Mangelsdorf I, et al. Healing of amyotrophic lateral sclerosis: a case report. Complement Med Res. 2017;24(3):175–181. doi: 10.1159/000477397. [DOI] [PubMed] [Google Scholar]
- 160.Arvidson B. Accumulation of inorganic mercury in lower motoneurons of mice. Neurotoxicology. 1992;13(1):277–280. [PubMed] [Google Scholar]
- 161.Chuu J-J, et al. Differential neurotoxic effects of methylmercury and mercuric sulfide in rats. Toxicol Lett. 2007;169(2):109–120. doi: 10.1016/j.toxlet.2006.12.006. [DOI] [PubMed] [Google Scholar]
- 162.Farina M, et al. Mechanisms of methylmercury-induced neurotoxicity: evidence from experimental studies. Life Sci. 2011;89(15–16):555–563. doi: 10.1016/j.lfs.2011.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Mitchell JD, et al. Trace elements in the spinal cord and other tissues in motor neuron disease. J Neurol Neurosurg Psychiatry. 1986;49(2):211–215. doi: 10.1136/jnnp.49.2.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Estevez AO, et al. Selenium induces cholinergic motor neuron degeneration in Caenorhabditis elegans. Neurotoxicology. 2012;33(5):1021–1032. doi: 10.1016/j.neuro.2012.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Vinceti M, et al. Are environmental exposures to selenium, heavy metals, and pesticides risk factors for amyotrophic lateral sclerosis? Rev Environ Health. 2012;27(1):19–41. doi: 10.1515/reveh-2012-0002. [DOI] [PubMed] [Google Scholar]
- 166.Vinceti M, et al. Cerebrospinal fluid of newly diagnosed amyotrophic lateral sclerosis patients exhibits abnormal levels of selenium species including elevated selenite. Neurotoxicology. 2013;38:25–32. doi: 10.1016/j.neuro.2013.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Bilen B, et al. Examination of metal mobilization from a gunshot by scanning acoustic microscopy, scanning electron microscopy, energy-dispersive X-ray spectroscopy, and inductively coupled plasma optical emission spectroscopy: a case report. J Med Case Reports. 2018;12(1):391. doi: 10.1186/s13256-018-1905-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Kilness AW, Hichberg FH. Amyotrophic lateral sclerosis in a high selenium environment. JAMA. 1977;237(26):2843–2844. [PubMed] [Google Scholar]
- 169.Vinceti M, et al. Amyotrophic lateral sclerosis after long-term exposure to drinking water with high selenium content. Epidemiology. 1996;7(5):529–532. [PubMed] [Google Scholar]
- 170.Markesbery WR, et al. Neutron activation analysis of trace elements in motor neuron disease spinal cord. Neurodegeneration. 1995;4(4):383–390. doi: 10.1006/neur.1995.0046. [DOI] [PubMed] [Google Scholar]
- 171.Moriwaka F, et al. Mercury and selenium contents in amyotrophic lateral sclerosis in Hokkaido, the northernmost island of Japan. J Neurol Sci. 1993;118(1):38–42. doi: 10.1016/0022-510x(93)90243-r. [DOI] [PubMed] [Google Scholar]
- 172.Gribble MO, et al. Mercury, selenium and fish oils in marine food webs and implications for human health. J Mar Biol Assoc U K. 2016;96(1):43–59. doi: 10.1017/S0025315415001356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ralston NV. Selenium health benefit values as seafood safety criteria. EcoHealth. 2008;5(4):442–455. doi: 10.1007/s10393-008-0202-0. [DOI] [PubMed] [Google Scholar]
- 174.Bergomi M, et al. 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. [DOI] [PubMed] [Google Scholar]
- 175.Dang TN, et al. Increased metal content in the TDP-43(A315T) transgenic mouse model of frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Front Aging Neurosci. 2014;6:15. doi: 10.3389/fnagi.2014.00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Lovejoy DB, Guillemin GJ. The potential for transition metal-mediated neurodegeneration in amyotrophic lateral sclerosis. Front Aging Neurosci. 2014;6:173. doi: 10.3389/fnagi.2014.00173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Choi BS, Zheng W. Copper transport to the brain by the blood–brain barrier and blood-CSF barrier. Brain Res. 2009;1248:14–21. doi: 10.1016/j.brainres.2008.10.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Guilarte TR. Manganese and Parkinson's disease: a critical review and new findings. Environ Health Perspect. 2010;118(8):1071–1080. doi: 10.1289/ehp.0901748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wiśniewska-Hejka Z, et al. Neurological examinations of workers with chronic exposure to manganese dioxide during the production of piles and batteries. Neurol Neurochir Pol. 1978;12(4):435–441. [PubMed] [Google Scholar]
- 180.Voss H. Progressive bulbar paralysis and amyotrophie lateral sclerosis following chronic manganese poisoning. Arch Gewerbepath Gewerbehyg. 1939;9:464–476. [Google Scholar]
- 181.Yanagihara R. Heavy metals and essential minerals in motor neuron disease. Adv Neurol. 1982;36:233–247. [PubMed] [Google Scholar]
- 182.Miyata S, et al. Increased manganese level in spinal cords of amyotrophic lateral sclerosis determined by radiochemical neutron activation analysis. J Neurol Sci. 1983;61(2):283–293. doi: 10.1016/0022-510x(83)90012-6. [DOI] [PubMed] [Google Scholar]
- 183.Hozumi I, et al. Patterns of levels of biological metals in CSF differ among neurodegenerative diseases. J Neurol Sci. 2011;303(1–2):95–99. doi: 10.1016/j.jns.2011.01.003. [DOI] [PubMed] [Google Scholar]
- 184.Roos PM, et al. Manganese in cerebrospinal fluid and blood plasma of patients with amyotrophic lateral sclerosis. Exp Biol Med (Maywood) 2012;237(7):803–810. doi: 10.1258/ebm.2012.011396. [DOI] [PubMed] [Google Scholar]
- 185.Kasarskis EJ, et al. Aluminum, calcium, and iron in the spinal cord of patients with sporadic amyotrophic lateral sclerosis using laser microprobe mass spectroscopy: a preliminary study. J Neurol Sci. 1995;130(2):203–208. doi: 10.1016/0022-510x(95)00037-3. [DOI] [PubMed] [Google Scholar]
- 186.Jeong SY, et al. Dysregulation of iron homeostasis in the CNS contributes to disease progression in a mouse model of amyotrophic lateral sclerosis. J Neurosci. 2009;29(3):610–619. doi: 10.1523/JNEUROSCI.5443-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kwan JY, et al. Iron accumulation in deep cortical layers accounts for MRI signal abnormalities in ALS: correlating 7 tesla MRI and pathology. PLoS ONE. 2012;7(4):e35241. doi: 10.1371/journal.pone.0035241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Wang Q, et al. Prevention of motor neuron degeneration by novel iron chelators in SOD1(G93A) transgenic mice of amyotrophic lateral sclerosis. Neurodegener Dis. 2011;8(5):310–321. doi: 10.1159/000323469. [DOI] [PubMed] [Google Scholar]
- 189.Vahidnia A, et al. Arsenic neurotoxicity—a review. Hum Exp Toxicol. 2007;26(10):823–832. doi: 10.1177/0960327107084539. [DOI] [PubMed] [Google Scholar]
- 190.Mazumdar S, et al. Arsenicosis: unusual neurological presentation from West Bengal. J Assoc Physicians India. 2010;58:201–202. [PubMed] [Google Scholar]
- 191.Tyler CR, Allan AM. The effects of arsenic exposure on neurological and cognitive dysfunction in human and rodent studies: a review. Curr Environ Health Rep. 2014;1(2):132–147. doi: 10.1007/s40572-014-0012-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Dubey M, Shea TB. Potentiation of arsenic neurotoxicity by folate deprivation: protective role of S-adenosyl methionine. Nutr Neurosci. 2007;10(5–6):199–204. doi: 10.1080/10284150701562703. [DOI] [PubMed] [Google Scholar]
- 193.Goh CW, et al. Chronic oxidative stress promotes GADD34-mediated phosphorylation of the TAR DNA-binding protein TDP-43, a modification linked to neurodegeneration. J Biol Chem. 2018;293(1):163–176. doi: 10.1074/jbc.M117.814111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Lukiw WJ, et al. Aluminum in neurological disease—a 36 year multicenter study. J Alzheimers Dis Parkinsonism. 2019;8(6):457. doi: 10.4172/2161-0460.1000457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Kanias GD, Kapaki E. Trace elements, age, and sex in amyotrophic lateral sclerosis disease. Biol Trace Elem Res. 1997;56(2):187–201. doi: 10.1007/BF02785392. [DOI] [PubMed] [Google Scholar]
- 196.Kapaki E, et al. Zinc, copper and magnesium concentration in serum and CSF of patients with neurological disorders. Acta Neurol Scand. 1989;79(5):373–378. doi: 10.1111/j.1600-0404.1989.tb03803.x. [DOI] [PubMed] [Google Scholar]
- 197.Bar-Sela S, et al. Amyotrophic lateral sclerosis in a battery-factory worker exposed to cadmium. Int J Occup Environ Health. 2001;7(2):109–112. doi: 10.1179/107735201800339470. [DOI] [PubMed] [Google Scholar]
- 198.Vinceti M, et al. Lead, cadmium and mercury in cerebrospinal fluid and risk of amyotrophic lateral sclerosis: a case–control study. J Trace Elem Med Biol. 2017;43:121–125. doi: 10.1016/j.jtemb.2016.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Dickerson AS, et al. Study of occupational chromium, iron, and nickel exposure and amyotrophic lateral sclerosis in Denmark. Int J Environ Res Public Health. 2020;17(21):8086. doi: 10.3390/ijerph17218086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Bourke CA. Molybdenum deficiency produces motor nervous effects that are consistent with amyotrophic lateral sclerosis. Front Neurol. 2016;7:28. doi: 10.3389/fneur.2016.00028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Grochowski C, et al. Analysis of trace elements in human brain: its aim, methods, and concentration levels. Front Chem. 2019;7:115. doi: 10.3389/fchem.2019.00115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Gellein K, et al. Concentrations of Cd Co, Cu, Fe, Mn, Rb, V, and Zn in formalin-fixed brain tissue in amyotrophic lateral sclerosis and Parkinsonism-dementia complex of Guam determined by High-resolution ICP-MS. Biol Trace Elem Res. 2003;96(1–3):39–60. doi: 10.1385/BTER:96:1-3:39. [DOI] [PubMed] [Google Scholar]
- 203.Huat TJ, et al. Metal toxicity links to Alzheimer's disease and neuroinflammation. J Mol Biol. 2019;431(9):1843–1868. doi: 10.1016/j.jmb.2019.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Waterman SJ, et al. Lead alters the immunogenicity of two neural proteins: a potential mechanism for the progression of lead-induced neurotoxicity. Environ Health Perspect. 1994;102(12):1052–1056. doi: 10.1289/ehp.941021052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Nakagawa Y, Yamada S. A novel hypothesis on metal dyshomeostasis and mitochondrial dysfunction in amyotrophic lateral sclerosis: potential pathogenetic mechanism and therapeutic implications. Eur J Pharmacol. 2021;892:173737. doi: 10.1016/j.ejphar.2020.173737. [DOI] [PubMed] [Google Scholar]
- 206.Hadjidemetriou M, Kostarelos K. Nanomedicine: evolution of the nanoparticle corona. Nat Nanotechnol. 2017;12(4):288–290. doi: 10.1038/nnano.2017.61. [DOI] [PubMed] [Google Scholar]
- 207.Cedervall T, et al. Understanding the nanoparticle-protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles. Proc Natl Acad Sci USA. 2007;104(7):2050–2055. doi: 10.1073/pnas.0608582104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Shang L, et al. Engineered nanoparticles interacting with cells: size matters. J Nanobiotechnology. 2014;12:5. doi: 10.1186/1477-3155-12-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Sawicki K, et al. Toxicity of metallic nanoparticles in the central nervous system. Nanotechnol Rev. 2019;8(1):175–200. [Google Scholar]
- 210.Salvati A, et al. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nat Nanotechnol. 2013;8(2):137–143. doi: 10.1038/nnano.2012.237. [DOI] [PubMed] [Google Scholar]
- 211.Gatoo MA, et al. Physicochemical properties of nanomaterials: implication in associated toxic manifestations. BioMed Res Int. 2014 doi: 10.1155/2014/498420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Aillon KL, et al. Effects of nanomaterial physicochemical properties on in vivo toxicity. Adv Drug Deliv Rev. 2009;61(6):457–466. doi: 10.1016/j.addr.2009.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Chen X, Schluesener HJ. Nanosilver: a nanoproduct in medical application. Toxicol Lett. 2008;176(1):1–12. doi: 10.1016/j.toxlet.2007.10.004. [DOI] [PubMed] [Google Scholar]
- 214.Ahamed M, et al. Silver nanoparticle applications and human health. Clin Chim Acta. 2010;411(23–24):1841–1848. doi: 10.1016/j.cca.2010.08.016. [DOI] [PubMed] [Google Scholar]
- 215.Singh AK. Engineered nanoparticles: structure, properties and mechanisms of toxicity. New York: Academic Press; 2015. [Google Scholar]
- 216.Khan I, et al. Nanoparticles: properties, applications and toxicities. Arab J Chem. 2019;12(7):908–931. [Google Scholar]
- 217.Zhang XQ, et al. Interactions of nanomaterials and biological systems: implications to personalized nanomedicine. Adv Drug Deliv Rev. 2012;64(13):1363–1384. doi: 10.1016/j.addr.2012.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Jeevanandam J, et al. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050–1074. doi: 10.3762/bjnano.9.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Bundschuh M, et al. Nanoparticles in the environment: where do we come from, where do we go to? Environ Sci Eur. 2018;30(1):6–6. doi: 10.1186/s12302-018-0132-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Teleanu DM, et al. Impact of nanoparticles on brain health: an up to date overview. J Clin Med. 2018;7(12):490. doi: 10.3390/jcm7120490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Calderón-Garcidueñas L, et al. Quadruple abnormal protein aggregates in brainstem pathology and exogenous metal-rich magnetic nanoparticles. The substantia nigrae is a very early target in young urbanites and the gastrointestinal tract likely a key brainstem portal. Environ Res. 2020;191:110139. doi: 10.1016/j.envres.2020.110139. [DOI] [PubMed] [Google Scholar]
- 222.Oberdörster G, et al. Translocation of inhaled ultrafine particles to the brain. Inhal Toxicol. 2004;16(6–7):437–445. doi: 10.1080/08958370490439597. [DOI] [PubMed] [Google Scholar]
- 223.Fröhlich E, Salar-Behzadi S. Toxicological assessment of inhaled nanoparticles: role of in vivo, ex vivo, in vitro, and in silico studies. Int J Mol Sci. 2014;15(3):4795–4822. doi: 10.3390/ijms15034795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.De Matteis V. Exposure to inorganic nanoparticles: routes of entry, immune response, biodistribution and in vitro/in vivo toxicity evaluation. Toxics. 2017;5(4):29. doi: 10.3390/toxics5040029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Gao H. Progress and perspectives on targeting nanoparticles for brain drug delivery. Acta Pharm Sin B. 2016;6(4):268–286. doi: 10.1016/j.apsb.2016.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Busquets MA, et al. Magnetic nanoparticles cross the blood-brain barrier: when physics rises to a challenge. Nanomaterials (Basel) 2015;5(4):2231–2248. doi: 10.3390/nano5042231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Zhou Y, et al. Crossing the blood–brain barrier with nanoparticles. J Control Release. 2018;270:290–303. doi: 10.1016/j.jconrel.2017.12.015. [DOI] [PubMed] [Google Scholar]
- 228.Lombardo SM, et al. Key for crossing the BBB with nanoparticles: the rational design. Beilstein J Nanotechnol. 2020;11:866–883. doi: 10.3762/bjnano.11.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Garcia GJM, et al. Olfactory deposition of inhaled nanoparticles in humans. Inhalation Toxicol. 2015;27(8):394–403. doi: 10.3109/08958378.2015.1066904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Pardeshi CV, Belgamwar VS. Direct nose to brain drug delivery via integrated nerve pathways bypassing the blood–brain barrier: an excellent platform for brain targeting. Expert Opin Drug Deliv. 2013;10(7):957–972. doi: 10.1517/17425247.2013.790887. [DOI] [PubMed] [Google Scholar]
- 231.Elder A, et al. Translocation of inhaled ultrafine manganese oxide particles to the central nervous system. Environ Health Perspect. 2006;114(8):1172–1178. doi: 10.1289/ehp.9030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Sutunkova M, et al. Toxic effects of low-level long-term inhalation exposures of rats to nickel oxide nanoparticles. Int J Mol Sci. 2019;20:1778. doi: 10.3390/ijms20071778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Riediker M, et al. Particle toxicology and health—where are we? Part Fibre Toxicol. 2019;16(1):19. doi: 10.1186/s12989-019-0302-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Lewinski N, et al. Cytotoxicity of nanoparticles. Small. 2008;4(1):26–49. doi: 10.1002/smll.200700595. [DOI] [PubMed] [Google Scholar]
- 235.Zoroddu MA, et al. Toxicity of nanoparticles. Curr Med Chem. 2014;21(33):3837–3853. doi: 10.2174/0929867321666140601162314. [DOI] [PubMed] [Google Scholar]
- 236.Li J, Ju D. Neurotoxicity of nanomaterials and nanomedicine. New York: Academic Press; 2017. [Google Scholar]
- 237.Kumar V et al (2018) Chapter 17—impact of nanoparticles on oxidative stress and responsive antioxidative defense in plants. In: Tripathi DK et al (eds) Nanomaterials in plants, algae, and microorganisms. Academic Press, New York, pp 393–406
- 238.Zuberek M, Grzelak A. Nanoparticles-caused oxidative imbalance. Adv Exp Med Biol. 2018;1048:85–98. doi: 10.1007/978-3-319-72041-8_6. [DOI] [PubMed] [Google Scholar]
- 239.Mauricio MD, et al. Nanoparticles in medicine: a focus on vascular oxidative stress. Oxid Med Cell Longev. 2018;2018:6231482–6231482. doi: 10.1155/2018/6231482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Ganguly R et al (2019) Chapter 3—nanoparticles as modulators of oxidative stress. In: Maurya PK, Singh S (eds) Nanotechnology in modern animal biotechnology. Elsevier, Amsterdam, pp 29–35
- 241.Lovisolo D, et al. Nanoparticles and potential neurotoxicity: focus on molecular mechanisms. AIMS Mol Sci. 2018;5:1–13. [Google Scholar]
- 242.Grissa I, et al. The effect of titanium dioxide nanoparticles on neuroinflammation response in rat brain. Environ Sci Pollut Res Int. 2016;23(20):20205–20213. doi: 10.1007/s11356-016-7234-8. [DOI] [PubMed] [Google Scholar]
- 243.Kim B-G, et al. Effects of nanoparticles on neuroinflammation in a mouse model of asthma. Respir Physiol Neurobiol. 2020;271:103292. doi: 10.1016/j.resp.2019.103292. [DOI] [PubMed] [Google Scholar]
- 244.Liang H, et al. Neuroinflammation is induced by tongue-instilled ZnO nanoparticles via the Ca2+-dependent NF-κB and MAPK pathways. Part Fibre Toxicol. 2018;15(1):39. doi: 10.1186/s12989-018-0274-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Manickam V, et al. Iron oxide nanoparticles induces cell cycle-dependent neuronal apoptosis in mice. J Mol Neurosci. 2018;64(3):352–362. doi: 10.1007/s12031-018-1030-5. [DOI] [PubMed] [Google Scholar]
- 246.Sheng L, et al. Mechanisms of TiO2 nanoparticle-induced neuronal apoptosis in rat primary cultured hippocampal neurons. J Biomed Mater Res A. 2015;103(3):1141–1149. doi: 10.1002/jbm.a.35263. [DOI] [PubMed] [Google Scholar]
- 247.Lebda MA, et al. Potential role of α-lipoic acid and Ginkgo biloba against silver nanoparticles-induced neuronal apoptosis and blood-brain barrier impairments in rats. Life Sci. 2018;212:251–260. doi: 10.1016/j.lfs.2018.10.011. [DOI] [PubMed] [Google Scholar]
- 248.Xue Y, et al. Four types of inorganic nanoparticles stimulate the inflammatory reaction in brain microglia and damage neurons in vitro. Toxicol Lett. 2012;214(2):91–98. doi: 10.1016/j.toxlet.2012.08.009. [DOI] [PubMed] [Google Scholar]
- 249.Imam SZ, et al. Iron oxide nanoparticles induce dopaminergic damage: in vitro pathways and in vivo imaging reveals mechanism of neuronal damage. Mol Neurobiol. 2015;52(2):913–926. doi: 10.1007/s12035-015-9259-2. [DOI] [PubMed] [Google Scholar]
- 250.Elsaesser A, Howard CV. Toxicology of nanoparticles. Adv Drug Deliv Rev. 2012;64(2):129–137. doi: 10.1016/j.addr.2011.09.001. [DOI] [PubMed] [Google Scholar]
- 251.Trickler WJ, et al. Silver nanoparticle induced blood-brain barrier inflammation and increased permeability in primary rat brain microvessel endothelial cells. Toxicol Sci. 2010;118(1):160–170. doi: 10.1093/toxsci/kfq244. [DOI] [PubMed] [Google Scholar]
- 252.Haase A, et al. Effects of silver nanoparticles on primary mixed neural cell cultures: uptake, oxidative stress and acute calcium responses. Toxicol Sci. 2012;126(2):457–468. doi: 10.1093/toxsci/kfs003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Calderón-Garcidueñas L, et al. Alzheimer disease starts in childhood in polluted Metropolitan Mexico City. A major health crisis in progress. Environ Res. 2020;183:109137. doi: 10.1016/j.envres.2020.109137. [DOI] [PubMed] [Google Scholar]
- 254.Calderón-Garcidueñas L, et al. Combustion and friction-derived nanoparticles and industrial-sourced nanoparticles: the culprit of Alzheimer and Parkinson's diseases. Environ Res. 2019;176:108574. doi: 10.1016/j.envres.2019.108574. [DOI] [PubMed] [Google Scholar]
- 255.Hopkins LE, et al. Repeated iron-soot exposure and nose-to-brain transport of inhaled ultrafine particles. Toxicol Pathol. 2018;46(1):75–84. doi: 10.1177/0192623317729222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Maher BA, et al. Magnetite pollution nanoparticles in the human brain. Proc Natl Acad Sci USA. 2016;113(39):10797–10801. doi: 10.1073/pnas.1605941113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Valdiglesias V, et al. Are iron oxide nanoparticles safe? Current knowledge and future perspectives. J Trace Elem Med Biol. 2016;38:53–63. doi: 10.1016/j.jtemb.2016.03.017. [DOI] [PubMed] [Google Scholar]
- 258.Castellani RJ, et al. Iron: the Redox-active center of oxidative stress in Alzheimer disease. Neurochem Res. 2007;32(10):1640–1645. doi: 10.1007/s11064-007-9360-7. [DOI] [PubMed] [Google Scholar]
- 259.Coccini T, et al. Cytotoxicity and proliferative capacity impairment induced on human brain cell cultures after short-and long-term exposure to magnetite nanoparticles. J Appl Toxicol. 2017;37(3):361–373. doi: 10.1002/jat.3367. [DOI] [PubMed] [Google Scholar]
- 260.Yarjanli Z, et al. Iron oxide nanoparticles may damage to the neural tissue through iron accumulation, oxidative stress, and protein aggregation. BMC Neurosci. 2017;18(1):51. doi: 10.1186/s12868-017-0369-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Teller S, et al. Magnetite-Amyloid-β deteriorates activity and functional organization in an in vitro model for Alzheimer’s disease. Sci Rep. 2015;5:17261. doi: 10.1038/srep17261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Zhou M, et al. Implications for blood-brain-barrier permeability, in vitro oxidative stress and neurotoxicity potential induced by mesoporous silica nanoparticles: effects of surface modification. RSC Adv. 2016;6(4):2800–2809. [Google Scholar]
- 263.You R, et al. Silica nanoparticles induce neurodegeneration-like changes in behavior, neuropathology, and affect synapse through MAPK activation. Part Fibre Toxicol. 2018;15(1):28. doi: 10.1186/s12989-018-0263-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Distasi C, et al. SiO(2) nanoparticles modulate the electrical activity of neuroendocrine cells without exerting genomic effects. Sci Rep. 2018;8(1):2760. doi: 10.1038/s41598-018-21157-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Li X, et al. SiO2 nanoparticles change colour preference and cause Parkinson's-like behaviour in zebrafish. Sci Rep. 2014;4:3810. doi: 10.1038/srep03810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Yang X, et al. Uptake of silica nanoparticles: neurotoxicity and Alzheimer-like pathology in human SK-N-SH and mouse neuro2a neuroblastoma cells. Toxicol Lett. 2014;229(1):240–249. doi: 10.1016/j.toxlet.2014.05.009. [DOI] [PubMed] [Google Scholar]
- 267.Song B, et al. Unraveling the neurotoxicity of titanium dioxide nanoparticles: focusing on molecular mechanisms. Beilstein J Nanotechnol. 2016;7:645–654. doi: 10.3762/bjnano.7.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Wu T, Tang M. The inflammatory response to silver and titanium dioxide nanoparticles in the central nervous system. Nanomedicine. 2018;13(2):233–249. doi: 10.2217/nnm-2017-0270. [DOI] [PubMed] [Google Scholar]
- 269.Disdier C, et al. Brain inflammation, blood brain barrier dysfunction and neuronal synaptophysin decrease after inhalation exposure to titanium dioxide nano-aerosol in aging rats. Sci Rep. 2017;7(1):1–13. doi: 10.1038/s41598-017-12404-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Calderón-Garcidueñas L, et al. Hallmarks of Alzheimer disease are evolving relentlessly in Metropolitan Mexico City infants, children and young adults. APOE4 carriers have higher suicide risk and higher odds of reaching NFT stage V at ≤ 40 years of age. Environ Res. 2018;164:475–487. doi: 10.1016/j.envres.2018.03.023. [DOI] [PubMed] [Google Scholar]
- 271.Calderón-Garcidueñas L, et al. Gait and balance disturbances are common in young urbanites and associated with cognitive impairment. Air pollution and the historical development of Alzheimer's disease in the young. Environ Res. 2020;191:110087. doi: 10.1016/j.envres.2020.110087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Kim SH, et al. Rapid doubling of Alzheimer's amyloid-β40 and 42 levels in brains of mice exposed to a nickel nanoparticle model of air pollution. F1000Res. 2012;1:70. doi: 10.12688/f1000research.1-70.v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Purdey M. Elevated levels of ferrimagnetic metals in foodchains supporting the Guam cluster of neurodegeneration: do metal nucleated crystal contaminants [corrected] evoke magnetic fields that initiate the progressive pathogenesis of neurodegeneration? Med Hypotheses. 2004;63(5):793–809. doi: 10.1016/j.mehy.2004.04.029. [DOI] [PubMed] [Google Scholar]
- 274.Afitska K, et al. (2019) α-Synuclein aggregation at low concentrations. Biochim Biophys Acta Proteins Proteom. 1867;7–8:701–709. doi: 10.1016/j.bbapap.2019.05.003. [DOI] [PubMed] [Google Scholar]
- 275.Coufalík P, et al. Content of metals in emissions from gasoline, diesel, and alternative mixed biofuels. Environ Sci Pollut Res Int. 2019;26(28):29012–29019. doi: 10.1007/s11356-019-06144-4. [DOI] [PubMed] [Google Scholar]
- 276.Sonntag DB, et al. Contribution of lubricating oil to particulate matter emissions from light-duty gasoline vehicles in Kansas City. Environ Sci Technol. 2012;46(7):4191–4199. doi: 10.1021/es203747f. [DOI] [PubMed] [Google Scholar]
- 277.Casado JE, et al. Unctuous ZrO(2) nanoparticles with improved functional attributes as lubricant additives. Nanotechnology. 2017;28(49):495704. doi: 10.1088/1361-6528/aa93ca. [DOI] [PubMed] [Google Scholar]
- 278.Yasar A, et al. A comparison of waste recycling facilities for their contribution of heavy metals and trace elements in ambient air. Environ Sci Pollut Res Int. 2021;28:1–9. doi: 10.1007/s11356-020-12101-3. [DOI] [PubMed] [Google Scholar]
- 279.Cenci MP, et al. Precious and critical metals from wasted LED lamps: characterization and evaluation. Environ Technol. 2020;10:1–12. doi: 10.1080/09593330.2020.1856939. [DOI] [PubMed] [Google Scholar]
- 280.Bhagat J, et al. Toxicological interactions of microplastics/nanoplastics and environmental contaminants: current knowledge and future perspectives. J Hazard Mater. 2021;405:123913. doi: 10.1016/j.jhazmat.2020.123913. [DOI] [PubMed] [Google Scholar]
- 281.Okeme JO, Arrandale VH. Electronic waste recycling: occupational exposures and work-related health effects. Curr Environ Health Rep. 2019;6(4):256–268. doi: 10.1007/s40572-019-00255-3. [DOI] [PubMed] [Google Scholar]
- 282.Ghoshdastidar AJ, Ariya PA. The existence of airborne mercury nanoparticles. Sci Rep. 2019;9(1):10733. doi: 10.1038/s41598-019-47086-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Cardoso CED, et al. Recovery of rare earth elements by carbon-based nanomaterials—a review. Nanomaterials (Basel) 2019;9(6):814. doi: 10.3390/nano9060814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Živančević K, et al. Elucidating the influence of environmentally relevant toxic metal mixture on molecular mechanisms involved in the development of neurodegenerative diseases: In silico toxicogenomic data-mining. Environ Res. 2021;194:110727. doi: 10.1016/j.envres.2021.110727. [DOI] [PubMed] [Google Scholar]
- 285.Hang BT, Anh TT. Controlled synthesis of various Fe(2)O(3) morphologies as energy storage materials. Sci Rep. 2021;11(1):5185. doi: 10.1038/s41598-021-84755-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Oggiano R, et al. An overview on amyotrophic lateral sclerosis and cadmium. Neurol Sci. 2021;42(2):531–537. doi: 10.1007/s10072-020-04957-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Roos E, et al. Amyotrophic lateral sclerosis after exposure to manganese from traditional medicine procedures in Kenya. Biol Trace Elem Res. 2020;199:3618–3624. doi: 10.1007/s12011-020-02501-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Engin AB. Combined toxicity of metal nanoparticles: comparison of individual and mixture particles effect. Adv Exp Med Biol. 2021;1275:165–193. doi: 10.1007/978-3-030-49844-3_7. [DOI] [PubMed] [Google Scholar]
- 289.Calderón-Garcidueñas L, et al. Air pollution and brain damage. Toxicol Pathol. 2002;30(3):373–389. doi: 10.1080/01926230252929954. [DOI] [PubMed] [Google Scholar]
- 290.Tesauro M, et al. Metal(loid)s role in the pathogenesis of amyotrophic lateral sclerosis: environmental, epidemiological, and genetic data. Environ Res. 2021;192:110292. doi: 10.1016/j.envres.2020.110292. [DOI] [PubMed] [Google Scholar]
- 291.National Research Council Committee for Review of the, D.s.E.P.M.S.P.R. (2010) In Review of the Department of Defense Enhanced Particulate Matter Surveillance Program Report, National Academies Press (US). Copyright 2010 by the National Academy of Sciences. All rights reserved [PubMed]
- 292.Olvera HA, et al. Ultrafine particle levels at an international port of entry between the US and Mexico: exposure implications for users, workers, and neighbors. J Expo Sci Environ Epidemiol. 2013;23(3):289–298. doi: 10.1038/jes.2012.119. [DOI] [PubMed] [Google Scholar]
- 293.Guerrero-Beltrán CE, et al. Silica nanoparticles induce cardiotoxicity interfering with energetic status and Ca(2+) handling in adult rat cardiomyocytes. Am J Physiol Heart Circ Physiol. 2017;312(4):H645–h661. doi: 10.1152/ajpheart.00564.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Limón-Pacheco JH, et al. Astrocytes are more vulnerable than neurons to silicon dioxide nanoparticle toxicity in vitro. Toxics. 2020;8(3):51. doi: 10.3390/toxics8030051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Wei J, et al. Low-dose exposure of silica nanoparticles induces neurotoxicity via neuroactive ligand-receptor interaction signaling pathway in Zebrafish embryos. Int J Nanomedicine. 2020;15:4407–4415. doi: 10.2147/IJN.S254480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Lojk J, et al. Toxicity mechanisms of selected engineered nanoparticles on human neural cells in vitro. Toxicology. 2020;432:152364. doi: 10.1016/j.tox.2020.152364. [DOI] [PubMed] [Google Scholar]
- 297.Gulati K, et al. Nanoengineered drug-releasing Ti wires as an alternative for local delivery of chemotherapeutics in the brain. Int J Nanomedicine. 2012;7:2069–2076. doi: 10.2147/IJN.S29917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Weiner MW, et al. Military risk factors for Alzheimer’s disease. Alzheimers Dement. 2013;9(4):445–451. doi: 10.1016/j.jalz.2013.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Heyburn L, et al. The role of TDP-43 in military-relevant TBI and chronic neurodegeneration. Front Neurol. 2019;10:680. doi: 10.3389/fneur.2019.00680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Weedn VW. Origins of the armed forces medical examiner system. Acad Forensic Pathol. 2020;10(1):16–34. doi: 10.1177/1925362120937916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Brady CB, et al. The Department of Veterans Affairs Biorepository Brain Bank: a national resource for amyotrophic lateral sclerosis research. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14(7–8):591–597. doi: 10.3109/21678421.2013.822516. [DOI] [PubMed] [Google Scholar]
- 302.O'Leary MC, et al. Development of a multi-study repository to support research on veteran health: the VA Cooperative Studies Program Epidemiology Center-Durham (CSPEC-Durham) Data and Specimen Repository. Front Public Health. 2021;9:612806. doi: 10.3389/fpubh.2021.612806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Obermann M, Lyon M. Financial cost of amyotrophic lateral sclerosis: a case study. Amyotroph Lateral Scler Frontotemporal Degener. 2015;16(1–2):54–57. doi: 10.3109/21678421.2014.951946. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.
Not applicable.