Next Article in Journal
Study of Clear Air Turbulence Related to Tropopause Folding over the Romanian Airspace
Next Article in Special Issue
Populus nigra Italica Leaves as a Valuable Tool for Mineralogical and Geochemical Interpretation of Inorganic Atmospheric Aerosols’ Genesis
Previous Article in Journal
Using a Statistical Crop Model to Predict Maize Yield by the End-Of-Century for the Azuero Region in Panama
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases

by
Deborah A. Cory-Slechta
*,
Marissa Sobolewski
and
Günter Oberdörster
Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
*
Author to whom correspondence should be addressed.
Atmosphere 2020, 11(10), 1098; https://doi.org/10.3390/atmos11101098
Submission received: 8 September 2020 / Revised: 5 October 2020 / Accepted: 7 October 2020 / Published: 14 October 2020
(This article belongs to the Special Issue Metals in Ambient Particles: Sources and Effects on Human Health)

Abstract

:
Increasing evidence links air pollution (AP) exposure to effects on the central nervous system structure and function. Particulate matter AP, especially the ultrafine (nanoparticle) components, can carry numerous metal and trace element contaminants that can reach the brain in utero and after birth. Excess brain exposure to either essential or non-essential elements can result in brain dyshomeostasis, which has been implicated in both neurodevelopmental disorders (NDDs; autism spectrum disorder, schizophrenia, and attention deficit hyperactivity disorder) and neurodegenerative diseases (NDGDs; Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis). This review summarizes the current understanding of the extent to which the inhalational or intranasal instillation of metals reproduces in vivo the shared features of NDDs and NDGDs, including enlarged lateral ventricles, alterations in myelination, glutamatergic dysfunction, neuronal cell death, inflammation, microglial activation, oxidative stress, mitochondrial dysfunction, altered social behaviors, cognitive dysfunction, and impulsivity. Although evidence is limited to date, neuronal cell death, oxidative stress, and mitochondrial dysfunction are reproduced by numerous metals. Understanding the specific contribution of metals/trace elements to this neurotoxicity can guide the development of more realistic animal exposure models of human AP exposure and consequently lead to a more meaningful approach to mechanistic studies, potential intervention strategies, and regulatory requirements.

1. Epidemiological Studies Increasingly Associate Air Pollution with Neurodevelopmental Disorders and Neurodegenerative Diseases

Over the past 10 years, it has become increasingly clear that the brain is a major target of air pollution (AP) [1,2]. For example, numerous epidemiological studies over the past several years have now linked the metrics of AP exposures with autism spectrum disorder (ASD) [3,4,5,6,7,8,9,10,11,12,13,14,15,16], a highly heterogeneously expressed neurodevelopmental disorder (NDD) [17,18,19] diagnosed on the basis of social communication/interaction deficits and restricted and repetitive behaviors, but that can also include hyperactivity, aggression, behavioral inflexibility, sensory-motor disturbances [20], impaired response inhibition, and impulsive responding [21,22]. AP has also now been associated with multiple reports of attention deficits or diagnoses of attention deficit hyperactivity disorder (ADHD) [23,24,25,26,27,28,29], another highly heterogeneous disorder [30], the features of which can be inattention, distractibility, impulsivity, and hyperactivity [31,32], but that can also include aggression, oppositional and externalizing behaviors, and alterations in social behaviors [33,34]}. Schizophrenia (SCZ), which generally has a late teen to early young adult onset and can include positive symptoms (hallucinations, delusions, and thought disorders) and negative symptoms (anhedonia, social communication deficits, and lack of motivation) [35], has also been linked to AP in several studies [36,37,38,39,40,41,42,43]. All three of these disorders are also male-biased in prevalence and/or severity [44], consistent with sex differences in brain development.
AP has also been linked to neurodegenerative diseases (NDGDs). A recent meta-analysis of four cohort studies (Canada, Taiwan, the UK, and the US) reported that increased PM2.5 levels were positively associated with dementia and Alzheimer’s disease (AD) [45], a progressive neurodegenerative disorder characterized by brain accumulation of β-amyloid and hyperphosphorylated tau proteins (plaques and tangles) with a corresponding loss of memory, and a recent study reports a decline in immediate recall and new learning [46]. Long-term PM2.5 exposure was also associated with an increased risk of dementia and AD in a meta-analysis of 80 studies [47], and with dementia and AD in a Swedish cohort [48] and in a Taiwanese cohort [49]. Consistent with prodromal symptoms of AD and other NDGDs, AP has also been related to olfactory dysfunction, consistent with the translocation of particulate matter via the olfactory axons directly into the olfactory cortex [50]. Parkinson’s disease (PD) is characterized by tremor, slowness of movement, and incoordination that results from the loss of the central nervous system nigrostriatal system dopamine. Several studies have linked AP exposures to PD in meta-analyses [51,52,53] and in individual studies [54,55,56,57], although such findings have not been completely invariant [58,59,60]. A role for AP exposure is also now indicated in some [61,62,63,64,65,66] but not all reports [67] for multiple sclerosis (MS), considered an autoimmune disease characterized by significant central nervous system and peripheral nervous system demyelination, with symptoms including pain, fatigue, incoordination, and motor impairment. AP also appears to be associated with amyotrophic lateral sclerosis (ALS), a disease damaging the neurons of the brain and spinal cord, leading to progressive muscle weakness [68,69,70]. Notably, a study of over 1000 women without any indication of dementia revealed that PM2.5 exposure resulted in a loss of brain white matter (myelin), including in the frontal and temporal lobes and in the corpus callosum, the largest white matter (myelinated) tract of the brain [2]. AP exposures have also been linked in human studies to alterations in brain connectivity [71], impaired motor performance [72,73], altered cerebral cortex structures related to impaired inhibitory control [74], and cognitive development [75].
While the breadth of effects seems already quite broad, it is important to remember that epidemiological studies are associative in nature. Clearly, critical evaluations of individual studies are warranted in terms of defining the strength of the epidemiological associations.

2. What Component(s) of Air Pollution Contribute to This Neurotoxicity?

AP, which is largely a product of vehicle exhaust and industrial emissions combined with atmospheric physico-chemical processes, is a complex mixture of particles, gases (CO2, CO, NO, ozone, SO2), and adhering contaminants. AP health effects are related to particulate matter (PM) size, designated as coarse (<10 µm or PM10), fine (<2.5 µm or PM2.5), or ultrafine (UFPs; <100 nm or PM0.1). UFPs are considered the most reactive component of AP [76] because of their greater surface area/mass for contamination by volatile organic and inorganic contaminants, including metals and trace elements that adhere to the carbon core [77]. In many parts of the world, the levels of PM10 and PM2.5 have declined over the years in response to regulations, thus reducing the levels of particulate AP. However, it is not clear that this is true for UFPs, based on studies reporting an absence of correlation between concentrations of UFPs and PM2.5 [78,79,80], due in part to the generation of UFPs via atmospheric gas to particle conversion [81,82].
Understanding the specific AP components that may contribute to NDD or NDGD risk is of critical importance for several reasons. First, such an understanding can guide the development of more realistic focused animal exposure models of human AP exposure and, consequently, a more meaningful approach to mechanistic studies, as well as to potential intervention strategies for NDDs and NDGDs. Further, a greater understanding of the neurotoxic components of AP will also help to focus epidemiological studies to consider speciation effects where possible. Of course, understanding the components of AP relating to this neurotoxicity is of translational importance to public health protection, as it can provide information pertinent to regulations of exposures, including the need for new or more stringent regulations.

3. Metals as a Source of AP-Related Neurotoxicity

One source of such neurotoxicity could be the metals and trace element contaminants of particulate matter [83], which include both metals essential to brain as well as non-essential elements. The metals and trace elements identified in PM arise from numerous anthropogenic sources, including mechanical engine wear, emissions from tail pipes, brake wear, coal-fired power plants, oil combustion processes, metal refineries, metal ore smelting and processing, and other industries [83]. Most such metals/elements, including those from roadway traffic, are insoluble, but can be solubilized and subsequently lead to redox cycling and oxidative stress in vivo. This can be achieved via secondary processing—e.g., in response to highly acidic sulfate aerosols [84]. SO2, for example, has been shown to enhance the Fe2O3 particle uptake, as seen in the mouse bronchial epithelium [85], and thereafter alter the Fe intracellular distribution.
Furthermore, as might be expected, the specific profile of metals adsorbed to particulates and their concentrations will vary significantly across time and by geographic location due to local industrial, traffic-related, and anthropogenic conditions [83]. Figure 1 shows the geographic distribution of PM2.5 in the United States (U.S.) derived from the 2001 National Emission Inventory, estimated at 5,050,000 tons/year, with the highest levels in the midwest, southeast, and the east and west coasts. Corresponding maps are shown for Fe, Al, Si, S, Zn, Pb, and Cu, refined using data from the 2002 Hazardous Air Pollution inventory [83]. As can be seen, the levels of Fe, Al, and Si are quite high in these same regions, with emissions of 223,000, 103,000, and 140,000 tons/year, respectively, with lesser levels of S, Zn, Pb, and Cu at 79,000, 3290, 895, and 1710 tons/year, respectively. As these maps demonstrate, these metals and trace elements are indeed present and thus could act as potential risk factors for NDDs and NGDGs [83].
Our studies of the inhalational exposures of mice to AP similarly highlight differences in metal and elemental constituents of AP by geographical location. Postnatal exposures of mice [86,87,88,89,90,91,92,93] were carried out via inhalational exposure to 10–20× concentrated ambient ultrafine particles (CAPs) in Rochester, NY, for 4 h/day from postnatal days (PND) 4–7 and 10–13, a time period analogous to the human 3rd trimester brain development, characterized by extensive neuro- and gliogenesis [94]. In another study, gestational (GE) CAPs exposures (gestational days 0.5–16.5; human brain 1st + 2nd trimester equivalent) to fine/UFP AP were carried out in Tuxedo, NY [95,96,97]. XRF analyses of filters from the exposure chambers from these studies (Figure 2) shows associated elemental tree-maps. The profiles of metals and elements differed by geographical site, as expected. Interestingly, the XRF analysis from Rochester showed a dominant S component and identified Fe, Al, Si, Zn, Pb, and Cu as well as numerous other constituents in the UFP. S also dominated in the GE CAPs in Tuxedo, NY, followed by a significant Fe component.
Studies from our laboratory in mice also demonstrate the potential for inhaled ambient AP to alter in vivo both the profile and level of metals in the brain [92]. Laser ablation ICP-MS analyses of a randomly chosen section of a male CAPs vs. air-exposed brains following postnatal CAPs exposures (Figure 3) confirmed a marked elevation of metal levels in the brain, including Fe, S, Cu, Ca, and Al, while concurrently suggesting reductions in Mn and Zn.

4. Evidence for the Involvement of Metals in NDDs and NDGDs

A role for trace elements/metal alterations in the etiology of NDDs and NDGDs has been suggested in numerous studies over the years. Metal/elemental disturbances have been associated with NDDs [98], suggesting a role of metal dyshomeostasis in their etiology, albeit with much of the focus of studies on NDDs on peripheral markers rather than the brain. For example, alterations in the levels of metals and/or trace metals in serum or in hair or nails are reported and sometimes correlate with the features or severity of ASD [99,100,101,102,103,104,105,106,107]; the observed metal profiles can differ by sex [108] and geographic location. Alterations in the brain metal levels in ASD have been reported [109,110], as have correlations of trace metal levels with neuroinflammatory markers [111]. Similar findings are reported in SCZ, particularly for Zn and Cu [112,113,114,115,116,117,118,119], as well as in ADHD [120,121,122,123,124,125,126]. During development, the brain acquires mechanisms to tightly regulate the levels of essential metals. However, increases in the brain in the levels of either essential or non-essential metals, particularly redox-active but required metals—e.g., Fe and Cu—results in imbalance (i.e., brain metal dyshomeostasis) that can then progress to oxidative stress and neuroinflammation, disrupting brain development and subsequent function [127].
A strong case for brain trace metal disturbances has emerged for NDGDs. For example, numerous NDGDs are associated with excess brain Fe accumulation. Fe accumulates in the human brain with age; however, excesses above aging-related increases appear in multiple NDGDs and are thought to underlie associated oxidative stress, protein aggregation, demyelination, and neuronal loss [73,128,129,130]. Both Fe and Cu have been reported to promote the formation and or accumulation of β-amyloid plaques, as well as the hyperphosphorylation of tau protein associated with AD [131], thereby contributing to the formation of intracellular plaques and tangles [73]. The accumulation of such metals can displace other essential metals such as Zn, mitigating its anti-inflammatory and anti-redox properties and its ability to precipitate aggregation intermediates [73]. Fe deposition has been shown to correlate with cognitive decline in AD [132]. Another metal that has repeatedly been implicated in AD is aluminum (Al) [110,133,134,135], which can likewise contribute to β-amyloid formation and tau protein aggregation.
PD, which includes the loss of brain dopamine and the presence of Lewy bodies with inclusions, most notably of α-synuclein, likewise includes excess Fe accumulation, particularly in the substantia nigra, the site of nigrostriatal dopamine system cell bodies. Fe has been reported to directly bind α-synuclein, with a resulting pathophysiological structural reorganization and post-translational modifications of the protein (e.g., nitration, phosphorylation) [136]. Cu has also been associated with the oligomerization of α-synuclein, as well as oxidative stress [73]. While Mn appears to have a poor binding affinity to α-synuclein, studies show that it can trigger the misfolding and accumulation of this protein [137,138]. Mn also influences numerous neurotransmitter systems, including dopamine systems where striatal reductions in dopamine release and in the levels of the D2 dopamine receptor have been reported [139,140].
Excess Fe has also been considered a key driver in MS, with effects hypothesized to occur through mechanisms including oxidative stress, such as could occur due to the release of Fe from Fe-rich oligodendrocytes, the precursor cells for myelination. Excess Fe is also likely, via oxidative stress mechanisms, to increase proinflammatory cytokines and thereby promote demyelination [73,141]. Low levels of Zn may result from the excess accumulation of other metals in the brain that then suppress its anti-inflammatory protective capacity.
Several metals have been implicated in ALS. As with other NDGDs, this includes Fe, considered a biomarker for ALS [142], likely through Fenton reaction-generated oxidative stress [143]. Albeit considered less important, Cu may be involved through the formation of aggregates of superoxide dismutase 1 and Cu transport proteins [127]. In epidemiological studies, ALS has been associated with reduced serum Zn levels and increased Cu levels [144], as well as with blood and bone levels of Pb [145].
Calcium is another required element that is found in AP. No studies to date appear to have examined the impact of Ca overload in brain via instillation or inhalation. It is notable, however, that voltage-gated calcium channel gene variants have been linked to the pathogenesis of ASD [146], and proteomic analyses of ASD brains have identified the upregulation of proteins involved in Ca signaling [147]. Ca-binding protein dysregulation is also seen in the brains of individuals with SCZ [148]. Alterations in mitochondrial Ca homeostasis are considered to contribute to neuronal loss in PD [149], and has also been postulated to serve as a key mechanism in the etiology of AD [150] and ALS [151].

5. Could AP-Related Brain Metals Contribute to Brain Metal Dyshomeostasis?

As the above suggests, metals may play pathophysiological roles in NDDs and NDGDs. Fe (chemical species unknown) has been reported to increase in the human brain with normal aging [152,153,154,155,156], whereas the increases in Cu appear to be less pronounced, at least as measured in the human substantia nigra [157]; however, the findings have been equivocal, with reports of no increase [158] as well as increases in some cases [159]. Similarly, the levels of Zn in the human brain have been reported to show age-related increases [158], whereas in a study focused on the frontal cortex and hippocampus the Zn levels significantly declined with age [159]. Such discrepancies likely reflect differences in various parametric features of individual studies, such as the chemical formulation of the metal/trace element, dose, exposure durations, ages, and sample sizes, etc., but all findings are consistent with brain metal dyshomeostasis. The mechanism(s) of this aging-related brain metal dyshomeostasis remain unclear, but the possibilities proposed have been age-related loss of the homeostatic control of metal levels, including in the periphery and the brain. Another is aging-related pathophysiological changes in barriers such as the blood brain barrier, or the gastrointestinal tract [160,161,162].
However, what appears never to have been considered is the possible life-long exposures to metals via air pollution. Inhaled metal or trace element contaminants in AP can reach the brain at any stage of life. During gestation, metals can move across the placenta, even against a concentration gradient [163,164], into the fetal blood stream and then be distributed to developing organs. For example, non-heme (i.e., unbound) Fe is detectable in rat brain choroid plexus epithelial cells by gestational day (GD)14 and in the developing axonal tracts of fetal corpus callosum by GD19 [165]. S moves across the placenta in humans as sulfate via sulfate transporters [166]; the fetus relies directly on maternal sources [166]. Numerous other nanoparticle-adsorbed or incorporated metal and trace elements also show placental transfer and accumulation in fetal tissue—e.g., As, Pb, Cd, Hg, Cu, Ni, Mn, Al, and V [167,168,169,170,171,172,173,174,175,176]. As the Fe regulatory proteins 1 and 2 and Fe transporters such as the transferrin receptor and divalent metal transporter-1, critical to the tight brain regulation of Fe levels, for example, are not fully expressed until PND 15–20 in rodents [177,178], an extended window is open during which excess Fe can reach and directly influence brain development. Similarly, the homeostatic regulation of Fe absorption appears in human infants at about 6–9 months of age [179].
At least two routes allow AP-related metals and trace elements to reach the brain by inhalation following birth. Particle passage, particularly of UFP, proceeds from deposits in the alveolar region of the lung into the bloodstream [180], followed by distribution to secondary organs [181]. In addition, uptake via sensory nerves in the upper respiratory tract (olfactory and trigeminal [182]) is known to occur. For example, proof of principle nasal olfactory uptake of nanosized elemental AP particles has been demonstrated in both humans and rodents [183], which for the latter has included the uptake of Fe, Mn, Cd, Ni, Hg, Al, Co, Zn, and Cu [183,184,185,186,187,188,189,190] via inhalation or intransal instillation, moving across olfactory epithelial barriers in the nasal passages, likely via uptake by dendrites of neuronal olfactory cells reaching out with cilia directly into the nasal lumen, followed by transport to the olfactory bulb and then to other brain regions [191]. With olfactory or trigeminal nerve uptake, trace elements and metals actually bypass the tight blood brain barrier, such that increased levels of trace elements in the brain may not be reflected in peripheral markers. For example, the intranasal instillation of Fe2O3 nanoparticles to rats over 7 days increased the Fe brain levels while significantly reducing the serum Fe levels [192]. Such discrepancies between the brain and serum levels are also seen in neurodegenerative diseases that include elevated brain Fe—e.g., PD [193,194,195]. In addition, sensory nerves in the upper and lower respiratory tract can translocate particles [196] that reach, e.g., the trigeminal ganglion, possibly via the hypoglossal nerve and tongue [182,191,197].
It is also interesting to consider AP as a source of neurotoxicity risk in the context of the dual-hit hypothesis proposed for the origins of PD [198]. According to this hypothesis, brain PD pathology actually originates in the olfactory bulb and brain stem via the vagal nerve (Figure 4). Either such route could potentially result from AP exposure—i.e., the olfactory/trigeminal uptake of metal-containing UFPs—or potentially from vagal transfer into the brain stem [199,200,201], as has been demonstrated for metals [202] but is as yet unknown for NPs.
Collectively, such findings raise the question of whether AP-derived metals contribute a risk for NDDs or NDGDs. One way to begin to address this question is to examine the consequences of inhalational exposures to AP-related metals and trace elements in relation to the features of NDDs and NDGDs. This paper examines that premise based upon inhalation exposure studies; some studies using intranasal instillation are also included, but it is important to note that non-physiological bolus-type instillation exposures are significantly different in terms of the exorbitant dose and dose rate, as well as, importantly, deposition throughout the respiratory tract. Furthermore, particles in suspension will have altered surface chemistry and agglomeration/aggregation properties compared to those in an aerosol [203]. Nevertheless, intranasal instillation studies can be confirmatory of the trigeminal pathway of uptake, despite their poor real-world relevance. This chapter also did not include studies utilizing engineered NPs, given that such reformulations could change activity and outcome.
To facilitate this assessment, the current review focuses on features that are common across NDDs and NDGDs that should also assist in determining the breadth of metal-related AP exposures as a risk factor. To date, the number of studies based on inhalation or intranasal instillation is relatively limited. Further, the studies also differ in relation to the particular trace element or metal under study; comparisons can be hindered by the fact that exposure concentrations and specific metal chemistry and other exposure parameters can vary widely across such studies, rendering direct comparisons difficult. Nevertheless, the assessment of the ability of AP-related inhaled trace element and metal NP exposures to reproduce specific known features of NDDs and NGDGs also provides the basis for hypothesis-forming studies for more precisely defining future research directions.

6. Shared Features of Neurodevelopmental Disorders and Neurodegenerative Diseases

While NDDs and NDGDs both have unique features, they also share multiple neuropathological and behavioral characteristics and hypothesized mechanisms [19,24,34,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254,255,256,257,258,259,260,261,262,263,264,265,266,267,268,269,270,271,272,273,274,275,276,277,278,279,280,281,282,283,284,285,286,287,288,289,290,291], suggesting that if AP-related trace element exposures produce these characteristics, they would be a significant risk factor for a wide range of disorders with high economic and societal costs. For example, as shown in Table 1, ventriculomegaly—i.e., enlarged lateral ventricles—is found in ASD, ADHD, and SCZ. This has been shown to persist across life in individuals with ASD [292,293,294,295]; ventriculomegaly is considered a hallmark characteristic of SCZ [207,208,209,210,211,212]. Ventriculomegaly has also been seen in studies of ADHD [23,204,205]. White matter reductions (i.e., loss of myelination) are seen not only in ASD [213,214,215,216,217], but are also a prominent feature of SCZ [219,220,221,222] and are reported in ADHD as well [214,218,296,297]. Ventriculomegaly is seen in AD [257,298,299,300,301], as well as in PD [302,303], ALS [304], and MS [305,306].
Myelination is critical to the connectivity of the two hemispheres and is required to mediate cognitive and other behavioral functions [223,224]; disconnectivity—i.e., loss of interhemispheric myelination—is considered a key mechanism of ASD-related behavioral disorders [225,227,228]. Additionally, such disconnectivity is a well-documented feature of SCZ [226,230,231,232,233,307] and is also reported as a mediator of behavioral deficits in ADHD [229,232,237,238]. Demyelination, including loss of white matter in the corpus callosum (the largest white matter tract in brain) and interhemispheric disconnectivity, is also a shared feature of AD [308,309,310], PD [311,312,313,314], MS [315,316,317,318], and ALS [319,320,321].
Alterations in glutamatergic functioning and excitatory-inhibitory imbalance are seen in ASD [234,235,236,239,244] and are prominent in SCZ [236,240,243,246,247] and ADHD [241,242,254]. Glutamatergic dysfunction occurs in AD [322,323,324], PD [325,326], MS [326,327,328,329,330], and ALS [331,332,333].
While each of these NDDS has some unique behavioral features, they can also share behavioral impairments, such as alterations in social functioning, impulsivity, and executive function deficits [29,334,335,336,337,338,339,340,341,342,343,344,345,346,347,348,349]. Cognitive/executive function deficits are also common in NDGDS (AD [350,351], PD [352,353], MS [354,355,356,357], ALS [358,359,360,361,362]), as are alterations in social behaviors [363,364,365] and manifestations of impulsive-like behaviors [366,367,368,369,370].
Hypothesized mechanisms of these disorders, such as mitochondrial dysfunction, have been reported in all NDDs, particularly in SCZ [276,277,283,285,286,288,289,291], and could be related to alterations in neuronal fate [371,372]. Inflammation and oxidative stress are also the hypothesized mechanisms of all three of these NDDs [23,261,373,374,375,376,377,378]. Neuronal cell death is likewise characteristic of NDGDs, as seen in AD [379,380], PD [381,382], MS [383,384,385], and ALS [331,385,386]. Potential mechanistic commonalities include mitochondrial dysfunction (AD [387,388,389], PD [390,391], MS [392,393,394], ALS [395,396,397]), as well as microglial activation/inflammation (AD [398,399,400,401,402], PD [403,404,405], MS [406,407,408,409], ALS [410,411,412]) and oxidative stress (AD [413,414,415,416], PD [413,414,415,417], MS [418,419,420,421], ALS [332,422,423]).

7. Current Evidence That AP-Related Metals and Trace Elements Can Reproduce Shared Characteristics of NDDs and NDGDs

Fe is the most abundant metal in the atmosphere [424] and is often present in the form of hematite (Fe2O3) or magnetite (Fe3O4), but can also be found as sulfates or carbonates. That atmospheric Fe associated with nanosized PM can reach the brain, and can do so via olfactory and trigeminal nerve uptake, bypassing the blood brain barrier, has been demonstrated. Mice of 4 weeks of age were exposed via intranasal instillation of the right nasal cavity to 40 mg/kg body weight of fine Fe2O3 particles (mean diameter 280 nm), with outcomes assessed 14 days later [184]; these exposures produced increases in olfactory bulb and brain stem Fe concentrations, accompanied by neuronal degeneration in the hippocampus. A subsequent study comparing the effects of particle sizes of 40 vs. 280 nm Fe2O3 in response to intranasal instillation in male mice found that smaller-sized Fe2O3 particles were present in the axons of the olfactory neurons, as well as in the mitochondria and lysosomes in the hippocampus [425]. In another nose-only inhalation study, exposures of rats to 1.0 to 1.14 mg/m3 of Fe2O3 nanoparticles (mean diameter 14 nm) for 4 h/day, 5 days/week, for 3, 6, or 10 months demonstrated the presence of Fe NPs within damaged axonal sheaths of the olfactory bulb [426]. Similarly, a study of the whole-body inhalation of Fe (40 ug/m3) soot (200 ug/m3) for 6 h/day for 5 days/week for 5 weeks in female mice showed that Fe reached brain via the olfactory bulb, where it produced neuroinflammation, including increased numbers of activated microglial cells as well as levels of the pro-inflammatory cytokine IL-1β [427]. Studies of the human frontal cortex have found the presence of magnetite NPs in the human brain with a composition, according to the authors, consistent with external rather than internal formation [428]. Levels of the neurotransmitters dopamine and norepinephrine were markedly increased in male rats seven days after the intranasal instillation of 10 mg/kg of 30 nm-diameter Fe2O3 for 7 days [192]. That such exposures can induce oxidative damage was demonstrated in a study in which intranasal instillation of 20 ug Fe3O4 (462–570 nm in diameter) in rats for 7 days increased striatal and hippocampal H2O2, the latter measured via colorometric assay, and also reduced oxidized glutathione in the striatum [429]. Assessment of behavioral function appears to be limited to a single study that reported that airborne Fe levels were negatively associated with fine motor function across four cohorts of children from 1 to 9 years of age [430] (Summarized in Supplemental Table S1).
Collectively, while limited in scope, studies using environmentally relevant inhaled or less relevant intranasally instilled Fe indicate that nanoparticles (NPs) can reach the brain, bypass the blood brain barrier, and produce features consistent with the hypothesized mechanisms of both NDDs and NDGDs, which, based on current studies, include effects on myelination, neuronal cell death, inflammation, and microglial activation, as well as oxidative stress and mitochondrial dysfunction (Table 1 and Supplemental Table S1). As noted above, such exposures would also be expected to be lifelong, as they can begin in utero and continue after birth. Further exacerbating the potential toxicity of elevated brain Fe in particular, studies have demonstrated its extremely slow turnover in rodent brains (half-time of ca. 9 mos), with estimates in the order of decades when extrapolated to humans, although this depends upon the underlying mechanisms of clearance and potential dissolution [431,432,433].
The Cu levels in AP can also be quite high depending upon geography [434]. Like any airborne nanosized particle, including Fe, Cu nanoparticles also reach brain via routes that include the olfactory bulb, where they produce effects consistent with those seen in NDDs and NDGDs. For example, female mice exposed via intranasal instillation to one of three doses (1, 10, and 40 mg/kg body weight, respectively) of Cu NPs (CuCl2·2H2O) for a total of either 7 or 15 days showed significant increases in Cu concentrations in the olfactory bulb, as well as numerous neurochemical changes in multiple brain regions [435]. These included reductions in dopamine (DA) and homovanillic acid (HVA) with increases 3,4-dihydoxyphenylacetic acid (DOPAC), as well as increased intracellular DA turnover in the olfactory bulb, whereas DA, DOPAC, and HVA were all increased in the hippocampus and cerebellum, as was DA turnover in the cerebellum. Increases in DA were observed in the striatum and cortex at the highest concentrations. The levels of serotonin (5-HT) and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) were also markedly elevated in the olfactory bulb [435]. A subsequent study by this group using the same exposure concentrations administered every other day for 19 days found dose-related increases in Cu concentrations in the hippocampus, cortex, cerebellum, and striatum, accompanied by a modified elemental distribution in the brain of Fe, Zn, and Ca, consistent with brain metal dyshomeostasis [436]. Damaged neurons were particularly evident in the cortex following exposures, with similar trends in the hippocampus, while astrocytic activation was present throughout the hippocampus. Reductions in cortical and cerebellar dopamine were accompanied by marked elevations in its metabolites, as well as in both intracellular and extracellular DA turnover, whereas the levels of DA and metabolites were increased in the cortex, accompanied by reductions in turnover. Some increases in 5-HT and reductions in 5-HT turnover were also found in the cerebellum. In another study, female mice exposed via intranasal instillation to either 1 mg/kg body weight or 40 mg/kg of 25 nanometer Cu particles (CuCl2·2H2O) or a comparable low dose of micro Cu nanoparticles (17 um), each delivered a total of three times across successive days, showed increased astrocytic activation and altered endoplasmic reticulum structure and dissociation in the hippocampus, as well as chromatin condensation and the aggregation of mitochondria in the olfactory bulb when examined 48 h after the termination of exposure [437], although 17 um particles are not likely to be inhalable by humans.
Interestingly, a study of 2897 children from 7 to 11 years of age from the BREATHE project revealed a positive trend between outdoor ambient Cu exposure and inattentiveness (p = 0.076), as measured by a computerized version of the Attentional Network test; more pronounced associations were found in children with polymorphisms of the ATPase copper-transporting beta gene (ATP7B) [438]. In another report incorporating 2836 children of 8–12 years of age, airborne Cu exposure was found to be significantly associated with poorer motor performance; in addition, higher Cu levels were significantly associated with higher fractional anisotropy, as measured via diffusion tensor imaging in white matter close to and within the caudate nucleus—i.e., areas surrounding the lateral ventricles [72]. In summary, to date, Cu nanoparticle inhalation reproduces features of NDDs and NDGDs that include alterations in myelination, neuronal cell death, inflammation, and microglial activation, as well as mitochondrial dysfunction, and have likewise been associated with inattention.
Studies of Zn NP exposure are limited and show that, like other metals, it too is transported via olfactory neurons directly into the brain [439,440]. In a recent study in 6-week-old male Wistar rats, daily intranasal instillation of ZnO NPs (10–30 nm in size; 20 ug/g body weight) was carried out for 15 or 30 days. Transmission electron micrographs were presented to show multiple ultrastructural changes in the olfactory bulb, hippocampus, striatum, and cortex that included mitochondrial changes, as well as changes in the endoplasmic reticulum, although these are difficult to visualize in the paper. Also reported was the presence of NPs in the cytoplasm and nerve fibers, along with histological changes in the hippocampus and striatum comprised of disordered cell arrangement, cell degeneration, and inflammatory cell infiltration. Changes in oxidative stress markers were quantified and revealed increased levels of malondialdehyde, TNF-α, and IL-1β in the olfactory bulb, striatum, hippocampus, and cortex at both 15 and 30 days, in conjunction with a corresponding reduction in glutathione [441]. Thus, inhaled Zn particles have been found to reproduce the neuronal cell death, inflammation, oxidative stress, and mitochondrial dysfunction features of NDDs and NDGDs.
Numerous studies have examined the impact of inhaled Mn, which has also been shown to be taken up via olfactory transport [442], as well as via the trigeminal nerve in Fischer 344/N rats and FVB/N mice [443], with the latter showing a clearance rate half-time of 9–10% per day that did not differ by sex. A MnCl2 inhalation study in CD-1 male mice to 0.02M for 1 h twice/week for a 12-week period confirmed mitochondrial and Golgi damage in olfactory neurons, vacuolization beginning at week 4, and a significant increase in the percentage of apoptotic cells from 8 weeks on [444]. Assessments in mice in which exocytosis results in green fluorescent protein fluorescence in the olfactory bulb nerve terminals following the intranasal instillation of 2–200 µg of MnCl2 demonstrated dose-dependent reductions in odorant-evoked neurotransmitter release even at the 2 µg levels, confirming a central rather than peripheral basis for these effects [445].
The subchronic inhalation exposure of male Sprague Dawley rats (34–38 days of age) to MnPO4 at 3000, 300, 30, or 0.3 μg/m3 of coarse and fine particulates for 6 h/day for 5 days/week for 13 consecutive weeks resulted in significant dose-dependent increases in the Mn concentrations in the olfactory bulb, frontal cortex, globus pallidus, caudate putamen, and cerebellum, but was not found to alter the neuronal cell counts in either the caudate or globus pallidus, nor to alter locomotor activity [446]. A subsequent study by this group using the same exposure paradigm but Mn SO4 rather than MnPO4 resulted in increases in the brain Mn in all regions at the two higher exposure concentrations, with reductions in neuronal counts in the globus pallidus and in ambulatory activity in both the diurnal and nocturnal periods [447]. MnSO4 exposure concentrations of 0.03, 0.3, or 3.0 mg Mn/m3 for 6 h/day for 7 days/week for 14 consecutive days in 6-week-old male Crl;CD (SD)BR rats increased the brain Mn levels in the order of olfactory bulb > striatum > cerebellum at medium and high doses; glutamine synthetase protein levels were increased in the olfactory bulb, and, to a lesser extent, in the hypothalamus. Total glutathione levels were significantly reduced and metallothionein levels increased only in the hypothalamus [448]. A subsequent study using young male and female and aged male CD rats with exposures to MnSO4 of 0.01, 0.1, and 0.5 mg Mn/m3 or MnPO4 at 0.1 mg Mn/m3 showed effects of Mn that were sex-, age-, and brain region-dependent [449] in relation to alterations of glutamine synthetase protein level mRNA, metallothionein levels, and total glutathione [450], as well as in corresponding pharmacokinetics [451]. Exposures of CD rats to MnSO4 (0.05, 0.5, or 1 mg/Mn/m3; mean particle diameter 1.0 um) from 28 days prior to breeding until postnatal day 18 resulted in sex-dependent differences in offspring at postnatal day 19, consisting of reductions in TH mRNA and glutathione in the olfactory bulb and striatum, while reductions in the ratio of glutathione to oxidized glutathione were present in the hippocampus, olfactory bulb, and striatum [452]. Three weeks post-termination of exposure, reductions in glutamine synthetase and metallothionein mRNA levels in females in the striatum were still present at all exposure concentrations, while reductions in glutathione levels were present in males in the cerebellum and in females in the olfactory bulb [453].
Intranasal instillation of MnCl2 solute (40 µL of aqueous 200mM) in rats resulted in olfactory bulb uptake as well as the impairment of olfactory discrimination [454]. Short term exposure of male Sprague Dawley rats (nose only to 1.2 µm, 39 mg/m3 for 5 days/week for 3 weeks) produced a Mn concentration gradient decreasing from olfactory bulb to striatum; while this exposure did not produce changes in rotarod performance, reductions in dopamine transporter levels were present in striatum [455]. Of direct relevance to PD, over the course of exposure of adult male Wistar rats to 0.04 M MnCl2 and 0.02 M Mn(OAc)3 for 1 h 3× weekly for 6 months (72 inhalations), beam walk test time increased significantly, and numbers of successful reaches in a single-pellet reaching task declined. Furthermore, the numbers of tyrosine hydroxylase positive neurons declined dramatically in the substantia nigra, but were, as in PD, not affected in the ventral tegmental area [456]. A study in male Fischer 344 rats of inhalational exposures to ~500 MnO ug/m3 NPs (30 nm diameter) with either both nostrils patent or the right nostril occluded demonstrated that inhaled Mn is translocated via the olfactory neuronal pathway as a solid UFP to the central nervous system, where it produced inflammatory changes [457].
A series of studies in male rhesus monkeys of Mn solutes likewise confirmed olfactory uptake following inhalational exposure to MnSO4 solute at ≥0.6 mg Mn/m3 and increases in the brain regional Mn concentrations in a rostral-caudal plane [458]. As in studies with rodents, inhalational exposures of juvenile male rhesus monkey (0.06, 0.3 or 1.5 mg Mn/m3 for 6 h/day for 5 days/week for 65 days, or 1.5 mg Mn/m3 for 6 h/day for 5 days/week for 15, 33 or 65 days followed by a 45 or 90 day post exposure recovery period) led to brain region-specific changes in markers of oxidative stress and glutamate function, as well as evidence of reversibility post-exposure [459,460]. Some indirect findings in humans are indicated in a study of 70 children of 7–12 years of age living near a ferro-manganese alloy plant; in these studies, Mn in hair, used as a surrogate biomarker, was found to be inversely related to IQ and measures of verbal working memory. However, it must be recognized that the relationship between hair Mn and brain Mn is not clear, and the potential for other metal contaminants, e.g., excess Fe, is also possible [461]. As these studies indicate, Mn solutes and nanoparticles can likewise influence measures common to both NDDs and NDGDs, including to date, producing glutamatergic dysfunction and neuronal cell death as well as oxidative stress and mitochondrial impairments.
Sulfur is a trace element within AP as well as a requisite to human health [462], with sulfur dioxide (SO2) levels regulated by the U.S. Environmental Protection Agency (EPA). When inhaled, SO2 can be hydrated to sulfurous acid that then dissociates to produce sulfite/bisulfite and hydrogen ions; bisulfite can produce sulfur trioxide radical anions that react with oxygen leading to peroxyl radicals. When administered as SO2 to young (3 mos), middle-aged (12 mos) or old (24 mos) Swiss albino rats (sex not specified) at 10 ppm for 1 h/day for 7 days/week for 6 weeks, increases in the antioxidant enzyme Cu, Zn-SOD and decreases in glutathione peroxidase were seen in all three age groups, as were increases in the lipid peroxidation marker, thiobarbituric acid reactive substances (TBARS) [463,464]. In contrast, reductions in these measures were seen in a study that exposed Kunming albino mice of both sexes to 22, 56 or 112 mg/m3 of SO2 for 6 h day for 7 days, with no systematic evidence of sex differences [465,466,467]. Further analyses revealed increased levels of sulfite in brains as measured 18 hr following the exposures [468]; exposures to 28 or 56 mg/m3 were associated with brain ultrastructural changes that included glial cell damage, comprised of swelling and disrupted mitochondria, mitochondrial swelling within neurons, and the detachment of myelin along with mitochondrial vacuolization and outer membrane rupture [469].
Exposures of male Wistar rats to lower, but still relatively high levels of SO2 (3.5, 7 or 14 mg/m3 for 4 h/day for 30 days) resulted in increased mitochondrial biogenesis in the cortex, which the authors interpreted as an adaptive response to mitochondrial depletion following oxidative damage [470]. Assessment of synaptic plasticity at the two highest concentrations revealed ultrastructural changes in hippocampus (widening of the synaptic cleft, thinning of the postsynaptic density, a shortening of the synaptic active zone and reductions of synaptic vesicles), concentration-related reductions in synaptophysin expression, a marker of presynaptic terminals, as well as in protein phosphorylation markers related to synaptic plasticity (ERK1/2 and p-CREB) and the scaffolding marker PSD-95 [471]. Importantly, the role of exposure duration was examined by these authors in comparing the effects of 3.5 or 7 mg/m3 for 6 h/day for 4 weeks to an equal mass concentration of 14 or 28 mg/m3 for 6 h day for 1 week. In this comparison, measures such as glutamate receptor subunits mRNA in the hippocampus were inhibited in both exposure conditions, while synaptic plasticity markers such as protein kinase A and protein kinase C were changed in opposite directions. Importantly, this suggests that trying to equate the effects of acute exposures at higher concentrations to the effects of a lower concentration over a longer period of time, as is sometimes done to suggest that the more acute high level exposures are actually relevant to lower exposure concentrations over a more protracted period, is likely invalid [472,473], as previously reported for inhalation [474]. In another study by this group [475], exposures to SO2 of 3.5 or 7.0 mg/m3 (4–8 times the U.S. EPA standard) for 90 days was reported to impair memory in the probe trial of a water maze paradigm at the high concentration, while both concentrations reduced glutamate receptor subunit mRNA levels and increased levels of the pro-inflammatory cytokines TNFα, IL-1β, and IL-6 [475]. With respect to NDDs and NDGDs, S exposures have been shown to reproduce features including altered myelination, both glutamatergic and mitochondrial dysfunction, and oxidative stress.
Calcium is another required element that is found in AP. However, to date, no studies appear to have examined the impact of Ca overload in brain via instillation or inhalation, despite its potential involvement in both NDDs and NDGDs as cited above.
Studies have also examined the impact of such exposures to non-essential metals. Al has repeatedly been implicated in AD [133,476,477] and reported in high concentrations in brain in ASD [109]; it is considered to be a consistent component of ambient particulate matter. Like other metals, studies have demonstrated that exposure of rabbits to Al dust (otherwise not specified) at a dose of 0.56 mg Al/m3 for 8 h/day for 5 days/week for 160 days resulted in a 247% increase in brain Al concentrations which were not seen in kidney, heart or bone [478]; notably, Al in serum was only slightly raised, indicating that serum Al was not a good marker for brain Al levels, findings similar to those reported since for other metals [193]. Intranasal instillation in 7-week-old male Sprague Dawley rats with Al NPs (5–100 nm diameter) at a dose of 1, 20 or 40 mg/kg body weight a total of 3 times within one week resulted in dose-related increases in olfactory bulb and whole brain levels of Al as determined 48 h post termination of exposure. It also resulted in significant changes in expression of genes primarily related to signal transduction, cell differentiation, transcription, transport, response to stress and apoptosis and in protein levels of MAPK signaling proteins p38 and ERK1, critical to signal transduction [479]. In a study of male and female mice exposed via inhalation to 0.5 mg/m3 Al2O3 NPs (average 40 nm diameter) for 6 h/day for 28 consecutive days, increases in cortex but not midbrain Al levels were seen in both sexes, but only females showed behavioral alterations that included increased time spent in the periphery of an open field, as well as increased immobility in a forced swim test, which the authors interpreted as a depressive-like behavioral phenotype [480]. H&E staining did not reveal any brain pathology, but numerous sex by Al exposure-related interactions were found in the altered expression of genes involved in neurotransmission, phosphatidic acid synthesis, and voltage-gated ion channel function. While a significant literature has accumulated demonstrating Al neurotoxicity, it has largely been via oral exposure routes, hence the ability to assess its impact on brain specifically from airborne pollution is as yet unknown.
Similarly, despite high emissions of Si, limited information is currently available on its in vivo toxicity via inhalation [481], although it is known to be translocated to brain via routes that include olfactory neurons [191]. Intranasal exposure of male Wistar rats to either 10 nm or 80 nm Si NPs (150 µg/50 µL) daily for 30 days [482] resulted in marked accumulation of Si in frontal cortex, hippocampus and striatum where it markedly increased levels of malondialdehyde, a marker of lipid peroxidation, as well as H2O2 levels, while decreasing levels of antioxidant function, including Mn SOD, glutathione reductase, catalase, and reduced glutathione. Additionally, increases in the protein levels of NF-κB, TNF-α, and IL-1β were also observed, consistent with oxidative stress-based effects.
Some recent studies have examined the impact of PbO NPs. In one such study, mice were exposed in whole-body inhalation chambers to PbO for 4–72 h (acute exposure: 4.05 × 106 PbO NPs/cm3) or for 1–11 weeks (subchronic) to a lower (3.83 × 105 particles/cm3) or higher (1.93 × 106 particles/cm3) concentration. Pb accumulation in brain increased over time in response to subchronic exposures, and histopathological analyses revealed changes in the hippocampus after 11 weeks consisting of spongiform changes consistent with degeneration, neuronal vacuolization and necrotic neurons [483]. In addition to producing an increase in brain Pb concentrations over time, a study of continuous inhalation exposure of mice to 1.93 × 106 nanoparticles/cm3 for 2, 5, or 13 weeks reported increases in brain in the oxidative stress-related markers TBARS, 8-isoPGF, and 8-isoPGE2 [484]. A 5-day exposure for 4 h/day via nose only of female rats to 1.3 mg/m3 PbO nanoparticles (mean diameter of 36 nm) revealed nanoparticle inclusion in neurons and vacuolized cytoplasm, as well as membrane demyelination [485]. Thus, inhaled Pb nanoparticles have been related to altered myelination, neuronal cell death, and oxidative stress.

8. Conclusions and Future Research Needs

To date, the number of studies examining in vivo inhalation exposures to the profile of trace elements and metals within AP in relation to central nervous system deficits is relatively limited, and comparisons can be hindered by the fact that the exposure concentrations and specific metal chemistry as well as exposure parameters do vary across studies, rendering direct comparisons difficult. Furthermore, not all studies have examined all of the shared characteristics (Table 1). Nevertheless, three generally consistent effects have begun to emerge across metals and trace elements in these studies to date—namely, neuronal death, oxidative stress, and mitochondrial dysfunction, thus including two features considered mechanistic drivers of NDDs and NDGDs. Similar trends for metals and trace elements reproducing characteristics of NDDs and NDGDs are also seen for alterations in myelination, as well as inflammation and microglial activation.
Unfortunately, however, there remain more questions in understanding than answers. For example, is one element more potent than others in producing these characteristics—i.e., which element most faithfully reproduces these characteristics? Alternatively, are combinations of metals responsible? It could be that single elements or even combinations of elements are primarily involved in specific NDDs or NDGDs, or that particular elements relate to specific features of these diseases and disorders. The determination of these relationships will require additional assessments of such shared features in response to exposures to individual metals and trace elements, followed by combinations of elements. In addition to the missing information, as seen in Table 1, very little information on the behavioral consequences of such exposures is available. An additional method that may help identify the role of an element in a specific NDD or NDGD is to examine its impact on behavioral functions known to be unique to specific NDDs or NDGDs, particularly the behavioral aberrations that are most characteristic of the disorder. For example, deficits in communications and social behaviors are distinctive in ASD, while alterations in pre-pulse inhibition are well documented in SCZ.
Most NDDs and NDGDs are highly heterogeneous in phenotypic expression. It is very likely in the case of NDDs in particular that this could reflect differences in the timing during the period of brain development when exposures to risk factors occur. In the case of metal and trace element contaminants from AP, there will be differences across populations of pregnant women based on geography in the profile of trace element metal exposures from AP, as well as in the timing of those exposures. Such differences in exposure timing mean that these metals and trace elements are impacting the brain at different points in development, and thus could contribute to the heterogeneity of NDDs; studies examining different periods of exposure during brain development will be critical to assess this possibility. In the case of NDGDs, in addition to differences in the profiles of trace elements within AP, the duration of exposures may be an important consideration, particularly as it relates to the cumulative level of increase in the brain metal concentrations or reductions. Understanding the role played by these factors will likewise be important.
Notably, with only a few exceptions, studies to date have utilized only males in assessments. While this might enhance sensitivity to the role of AP-related trace elements in NDDs, most of which are male-biased, it eliminates any understanding of the differences between the sexes that may serve as an underlying point of vulnerability and thus aid the development of therapeutic interventions. In addition, while NDDs certainly exist in females, the associated symptoms often differ by sex. Moreover, some NDGDs, particularly AD and MS, have a female bias. Consequently, the inclusion of both sexes in these studies is imperative for a full understanding.
Inhalation as the route of exposure for studies of the questions posed here is the only directly relevant method. Many studies have utilized non-physiological intranasal or intratracheal instillation routes as a substitute, but deposition directly onto tissue via an aqueous medium differs from that in an airborne state resulting from inhalation; the physico-chemical processes underlying target cell–toxicant interactions are different. As just one example, although entering the blood stream after deposition in the respiratory tract, nanoparticles are unlikely to enter the CNS via the very tight blood brain barrier; however, they can directly access the brain through olfactory and trigeminal neural pathways, bypassing the blood brain barrier. Such limitations are also relevant to studies reported to assess air pollution using in vitro approaches, as the significant particle chemistry that would have occurred in air and other biological sites before it reaches isolated cells of interest is lacking.
Once uptake occurs, NPs adsorb biomolecules, forming a protein corona; some of these biomolecules adsorb with greater affinity and some with lesser affinity, such that the protein corona remains in a state of flux with the composition varying with time, resulting in both nanoparticle–protein and protein–protein interactions [486]. The composition of the protein corona has been shown to be size-dependent [487] and is influenced by composition of the media in in vitro studies [488]. The composition of the protein corona influences cellular uptake [489] as well. Collectively, these factors influence the biological and physiological responses of NPs, including toxicity. Of particular relevance to the impact on the brain, a recent study of engineered NPs found that the composition of the protein corona of gold NPs underwent marked molecular modifications, both quantitative and qualitative, during passage from the “blood” to the “brain” side of an in vitro transwell model of the blood brain barrier [490]. Clearly, a far better understanding of the protein corona composition and its impact on effects and associated dynamic changes in vivo for different NPs are needed, particularly for the brain.
Finally, many studies to date have utilized high or extremely high levels of exposure, even recognizing the differences in the routes of uptake (nasal breathing vs. oro-nasal breathing) and other kinetic differences between humans and rodents. Clearly, exposure-dose-response information will be important for furthering public health protection in determining the need for regulations.

Supplementary Materials

The following are available online at https://www.mdpi.com/2073-4433/11/10/1098/s1, Table S1: Cited Studies of Metal Inhalation or Intranasal Instillation in Relation to Impacts on Shared Characteristics of Neurodevelopmental Disorders and Neurodegenerative Diseases.

Funding

Funded by NIH P30 ES001247 and R01 ES025541.

Acknowledgments

Special thanks to Bob Gelein, David Chalupa, Elena Marvin and Katherine Conrad.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sunyer, J.; Dadvand, P. Pre-natal brain development as a target for urban air pollution. Basic Clin. Pharmacol. Toxicol. 2019, 125, 81–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Chen, J.C.; Wang, X.; Wellenius, G.A.; Serre, M.L.; Driscoll, I.; Casanova, R.; McArdle, J.J.; Manson, J.E.; Chui, H.C.; Espeland, M.A. Ambient air pollution and neurotoxicity on brain structure: Evidence from women’s health initiative memory study. Ann. Neurol. 2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Flores-Pajot, M.C.; Ofner, M.; Do, M.T.; Lavigne, E.; Villeneuve, P.J. Childhood autism spectrum disorders and exposure to nitrogen dioxide, and particulate matter air pollution: A review and meta-analysis. Environ. Res. 2016, 151, 763–776. [Google Scholar] [CrossRef]
  4. Annavarapu, R.N.; Kathi, S. Cognitive disorders in children associated with urban vehicular emissions. Environ. Pollut. 2016, 208, 74–78. [Google Scholar] [CrossRef] [PubMed]
  5. Kalkbrenner, A.E.; Windham, G.C.; Serre, M.L.; Akita, Y.; Wang, X.; Hoffman, K.; Thayer, B.P.; Daniels, J.L. Particulate matter exposure, prenatal and postnatal windows of susceptibility, and autism spectrum disorders. Epidemiology 2015, 26, 30–42. [Google Scholar] [CrossRef]
  6. Volk, H.E.; Lurmann, F.; Penfold, B.; Hertz-Picciotto, I.; McConnell, R. Traffic-related air pollution, particulate matter, and autism. JAMA Psychiatry 2013, 70, 71–77. [Google Scholar] [CrossRef]
  7. Volk, H.E.; Hertz-Picciotto, I.; Delwiche, L.; Lurmann, F.; McConnell, R. Residential proximity to freeways and autism in the CHARGE study. Environ. Health Perspect. 2011, 119, 873–877. [Google Scholar] [CrossRef]
  8. Fluegge, K. Does environmental exposure to the greenhouse gas, N2O, contribute to etiological factors in neurodevelopmental disorders? A mini-review of the evidence. Environ. Toxicol. Pharmacol. 2016, 47, 6–18. [Google Scholar] [CrossRef]
  9. Becerra, T.A.; Wilhelm, M.; Olsen, J.; Cockburn, M.; Ritz, B. Ambient air pollution and autism in Los Angeles county, California. Environ. Health Perspect. 2013, 121, 380–386. [Google Scholar] [CrossRef]
  10. Dickerson, A.S.; Rahbar, M.H.; Bakian, A.V.; Bilder, D.A.; Harrington, R.A.; Pettygrove, S.; Kirby, R.S.; Durkin, M.S.; Han, I.; Moye, L.A., 3rd; et al. Autism spectrum disorder prevalence and associations with air concentrations of lead, mercury, and arsenic. Environ. Monit. Assess. 2016, 188, 407. [Google Scholar] [CrossRef]
  11. Suades-Gonzalez, E.; Gascon, M.; Guxens, M.; Sunyer, J. Air pollution and neuropsychological development: A review of the latest evidence. Endocrinology 2015, 156, 3473–3482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Talbott, E.O.; Arena, V.C.; Rager, J.R.; Clougherty, J.E.; Michanowicz, D.R.; Sharma, R.K.; Stacy, S.L. Fine particulate matter and the risk of autism spectrum disorder. Environ. Res. 2015, 140, 414–420. [Google Scholar] [CrossRef]
  13. Raz, R.; Roberts, A.L.; Lyall, K.; Hart, J.E.; Just, A.C.; Laden, F.; Weisskopf, M.G. Autism spectrum disorder and particulate matter air pollution before, during, and after pregnancy: A nested case–control analysis within the Nurses’ Health Study II cohort. Environ. Health Perspect. 2015, 123, 264–270. [Google Scholar] [CrossRef]
  14. Jung, C.R.; Lin, Y.T.; Hwang, B.F. Air pollution and newly diagnostic autism spectrum disorders: A population-based cohort study in Taiwan. PLoS ONE 2013, 8, e75510. [Google Scholar] [CrossRef] [Green Version]
  15. Roberts, A.L.; Lyall, K.; Hart, J.E.; Laden, F.; Just, A.C.; Bobb, J.F.; Koenen, K.C.; Ascherio, A.; Weisskopf, M.G. Perinatal air pollutant exposures and autism spectrum disorder in the children of Nurses’ Health Study II participants. Environ. Health Perspect. 2013, 121, 978–984. [Google Scholar] [CrossRef] [Green Version]
  16. Pagalan, L.; Bickford, C.; Weikum, W.; Lanphear, B.; Brauer, M.; Lanphear, N.; Hanley, G.E.; Oberlander, T.F.; Winters, M. Association of prenatal exposure to air pollution with autism spectrum disorder. JAMA Pediatrics 2018. [Google Scholar] [CrossRef] [PubMed]
  17. Mintz, M. Evolution in the understanding of autism spectrum disorder: Historical perspective. Indian J. Pediatr. 2017, 84, 44–52. [Google Scholar] [CrossRef]
  18. Goldson, E. Advances in autism-2016. Adv. Pediatr. 2016, 63, 333–355. [Google Scholar] [CrossRef]
  19. van Rees, G.F.; Lago, S.G.; Cox, D.A.; Tomasik, J.; Rustogi, N.; Weigelt, K.; Ozcan, S.; Cooper, J.; Drexhage, H.; Leweke, F.M.; et al. Evidence of microglial activation following exposure to serum from first-onset drug-naive schizophrenia patients. Brain Behav. Immun. 2018, 67, 364–373. [Google Scholar] [CrossRef]
  20. Merchán-Naranjo, J.; Boada, L.; del Rey-Mejías, Á.; Mayoral, M.; Llorente, C.; Arango, C.; Parellada, M. Executive function is affected in autism spectrum disorder, but does not correlate with intelligence. Rev. Psiquiatr. Salud Ment. 2016, 9, 39–50. [Google Scholar] [CrossRef]
  21. Carter Leno, V.; Chandler, S.; White, P.; Pickles, A.; Baird, G.; Hobson, C.; Smith, A.B.; Charman, T.; Rubia, K.; Simonoff, E. Testing the specificity of executive functioning impairments in adolescents with ADHD, ODD/CD and ASD. Eur. Child Adolesc. Psychiatry 2018, 27, 899–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Uzefovsky, F.; Allison, C.; Smith, P.; Baron-Cohen, S. Brief Report: The go/no-go task online: Inhibitory control deficits in autism in a large sample. J. Autism Dev. Disord. 2016, 46, 2774–2779. [Google Scholar] [CrossRef] [Green Version]
  23. Allred, E.N.; Dammann, O.; Fichorova, R.N.; Hooper, S.R.; Hunter, S.J.; Joseph, R.M.; Kuban, K.; Leviton, A.; O’Shea, T.M.; Scott, M.N.; et al. Systemic inflammation during the first postnatal month and the risk of attention deficit hyperactivity disorder characteristics among 10 year-old children born extremely preterm. J. Neuroimmune Pharmacol. 2017, 12, 531–543. [Google Scholar] [CrossRef]
  24. Alemany, S.; Vilor-Tejedor, N.; Garcia-Esteban, R.; Bustamante, M.; Dadvand, P.; Esnaola, M.; Mortamais, M.; Forns, J.; van Drooge, B.L.; Alvarez-Pedrerol, M.; et al. Traffic-related air pollution, APOEepsilon4 status, and neurodevelopmental outcomes among school children enrolled in the BREATHE project (Catalonia, Spain). Environ. Health Perspect. 2018, 126, 087001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Fuertes, E.; Standl, M.; Forns, J.; Berdel, D.; Garcia-Aymerich, J.; Markevych, I.; Schulte-Koerne, G.; Sugiri, D.; Schikowski, T.; Tiesler, C.M.; et al. Traffic-related air pollution and hyperactivity/inattention, dyslexia and dyscalculia in adolescents of the German GINIplus and LISAplus birth cohorts. Environ. Int. 2016, 97, 85–92. [Google Scholar] [CrossRef]
  26. Sentis, A.; Sunyer, J.; Dalmau-Bueno, A.; Andiarena, A.; Ballester, F.; Cirach, M.; Estarlich, M.; Fernandez-Somoano, A.; Ibarluzea, J.; Iniguez, C.; et al. Prenatal and postnatal exposure to NO2 and child attentional function at 4-5years of age. Environ. Int. 2017, 106, 170–177. [Google Scholar] [CrossRef] [Green Version]
  27. Min, J.Y.; Min, K.B. Exposure to ambient PM10 and NO2 and the incidence of attention-deficit hyperactivity disorder in childhood. Environ. Int. 2017, 99, 221–227. [Google Scholar] [CrossRef]
  28. Newman, N.C.; Ryan, P.; Lemasters, G.; Levin, L.; Bernstein, D.; Hershey, G.K.; Lockey, J.E.; Villareal, M.; Reponen, T.; Grinshpun, S.; et al. Traffic-related air pollution exposure in the first year of life and behavioral scores at 7 years of age. Environ. Health Perspect. 2013, 121, 731–736. [Google Scholar] [CrossRef]
  29. Siddique, S.; Banerjee, M.; Ray, M.R.; Lahiri, T. Attention-deficit hyperactivity disorder in children chronically exposed to high level of vehicular pollution. Eur. J. Pediatrics 2011, 170, 923–929. [Google Scholar] [CrossRef]
  30. Luo, Y.; Weibman, D.; Halperin, J.M.; Li, X. A Review of heterogeneity in attention deficit/hyperactivity disorder (ADHD). Front. Hum. Neurosci. 2019, 13, 42. [Google Scholar] [CrossRef] [Green Version]
  31. Magnus, W.; Nazir, S.; Anilkumar, A.C.; Shaban, K. Attention Deficit Hyperactivity Disorder (ADHD); StatPearls Publishing LLC.: Treasure Island, FL, USA, 2019. [Google Scholar]
  32. Tandon, M.; Pergjika, A. Attention deficit hyperactivity disorder in preschool-age children. Child Adolesc. Psychiatr. Clin. N Am. 2017, 26, 523–538. [Google Scholar] [CrossRef]
  33. Hinshaw, S.P. Attention deficit hyperactivity disorder (ADHD): Controversy, developmental mechanisms, and multiple levels of analysis. Annu. Rev. Clin. Psychol. 2018, 14, 291–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ferretti, N.M.; King, S.L.; Hilton, D.C.; Rondon, A.T.; Jarrett, M.A. Social functioning in youth with attention-deficit/hyperactivity disorder and sluggish cognitive tempo. Yale J. Biol. Med. 2019, 92, 29–35. [Google Scholar]
  35. Dean, L. Schizophrenia. In Medical Genetics Summaries; Pratt, V., McLeod, H., Rubinstein, W., Dean, L., Kattman, B., Malheiro, A., Eds.; National Center for Biotechnology Information: Bethesda, MD, USA, 2012. [Google Scholar]
  36. Newbury, J.B.; Arseneault, L.; Beevers, S.; Kitwiroon, N.; Roberts, S.; Pariante, C.M.; Kelly, F.J.; Fisher, H.L. Association of air pollution exposure with psychotic experiences during adolescence. JAMA Psychiatry 2019, 76, 614–623. [Google Scholar] [CrossRef]
  37. Duan, J.; Luo, X.L.; Chu, W.W.; Gao, J.J.; Xu, Z.H.; Zhang, Y.W.; Cheng, Q.; Bai, L.J.; Wei, Q.N.; Su, H. Time series analysis on the effect of ambient fine particulate matters and temperature interactions on schizophrenia admission in Tongling City of Anhui Province, 2014–2017. Zhonghua Yu Fang Yi Xue Za Zhi 2019, 53, 51–56. [Google Scholar] [CrossRef]
  38. Qiu, H.; Zhu, X.; Wang, L.; Pan, J.; Pu, X.; Zeng, X.; Zhang, L.; Peng, Z.; Zhou, L. Attributable risk of hospital admissions for overall and specific mental disorders due to particulate matter pollution: A time-series study in Chengdu, China. Environ. Res. 2019, 170, 230–237. [Google Scholar] [CrossRef]
  39. Liang, Z.; Xu, C.; Cao, Y.; Kan, H.D.; Chen, R.J.; Yao, C.Y.; Liu, X.L.; Xiang, Y.; Wu, N.; Wu, L.; et al. The association between short-term ambient air pollution and daily outpatient visits for schizophrenia: A hospital-based study. Environ. Pollut. 2019, 244, 102–108. [Google Scholar] [CrossRef]
  40. Duan, J.; Cheng, Q.; Luo, X.; Bai, L.; Zhang, H.; Wang, S.; Xu, Z.; Gao, J.; Zhang, Y.; Su, H. Is the serious ambient air pollution associated with increased admissions for schizophrenia? Sci. Total Environ. 2018, 644, 14–19. [Google Scholar] [CrossRef]
  41. Eguchi, R.; Onozuka, D.; Ikeda, K.; Kuroda, K.; Ieiri, I.; Hagihara, A. The relationship between fine particulate matter (PM2.5) and schizophrenia severity. Int. Arch. Occup. Environ. Health 2018, 91, 613–622. [Google Scholar] [CrossRef]
  42. Yackerson, N.S.; Zilberman, A.; Todder, D.; Kaplan, Z. The influence of air-suspended particulate concentration on the incidence of suicide attempts and exacerbation of schizophrenia. Int. J. Biometeorol. 2014, 58, 61–67. [Google Scholar] [CrossRef]
  43. Pedersen, C.B.; Raaschou-Nielsen, O.; Hertel, O.; Mortensen, P.B. Air pollution from traffic and schizophrenia risk. Schizophr. Res. 2004, 66, 83–85. [Google Scholar] [CrossRef]
  44. Pinares-Garcia, P.; Stratikopoulos, M.; Zagato, A.; Loke, H.; Lee, J. Sex: A significant risk factor for neurodevelopmental and neurodegenerative disorders. Brain Sci. 2018, 8, 154. [Google Scholar] [CrossRef] [Green Version]
  45. Tsai, T.L.; Lin, Y.T.; Hwang, B.F.; Nakayama, S.F.; Tsai, C.H.; Sun, X.L.; Ma, C.; Jung, C.R. Fine particulate matter is a potential determinant of Alzheimer’s disease: A systemic review and meta-analysis. Environ. Res. 2019, 177, 108638. [Google Scholar] [CrossRef]
  46. Younan, D.; Petkus, A.J.; Widaman, K.F.; Wang, X.; Casanova, R.; Espeland, M.A.; Gatz, M.; Henderson, V.W.; Manson, J.E.; Rapp, S.R.; et al. Particulate matter and episodic memory decline mediated by early neuroanatomic biomarkers of Alzheimer’s disease. Brain 2020, 143, 289–302. [Google Scholar] [CrossRef] [Green Version]
  47. Fu, P.; Guo, X.; Cheung, F.M.H.; Yung, K.K.L. The association between PM(2.5) exposure and neurological disorders: A systematic review and meta-analysis. Sci. Total Environ. 2019, 655, 1240–1248. [Google Scholar] [CrossRef]
  48. Oudin, A.; Forsberg, B.; Adolfsson, A.N.; Lind, N.; Modig, L.; Nordin, M.; Nordin, S.; Adolfsson, R.; Nilsson, L.G. Traffic-related air pollution and dementia incidence in northern Sweden: A longitudinal study. Environ. Health Perspect. 2016, 124, 306–312. [Google Scholar] [CrossRef]
  49. Jung, C.R.; Lin, Y.T.; Hwang, B.F. Ozone, particulate matter, and newly diagnosed Alzheimer’s disease: A population-based cohort study in Taiwan. J. Alzheimers Dis. 2015, 44, 573–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Ajmani, G.S.; Suh, H.H.; Pinto, J.M. Effects of ambient air pollution exposure on olfaction: A review. Environ. Health Perspect. 2016, 124, 1683–1693. [Google Scholar] [CrossRef]
  51. Han, C.; Lu, Y.; Cheng, H.; Wang, C.; Chan, P. The impact of long-term exposure to ambient air pollution and second-hand smoke on the onset of Parkinson disease: A review and meta-analysis. Public Health 2020, 179, 100–110. [Google Scholar] [CrossRef]
  52. Kasdagli, M.I.; Katsouyanni, K.; Dimakopoulou, K.; Samoli, E. Air pollution and Parkinson’s disease: A systematic review and meta-analysis up to 2018. Int. J. Hyg. Environ. Health 2019, 222, 402–409. [Google Scholar] [CrossRef]
  53. Hu, C.Y.; Fang, Y.; Li, F.L.; Dong, B.; Hua, X.G.; Jiang, W.; Zhang, H.; Lyu, Y.; Zhang, X.J. Association between ambient air pollution and Parkinson’s disease: Systematic review and meta-analysis. Environ. Res. 2019, 168, 448–459. [Google Scholar] [CrossRef]
  54. Shin, S.; Burnett, R.T.; Kwong, J.C.; Hystad, P.; van Donkelaar, A.; Brook, J.R.; Copes, R.; Tu, K.; Goldberg, M.S.; Villeneuve, P.J.; et al. Effects of ambient air pollution on incident Parkinson’s disease in Ontario, 2001 to 2013: A population-based cohort study. Int. J. Epidemiol. 2018, 47, 2038–2048. [Google Scholar] [CrossRef]
  55. Chen, C.Y.; Hung, H.J.; Chang, K.H.; Hsu, C.Y.; Muo, C.H.; Tsai, C.H.; Wu, T.N. Long-term exposure to air pollution and the incidence of Parkinson’s disease: A nested case-control study. PLoS ONE 2017, 12, e0182834. [Google Scholar] [CrossRef] [Green Version]
  56. Lee, P.C.; Liu, L.L.; Sun, Y.; Chen, Y.A.; Liu, C.C.; Li, C.Y.; Yu, H.L.; Ritz, B. Traffic-related air pollution increased the risk of Parkinson’s disease in Taiwan: A nationwide study. Environ. Int. 2016, 96, 75–81. [Google Scholar] [CrossRef]
  57. Ritz, B.; Lee, P.C.; Hansen, J.; Lassen, C.F.; Ketzel, M.; Sorensen, M.; Raaschou-Nielsen, O. Traffic-related air pollution and Parkinson’s disease in Denmark: A case-control study. Environ. Health Perspect. 2016, 124, 351–356. [Google Scholar] [CrossRef] [Green Version]
  58. Toro, R.; Downward, G.S.; van der Mark, M.; Brouwer, M.; Huss, A.; Peters, S.; Hoek, G.; Nijssen, P.; Mulleners, W.M.; Sas, A.; et al. Parkinson’s disease and long-term exposure to outdoor air pollution: A matched case-control study in The Netherlands. Environ. Int. 2019, 129, 28–34. [Google Scholar] [CrossRef]
  59. Palacios, N.; Fitzgerald, K.C.; Hart, J.E.; Weisskopf, M.; Schwarzschild, M.A.; Ascherio, A.; Laden, F. Air pollution and risk of Parkinson’s disease in a large prospective study of men. Environ. Health Perspect. 2017, 125, 087011. [Google Scholar] [CrossRef]
  60. Palacios, N.; Fitzgerald, K.C.; Hart, J.E.; Weisskopf, M.G.; Schwarzschild, M.A.; Ascherio, A.; Laden, F. Particulate matter and risk of Parkinson disease in a large prospective study of women. Environ. Health 2014, 13, 80. [Google Scholar] [CrossRef] [Green Version]
  61. Tateo, F.; Grassivaro, F.; Ermani, M.; Puthenparampil, M.; Gallo, P. PM2.5 levels strongly associate with multiple sclerosis prevalence in the Province of Padua, Veneto Region, North-East Italy. Mult. Scler. J. 2019, 25, 1719–1727. [Google Scholar] [CrossRef]
  62. Jeanjean, M.; Bind, M.A.; Roux, J.; Ongagna, J.C.; de Sèze, J.; Bard, D.; Leray, E. Ozone, NO(2) and PM(10) are associated with the occurrence of multiple sclerosis relapses. Evidence from seasonal multi-pollutant analyses. Environ. Res. 2018, 163, 43–52. [Google Scholar] [CrossRef] [Green Version]
  63. Bergamaschi, R.; Cortese, A.; Pichiecchio, A.; Berzolari, F.G.; Borrelli, P.; Mallucci, G.; Bollati, V.; Romani, A.; Nosari, G.; Villa, S.; et al. Air pollution is associated to the multiple sclerosis inflammatory activity as measured by brain MRI. Mult. Scler. J. 2018, 24, 1578–1584. [Google Scholar] [CrossRef]
  64. Roux, J.; Bard, D.; Le Pabic, E.; Segala, C.; Reis, J.; Ongagna, J.C.; de Sèze, J.; Leray, E. Air pollution by particulate matter PM(10) may trigger multiple sclerosis relapses. Environ. Res. 2017, 156, 404–410. [Google Scholar] [CrossRef]
  65. Heydarpour, P.; Amini, H.; Khoshkish, S.; Seidkhani, H.; Sahraian, M.A.; Yunesian, M. Potential impact of air pollution on multiple sclerosis in Tehran, Iran. Neuroepidemiology 2014, 43, 233–238. [Google Scholar] [CrossRef] [Green Version]
  66. Oikonen, M.; Laaksonen, M.; Laippala, P.; Oksaranta, O.; Lilius, E.M.; Lindgren, S.; Rantio-Lehtimaki, A.; Anttinen, A.; Koski, K.; Eralinna, J.P. Ambient air quality and occurrence of multiple sclerosis relapse. Neuroepidemiology 2003, 22, 95–99. [Google Scholar] [CrossRef]
  67. Palacios, N.; Munger, K.L.; Fitzgerald, K.C.; Hart, J.E.; Chitnis, T.; Ascherio, A.; Laden, F. Exposure to particulate matter air pollution and risk of multiple sclerosis in two large cohorts of US nurses. Environ. Int. 2017, 109, 64–72. [Google Scholar] [CrossRef]
  68. Visser, A.E.; D’Ovidio, F.; Peters, S.; Vermeulen, R.C.; Beghi, E.; Chiò, A.; Veldink, J.H.; Logroscino, G.; Hardiman, O.; van den Berg, L.H. Multicentre, population-based, case-control study of particulates, combustion products and amyotrophic lateral sclerosis risk. J. Neurol. Neurosurg. Psychiatry 2019, 90, 854–860. [Google Scholar] [CrossRef]
  69. Myung, W.; Lee, H.; Kim, H. Short-term air pollution exposure and emergency department visits for amyotrophic lateral sclerosis: A time-stratified case-crossover analysis. Environ. Int. 2019, 123, 467–475. [Google Scholar] [CrossRef]
  70. Seelen, M.; Toro Campos, R.A.; Veldink, J.H.; Visser, A.E.; Hoek, G.; Brunekreef, B.; van der Kooi, A.J.; de Visser, M.; Raaphorst, J.; van den Berg, L.H.; et al. Long-term air pollution exposure and amyotrophic lateral sclerosis in Netherlands: A population-based case-control study. Environ. Health Perspect. 2017, 125, 097023. [Google Scholar] [CrossRef] [Green Version]
  71. Pujol, J.; Martinez-Vilavella, G.; Macia, D.; Fenoll, R.; Alvarez-Pedrerol, M.; Rivas, I.; Forns, J.; Blanco-Hinojo, L.; Capellades, J.; Querol, X.; et al. Traffic pollution exposure is associated with altered brain connectivity in school children. NeuroImage 2016, 129, 175–184. [Google Scholar] [CrossRef] [Green Version]
  72. Pujol, J.; Fenoll, R.; Macia, D.; Martinez-Vilavella, G.; Alvarez-Pedrerol, M.; Rivas, I.; Forns, J.; Deus, J.; Blanco-Hinojo, L.; Querol, X.; et al. Airborne copper exposure in school environments associated with poorer motor performance and altered basal ganglia. Brain Behav. 2016, 6, e00467. [Google Scholar] [CrossRef] [Green Version]
  73. Mezzaroba, L.; Alfieri, D.F.; Simão, A.N.C.; Reiche, E.M.V. The role of zinc, copper, manganese and iron in neurodegenerative diseases. Neurotoxicology 2019, 74, 230–241. [Google Scholar] [CrossRef]
  74. Guxens, M.; Lubczynska, M.J.; Muetzel, R.L.; Dalmau-Bueno, A.; Jaddoe, V.W.V.; Hoek, G.; van der Lugt, A.; Verhulst, F.C.; White, T.; Brunekreef, B.; et al. Air pollution exposure during fetal life, brain morphology, and cognitive function in school-age children. Biol. Psychiatry 2018, 84, 295–303. [Google Scholar] [CrossRef] [Green Version]
  75. Harris, M.H.; Gold, D.R.; Rifas-Shiman, S.L.; Melly, S.J.; Zanobetti, A.; Coull, B.A.; Schwartz, J.D.; Gryparis, A.; Kloog, I.; Koutrakis, P.; et al. Prenatal and childhood traffic-related pollution exposure and childhood cognition in the Project viva cohort (Massachusetts, USA). Environ. Health Perspect. 2015, 123, 1072–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Lin, C.C.; Chen, S.J.; Huang, K.L.; Hwang, W.I.; Chang-Chien, G.P.; Lin, W.Y. Characteristics of metals in nano/ultrafine/fine/coarse particles collected beside a heavily trafficked road. Environ. Sci. Technol. 2005, 39, 8113–8122. [Google Scholar] [CrossRef]
  77. Friedlander, S.K. Chemical element balances and identification of air pollution sources. Environ. Sci. Technol. 1973, 7, 235–240. [Google Scholar] [CrossRef]
  78. Ruuskanen, J.; Tuch, T.; Brink, T.; Peters, A.; Khlystov, A.; Mirme, A.; Kos, G.P.A.; Brunekreef, B.; Wichmann, H.E.; Buzorius, G.; et al. Concentrations of ultrafine, fine and PM2.5 particles in three European cities. Atmos. Environ. 2001, 35, 3729–3738. [Google Scholar] [CrossRef]
  79. Tuch, T.; Brand, P.; Wichmann, H.E.; Heyder, J. Variation of particle number and mass concentration in various size ranges of ambient aerosols in Eastern Germany. Atmos. Environ. 1997, 31, 4193–4197. [Google Scholar] [CrossRef]
  80. Knibbs, L.D.; de Dear, R.J. Exposure to ultrafine particles and PM2.5 in four Sydney transport modes. Atmos. Environ. 2010, 44, 3224–3227. [Google Scholar] [CrossRef] [Green Version]
  81. Friedlander, S.K. Gas-to-particle conversion. In Smoke, Dust and Haze: Fundamentals of Aerosol Dynamics; Oxford University Press: New York, NY, USA, 2000; pp. 275–305. [Google Scholar]
  82. Kulmala, M.; Kerminen, V.M.; Petäjä, T.; Ding, A.J.; Wang, L. Atmospheric gas-to-particle conversion: Why NPF events are observed in megacities? Faraday Discuss. 2017, 200, 271–288. [Google Scholar] [CrossRef]
  83. Reff, A.; Bhave, P.V.; Simon, H.; Pace, T.G.; Pouliot, G.A.; Mobley, J.D.; Houyoux, M. Emissions inventory of PM2.5 trace elements across the United States. Environ. Sci. Technol. 2009, 43, 5790–5796. [Google Scholar] [CrossRef]
  84. Fang, T.; Guo, H.Y.; Zeng, L.H.; Verma, V.; Nenes, A.; Weber, R.J. Highly acidic ambient particles, soluble metals, and oxidative potential: A link between sulfate and aerosol toxicity. Environ. Sci. Technol. 2017, 51, 2611–2620. [Google Scholar] [CrossRef] [PubMed]
  85. Watson, A.Y.; Brain, J.D. The effect of SO2 on the uptake of particles by mouse bronchial epithelium. Exp. Lung Res. 1980, 1, 67–87. [Google Scholar] [CrossRef] [PubMed]
  86. Allen, J.L.; Conrad, K.; Oberdorster, G.; Johnston, C.J.; Sleezer, B.; Cory-Slechta, D.A. Developmental exposure to concentrated ambient particles and preference for immediate reward in mice. Environ. Health Perspect. 2013, 121, 32–38. [Google Scholar] [CrossRef]
  87. Allen, J.L.; Klocke, C.; Morris-Schaffer, K.; Conrad, K.; Sobolewski, M.; Cory-Slechta, D.A. Cognitive effects of air pollution exposures and potential mechanistic underpinnings. Curr. Environ. Health Rep. 2017, 4, 180–191. [Google Scholar] [CrossRef]
  88. Allen, J.L.; Liu, X.; Pelkowski, S.; Palmer, B.; Conrad, K.; Oberdorster, G.; Weston, D.; Mayer-Proschel, M.; Cory-Slechta, D.A. Early postnatal exposure to ultrafine particulate matter air pollution: Persistent ventriculomegaly, neurochemical disruption, and glial activation preferentially in male mice. Environ. Health Perspect. 2014, 122, 939–945. [Google Scholar] [CrossRef]
  89. Allen, J.L.; Liu, X.; Weston, D.; Conrad, K.; Oberdorster, G.; Cory-Slechta, D.A. Consequences of developmental exposure to concentrated ambient ultrafine particle air pollution combined with the adult paraquat and maneb model of the Parkinson’s disease phenotype in male mice. Neurotoxicology 2014, 41, 80–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Allen, J.L.; Liu, X.; Weston, D.; Prince, L.; Oberdorster, G.; Finkelstein, J.N.; Johnston, C.J.; Cory-Slechta, D.A. Developmental exposure to concentrated ambient ultrafine particulate matter air pollution in mice results in persistent and sex-dependent behavioral neurotoxicity and glial activation. Toxicol. Sci. 2014, 140, 160–178. [Google Scholar] [CrossRef] [Green Version]
  91. Cory-Slechta, D.A.; Allen, J.L.; Conrad, K.; Marvin, E.; Sobolewski, M. Developmental exposure to low level ambient ultrafine particle air pollution and cognitive dysfunction. Neurotoxicology 2017. [Google Scholar] [CrossRef]
  92. Cory-Slechta, D.A.; Sobolewski, M.; Marvin, E.; Conrad, K.; Merrill, A.; Anderson, T.; Jackson, B.P.; Oberdorster, G. The impact of inhaled ambient ultrafine particulate matter on developing brain: Potential importance of elemental contaminants. Toxicol. Pathol. 2019, 47, 976–992. [Google Scholar] [CrossRef]
  93. Sobolewski, M.; Anderson, T.; Conrad, K.; Marvin, E.; Klocke, C.; Morris-Schaffer, K.; Allen, J.L.; Cory-Slechta, D.A. Developmental exposures to ultrafine particle air pollution reduces early testosterone levels and adult male social novelty preference: Risk for children’s sex-biased neurobehavioral disorders. NeuroToxicology 2018, 68, 203–211. [Google Scholar] [CrossRef]
  94. Bandeira, F.; Lent, R.; Herculano-Houzel, S. Changing numbers of neuronal and non-neuronal cells underlie postnatal brain growth in the rat. Proc. Natl. Acad. Sci. USA 2009, 106, 14108–14113. [Google Scholar] [CrossRef] [Green Version]
  95. Klocke, C.; Allen, J.L.; Sobolewski, M.; Blum, J.L.; Zelikoff, J.T.; Cory-Slechta, D.A. Exposure to fine and ultrafine particulate matter during gestation alters postnatal oligodendrocyte maturation, proliferation capacity, and myelination. Neurotoxicology 2018, 65, 196–206. [Google Scholar] [CrossRef]
  96. Klocke, C.; Allen, J.L.; Sobolewski, M.; Mayer-Pröschel, M.; Blum, J.L.; Lauterstein, D.; Zelikoff, J.T.; Cory-Slechta, D.A. Neuropathological consequences of gestational exposure to concentrated ambient fine and ultrafine particles in the mouse. Toxicol. Sci. 2017, 156, 492–508. [Google Scholar] [CrossRef]
  97. Klocke, C.; Sherina, V.; Graham, U.M.; Gunderson, J.; Allen, J.L.; Sobolewski, M.; Blum, J.L.; Zelikoff, J.T.; Cory-Slechta, D.A. Enhanced cerebellar myelination with concomitant iron elevation and ultrastructural irregularities following prenatal exposure to ambient particulate matter in the mouse. Inhal. Toxicol. 2018, 30, 381–396. [Google Scholar] [CrossRef]
  98. Scassellati, C.; Bonvicini, C.; Benussi, L.; Ghidoni, R.; Squitti, R. Neurodevelopmental disorders: Metallomics studies for the identification of potential biomarkers associated to diagnosis and treatment. J. Trace Elem. Med. Biol. 2020, 60, 126499. [Google Scholar] [CrossRef]
  99. Adams, J.B.; Audhya, T.; McDonough-Means, S.; Rubin, R.A.; Quig, D.; Geis, E.; Gehn, E.; Loresto, M.; Mitchell, J.; Atwood, S.; et al. Toxicological status of children with autism vs. neurotypical children and the association with autism severity. Biol. Trace Elem. Res. 2013, 151, 171–180. [Google Scholar] [CrossRef]
  100. Fiore, M.; Barone, R.; Copat, C.; Grasso, A.; Cristaldi, A.; Rizzo, R.; Ferrante, M. Metal and essential element levels in hair and association with autism severity. J. Trace Elem. Med. Biol. 2020, 57, 126409. [Google Scholar] [CrossRef]
  101. Lakshmi Priya, M.D.; Geetha, A. Level of trace elements (copper, zinc, magnesium and selenium) and toxic elements (lead and mercury) in the hair and nail of children with autism. Biol. Trace Elem. Res. 2011, 142, 148–158. [Google Scholar] [CrossRef]
  102. Li, S.O.; Wang, J.L.; Bjørklund, G.; Zhao, W.N.; Yin, C.H. Serum copper and zinc levels in individuals with autism spectrum disorders. Neuroreport 2014, 25, 1216–1220. [Google Scholar] [CrossRef]
  103. Saghazadeh, A.; Ahangari, N.; Hendi, K.; Saleh, F.; Rezaei, N. Status of essential elements in autism spectrum disorder: Systematic review and meta-analysis. Rev. Neurosci. 2017, 28, 783–809. [Google Scholar] [CrossRef] [PubMed]
  104. Skalny, A.V.; Simashkova, N.V.; Klyushnik, T.P.; Grabeklis, A.R.; Radysh, I.V.; Skalnaya, M.G.; Tinkov, A.A. Analysis of hair trace elements in children with autism spectrum disorders and communication disorders. Biol. Trace Elem. Res. 2017, 177, 215–223. [Google Scholar] [CrossRef]
  105. Tinkov, A.A.; Skalnaya, M.G.; Simashkova, N.V.; Klyushnik, T.P.; Skalnaya, A.A.; Bjørklund, G.; Notova, S.V.; Kiyaeva, E.V.; Skalny, A.V. Association between catatonia and levels of hair and serum trace elements and minerals in autism spectrum disorder. Biomed. Pharmacother. 2019, 109, 174–180. [Google Scholar] [CrossRef] [PubMed]
  106. Wu, J.; Liu, D.J.; Shou, X.J.; Zhang, J.S.; Meng, F.C.; Liu, Y.Q.; Han, S.P.; Zhang, R.; Jia, J.Z.; Wang, J.Y.; et al. Chinese children with autism: A multiple chemical elements profile in erythrocytes. Autism Res. 2018, 11, 834–845. [Google Scholar] [CrossRef]
  107. Zhai, Q.; Cen, S.; Jiang, J.; Zhao, J.; Zhang, H.; Chen, W. Disturbance of trace element and gut microbiota profiles as indicators of autism spectrum disorder: A pilot study of Chinese children. Environ. Res. 2019, 171, 501–509. [Google Scholar] [CrossRef]
  108. Skalny, A.V.; Simashkova, N.V.; Skalnaya, A.A.; Klyushnik, T.P.; Bjørklund, G.; Skalnaya, M.G.; Tinkov, A.A. Assessment of gender and age effects on serum and hair trace element levels in children with autism spectrum disorder. Metab. Brain Dis. 2017, 32, 1675–1684. [Google Scholar] [CrossRef] [PubMed]
  109. Mold, M.; Umar, D.; King, A.; Exley, C. Aluminium in brain tissue in autism. J. Trace Elem. Med. Biol. 2018, 46, 76–82. [Google Scholar] [CrossRef]
  110. Exley, C.; Clarkson, E. Aluminium in human brain tissue from donors without neurodegenerative disease: A comparison with Alzheimer’s disease, multiple sclerosis and autism. Sci. Rep. 2020, 10, 7770. [Google Scholar] [CrossRef]
  111. Skalny, A.V.; Simashkova, N.V.; Skalnaya, A.A.; Klyushnik, T.P.; Zhegalova, I.V.; Grabeklis, A.R.; Skalnaya, M.G.; Tinkov, A.A. Trace element levels are associated with neuroinflammatory markers in children with autistic spectrum disorder. J. Trace Elem. Med. Biol. 2018, 50, 622–628. [Google Scholar] [CrossRef]
  112. Cai, L.; Chen, T.; Yang, J.; Zhou, K.; Yan, X.; Chen, W.; Sun, L.; Li, L.; Qin, S.; Wang, P.; et al. Serum trace element differences between Schizophrenia patients and controls in the Han Chinese population. Sci. Rep. 2015, 5, 15013. [Google Scholar] [CrossRef] [Green Version]
  113. Gillin, J.C.; Carpenter, W.T.; Hambidge, K.M.; Wyatt, R.J.; Henkin, R.I. Zinc and copper in patients with schizophrenia. Encephale 1982, 8, 435–444. [Google Scholar]
  114. Joe, P.; Petrilli, M.; Malaspina, D.; Weissman, J. Zinc in schizophrenia: A meta-analysis. Gen. Hosp. Psychiatry 2018, 53, 19–24. [Google Scholar] [CrossRef]
  115. Ma, J.; Wang, B.; Gao, X.; Wu, H.; Wang, D.; Li, N.; Tan, J.; Wang, J.; Yan, L. A comparative study of the typical toxic metals in serum by patients of schizophrenia and healthy controls in China. Psychiatry Res. 2018, 269, 558–564. [Google Scholar] [CrossRef]
  116. Ma, J.; Yan, L.; Guo, T.; Yang, S.; Guo, C.; Liu, Y.; Xie, Q.; Wang, J. Association of typical toxic heavy metals with Schizophrenia. Int. J. Environ. Res. Public Health 2019, 16, 4200. [Google Scholar] [CrossRef] [Green Version]
  117. Modabbernia, A.; Velthorst, E.; Gennings, C.; De Haan, L.; Austin, C.; Sutterland, A.; Mollon, J.; Frangou, S.; Wright, R.; Arora, M.; et al. Early-life metal exposure and schizophrenia: A proof-of-concept study using novel tooth-matrix biomarkers. Eur. Psychiatry 2016, 36, 1–6. [Google Scholar] [CrossRef] [Green Version]
  118. Vidović, B.; Dorđević, B.; Milovanović, S.; Škrivanj, S.; Pavlović, Z.; Stefanović, A.; Kotur-Stevuljević, J. Selenium, zinc, and copper plasma levels in patients with schizophrenia: Relationship with metabolic risk factors. Biol. Trace Elem. Res. 2013, 156, 22–28. [Google Scholar] [CrossRef]
  119. Yanik, M.; Kocyigit, A.; Tutkun, H.; Vural, H.; Herken, H. Plasma manganese, selenium, zinc, copper, and iron concentrations in patients with schizophrenia. Biol. Trace Elem. Res. 2004, 98, 109–117. [Google Scholar] [CrossRef]
  120. Adisetiyo, V.; Helpern, J.A. Brain iron: A promising noninvasive biomarker of attention-deficit/hyperactivity disorder that warrants further investigation. Biomark. Med. 2015, 9, 403–406. [Google Scholar] [CrossRef]
  121. Adisetiyo, V.; Jensen, J.H.; Tabesh, A.; Deardorff, R.L.; Fieremans, E.; Di Martino, A.; Gray, K.M.; Castellanos, F.X.; Helpern, J.A. Multimodal MR imaging of brain iron in attention deficit hyperactivity disorder: A noninvasive biomarker that responds to psychostimulant treatment? Radiology 2014, 272, 524–532. [Google Scholar] [CrossRef] [Green Version]
  122. Arnold, L.E.; DiSilvestro, R.A. Zinc in attention-deficit/hyperactivity disorder. J. Child Adolesc. Psychopharmacol. 2005, 15, 619–627. [Google Scholar] [CrossRef]
  123. Cortese, S.; Azoulay, R.; Castellanos, F.X.; Chalard, F.; Lecendreux, M.; Chechin, D.; Delorme, R.; Sebag, G.; Sbarbati, A.; Mouren, M.C.; et al. Brain iron levels in attention-deficit/hyperactivity disorder: A pilot MRI study. World J. Biol. Psychiatry 2012, 13, 223–231. [Google Scholar] [CrossRef]
  124. Huang, Y.H.; Zeng, B.Y.; Li, D.J.; Cheng, Y.S.; Chen, T.Y.; Liang, H.Y.; Yang, W.C.; Lin, P.Y.; Chen, Y.W.; Tseng, P.T.; et al. Significantly lower serum and hair magnesium levels in children with attention deficit hyperactivity disorder than controls: A systematic review and meta-analysis. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 90, 134–141. [Google Scholar] [CrossRef]
  125. Mahmoud, M.M.; El-Mazary, A.A.; Maher, R.M.; Saber, M.M. Zinc, ferritin, magnesium and copper in a group of Egyptian children with attention deficit hyperactivity disorder. Ital. J. Pediatr. 2011, 37, 60. [Google Scholar] [CrossRef] [Green Version]
  126. Scassellati, C.; Bonvicini, C.; Faraone, S.V.; Gennarelli, M. Biomarkers and attention-deficit/hyperactivity disorder: A systematic review and meta-analyses. J. Am. Acad. Child Adolesc. Psychiatry 2012, 51, 1003–1019.e1020. [Google Scholar] [CrossRef] [PubMed]
  127. Garza-Lombo, C.; Posadas, Y.; Quintanar, L.; Gonsebatt, M.E.; Franco, R. Neurotoxicity linked to dysfunctional metal ion homeostasis and xenobiotic metal exposure: Redox signaling and oxidative stress. Antioxid. Redox Signal. 2018. [Google Scholar] [CrossRef] [Green Version]
  128. Abeyawardhane, D.L.; Lucas, H.R. Iron redox chemistry and implications in the Parkinson’s disease brain. Oxid. Med. Cell. Longev. 2019, 2019, 4609702. [Google Scholar] [CrossRef] [Green Version]
  129. Bjørklund, G.; Hofer, T.; Nurchi, V.M.; Aaseth, J. Iron and other metals in the pathogenesis of Parkinson’s disease: Toxic effects and possible detoxification. J. Inorg. Biochem. 2019, 199, 110717. [Google Scholar] [CrossRef]
  130. Bu, X.L.; Xiang, Y.; Guo, Y. The role of iron in amyotrophic Lateral sclerosis. Adv. Exp. Med. Biol. 2019, 1173, 145–152. [Google Scholar] [CrossRef]
  131. Rao, S.S.; Adlard, P.A. Untangling tau and iron: Exploring the interaction between iron and tau in neurodegeneration. Front. Mol. Neurosci. 2018, 11, 276. [Google Scholar] [CrossRef]
  132. Du, L.; Zhao, Z.; Cui, A.; Zhu, Y.; Zhang, L.; Liu, J.; Shi, S.; Fu, C.; Han, X.; Gao, W.; et al. Increased iron deposition on brain quantitative Susceptibility mapping correlates with decreased cognitive function in Alzheimer’s disease. ACS Chem. Neurosci. 2018, 9, 1849–1857. [Google Scholar] [CrossRef]
  133. Bondy, S.C. Low levels of aluminum can lead to behavioral and morphological changes associated with Alzheimer’s disease and age-related neurodegeneration. Neurotoxicology 2016, 52, 222–229. [Google Scholar] [CrossRef] [Green Version]
  134. Kandimalla, R.; Vallamkondu, J.; Corgiat, E.B.; Gill, K.D. Understanding aspects of aluminum exposure in Alzheimer’s disease development. Brain Pathol. 2016, 26, 139–154. [Google Scholar] [CrossRef]
  135. Maya, S.; Prakash, T.; Madhu, K.D.; Goli, D. Multifaceted effects of aluminium in neurodegenerative diseases: A review. Biomed. Pharmacother. 2016, 83, 746–754. [Google Scholar] [CrossRef]
  136. Chen, B.; Wen, X.; Jiang, H.; Wang, J.; Song, N.; Xie, J. Interactions between iron and alpha-synuclein pathology in Parkinson’s disease. Free Radic. Biol. Med. 2019, 141, 253–260. [Google Scholar] [CrossRef]
  137. Peres, T.V.; Schettinger, M.R.; Chen, P.; Carvalho, F.; Avila, D.S.; Bowman, A.B.; Aschner, M. Manganese-induced neurotoxicity: A review of its behavioral consequences and neuroprotective strategies. BMC Pharmacol. Toxicol. 2016, 17, 57. [Google Scholar] [CrossRef] [Green Version]
  138. Binolfi, A.; Rasia, R.M.; Bertoncini, C.W.; Ceolin, M.; Zweckstetter, M.; Griesinger, C.; Jovin, T.M.; Fernandez, C.O. Interaction of alpha-synuclein with divalent metal ions reveals key differences: A link between structure, binding specificity and fibrillation enhancement. J. Am. Chem. Soc. 2006, 128, 9893–9901. [Google Scholar] [CrossRef]
  139. Huang, C.C. Parkinsonism induced by chronic manganese intoxication—An experience in Taiwan. Chang Gung Med. J. 2007, 30, 385–395. [Google Scholar]
  140. Guilarte, T.R.; Burton, N.C.; McGlothan, J.L.; Verina, T.; Zhou, Y.; Alexander, M.; Pham, L.; Griswold, M.; Wong, D.F.; Syversen, T.; et al. Impairment of nigrostriatal dopamine neurotransmission by manganese is mediated by pre-synaptic mechanism(s): Implications to manganese-induced parkinsonism. J. Neurochem. 2008, 107, 1236–1247. [Google Scholar] [CrossRef] [Green Version]
  141. Sheykhansari, S.; Kozielski, K.; Bill, J.; Sitti, M.; Gemmati, D.; Zamboni, P.; Singh, A.V. Redox metals homeostasis in multiple sclerosis and amyotrophic lateral sclerosis: A review. Cell Death Dis. 2018, 9, 348. [Google Scholar] [CrossRef] [PubMed]
  142. Ignjatovic, A.; Stevic, Z.; Lavrnic, S.; Dakovic, M.; Bacic, G. Brain iron MRI: A biomarker for amyotrophic lateral sclerosis. J. Magn. Reson. Imaging 2013, 38, 1472–1479. [Google Scholar] [CrossRef] [PubMed]
  143. Hadzhieva, M.; Kirches, E.; Mawrin, C. Review: Iron metabolism and the role of iron in neurodegenerative disorders. Neuropathol. Appl. Neurobiol. 2014, 40, 240–257. [Google Scholar] [CrossRef]
  144. Peters, T.L.; Beard, J.D.; Umbach, D.M.; Allen, K.; Keller, J.; Mariosa, D.; Sandler, D.P.; Schmidt, S.; Fang, F.; Ye, W.; et al. Blood levels of trace metals and amyotrophic lateral sclerosis. Neurotoxicology 2016, 54, 119–126. [Google Scholar] [CrossRef] [Green Version]
  145. Kamel, F.; Umbach, D.M.; Hu, H.; Munsat, T.L.; Shefner, J.M.; Taylor, J.A.; Sandler, D.P. Lead exposure as a risk factor for amyotrophic lateral sclerosis. Neuro Degener. Dis. 2005, 2, 195–201. [Google Scholar] [CrossRef] [PubMed]
  146. Liao, X.; Li, Y. Genetic associations between voltage-gated calcium channels and autism spectrum disorder: A systematic review. Mol. Brain 2020, 13, 96. [Google Scholar] [CrossRef]
  147. Abraham, J.R.; Szoko, N.; Barnard, J.; Rubin, R.A.; Schlatzer, D.; Lundberg, K.; Li, X.; Natowicz, M.R. Proteomic investigations of autism brain identify known and novel pathogenetic processes. Sci. Rep. 2019, 9, 13118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Vidal-Domènech, F.; Riquelme, G.; Pinacho, R.; Rodriguez-Mias, R.; Vera, A.; Monje, A.; Ferrer, I.; Callado, L.F.; Meana, J.J.; Villén, J.; et al. Calcium-binding proteins are altered in the cerebellum in schizophrenia. PLoS ONE 2020, 15, e0230400. [Google Scholar] [CrossRef]
  149. Dey, K.; Bazala, M.A.; Kuznicki, J. Targeting mitochondrial calcium pathways as a potential treatment against Parkinson’s disease. Cell Calcium 2020, 89, 102216. [Google Scholar] [CrossRef]
  150. Alzheimer’s Association Calcium Hypothesis, W.; Khachaturian, Z.S. Calcium hypothesis of Alzheimer’s disease and brain aging: A framework for integrating new evidence into a comprehensive theory of pathogenesis. Alzheimer’s Dement. 2017, 13, 178–182.e117. [Google Scholar] [CrossRef] [PubMed]
  151. Grosskreutz, J.; Van Den Bosch, L.; Keller, B.U. Calcium dysregulation in amyotrophic lateral sclerosis. Cell Calcium 2010, 47, 165–174. [Google Scholar] [CrossRef] [PubMed]
  152. Xu, X.; Wang, Q.; Zhang, M. Age, gender, and hemispheric differences in iron deposition in the human brain: An in vivo MRI study. NeuroImage 2008, 40, 35–42. [Google Scholar] [CrossRef] [PubMed]
  153. Hallgren, B.; Sourander, P. THE effect of age on the non-haemin iron in the human brain. J. Neurochem. 1958, 3, 41–51. [Google Scholar] [CrossRef]
  154. Loeffler, D.A.; Connor, J.R.; Juneau, P.L.; Snyder, B.S.; Kanaley, L.; DeMaggio, A.J.; Nguyen, H.; Brickman, C.M.; LeWitt, P.A. Transferrin and iron in normal, Alzheimer’s disease, and Parkinson’s disease brain regions. J. Neurochem. 1995, 65, 710–716. [Google Scholar] [CrossRef]
  155. Bartzokis, G.; Mintz, J.; Sultzer, D.; Marx, P.; Herzberg, J.S.; Phelan, C.K.; Marder, S.R. In vivo MR evaluation of age-related increases in brain iron. Am. J. Neuroradiol. 1994, 15, 1129–1138. [Google Scholar]
  156. Martin, W.R.W.; Ye, F.Q.; Allen, P.S. Increasing striatal iron content associated with normal aging. Mov. Disord. 1998, 13, 281–286. [Google Scholar] [CrossRef] [PubMed]
  157. Zecca, L.; Zucca, F.A.; Toscani, M.; Adorni, F.; Giaveri, G.; Rizzio, E.; Gallorini, M. Iron, copper and their proteins in substantia nigra of human brain during aging. J. Radioanal. Nucl. Chem. 2005, 263, 733–737. [Google Scholar] [CrossRef]
  158. Ramos, P.; Santos, A.; Pinto, N.R.; Mendes, R.; Magalhães, T.; Almeida, A. Anatomical region differences and age-related changes in copper, zinc, and manganese levels in the human brain. Biol. Trace Elem. Res. 2014, 161, 190–201. [Google Scholar] [CrossRef]
  159. Vasudevaraju, P.; Bharathi, G.; Jyothsna, T.; Shamasundar, N.M.; Subba Rao, K.; Balaraj, B.M.; Rao, K.S.J.; Sathyanarayana Rao, T.S. New evidence on iron, copper accumulation and zinc depletion and its correlation with DNA integrity in aging human brain regions. Indian J. Psychiatry 2010, 52, 140–144. [Google Scholar] [CrossRef]
  160. Bolognin, S.; Messori, L.; Zatta, P. Metal ion physiopathology in neurodegenerative disorders. Neuromol. Med. 2009, 11, 223–238. [Google Scholar] [CrossRef]
  161. Jellinger, K.A. Chapter one—The relevance of metals in the pathophysiology of neurodegeneration, pathological considerations. In International Review of Neurobiology; Bhatia, K.P., Schneider, S.A., Eds.; Academic Press: Cambridge, MA, USA, 2013; Volume 110, pp. 1–47. [Google Scholar]
  162. Jomova, K.; Valko, M. Advances in metal-induced oxidative stress and human disease. Toxicology 2011, 283, 65–87. [Google Scholar] [CrossRef]
  163. Gambling, L.; Lang, C.; McArdle, H.J. Fetal regulation of iron transport during pregnancy. Am. J. Clin. Nutr. 2011, 94, 1903S–1907S. [Google Scholar] [CrossRef] [Green Version]
  164. Cao, C.; Fleming, M.D. The placenta: The forgotten essential organ of iron transport. Nutr. Rev. 2016, 74, 421–431. [Google Scholar] [CrossRef] [Green Version]
  165. Moos, T. Developmental profile of non-heme iron distribution in the rat brain during ontogenesis. Dev. Brain Res. 1995, 87, 203–213. [Google Scholar] [CrossRef]
  166. Dawson, P.A.; Elliott, A.; Bowling, F.G. Sulphate in pregnancy. Nutrients 2015, 7, 1594–1606. [Google Scholar] [CrossRef] [Green Version]
  167. Semmler-Behnke, M.; Lipka, J.; Wenk, A.; Hirn, S.; Schaffler, M.; Tian, F.; Schmid, G.; Oberdorster, G.; Kreyling, W.G. Size dependent translocation and fetal accumulation of gold nanoparticles from maternal blood in the rat. Part Fibre Toxicol. 2014, 11, 33. [Google Scholar] [CrossRef]
  168. Pietroiusti, A.; Campagnolo, L.; Fadeel, B. Interactions of engineered nanoparticles with organs protected by internal biological barriers. Small 2013, 9, 1557–1572. [Google Scholar] [CrossRef] [Green Version]
  169. McArdle, H.J.; Andersen, H.S.; Jones, H.; Gambling, L. Copper and iron transport across the placenta: Regulation and interactions. J. Neuroendocrinol. 2008, 20, 427–431. [Google Scholar] [CrossRef]
  170. Edel, J.; Sabbioni, E. Vanadium transport across placenta and milk of rats to the fetus and newborn. Biol. Trace Elem. Res. 1989, 22, 265–275. [Google Scholar] [CrossRef]
  171. Goyer, R.A. Transplacental transport of lead. Environ. Health Perspect. 1990, 89, 101–105. [Google Scholar] [CrossRef]
  172. Gundacker, C.; Hengstschlager, M. The role of the placenta in fetal exposure to heavy metals. Wien Med. Wochenschr. 2012, 162, 201–206. [Google Scholar] [CrossRef]
  173. Iwai-Shimada, M.; Kameo, S.; Nakai, K.; Yaginuma-Sakurai, K.; Tatsuta, N.; Kurokawa, N.; Nakayama, S.F.; Satoh, H. Exposure profile of mercury, lead, cadmium, arsenic, antimony, copper, selenium and zinc in maternal blood, cord blood and placenta: The Tohoku Study of Child Development in Japan. Environ. Health Prev. Med. 2019, 24, 35. [Google Scholar] [CrossRef] [Green Version]
  174. Li, A.; Zhuang, T.; Shi, J.; Liang, Y.; Song, M. Heavy metals in maternal and cord blood in Beijing and their efficiency of placental transfer. J. Environ. Sci. (China) 2019, 80, 99–106. [Google Scholar] [CrossRef]
  175. Chen, Z.; Myers, R.; Wei, T.; Bind, E.; Kassim, P.; Wang, G.; Ji, Y.; Hong, X.; Caruso, D.; Bartell, T.; et al. Placental transfer and concentrations of cadmium, mercury, lead, and selenium in mothers, newborns, and young children. J. Expo. Sci. Environ. Epidemiol. 2014, 24, 537–544. [Google Scholar] [CrossRef] [Green Version]
  176. Sakamoto, M.; Yasutake, A.; Domingo, J.L.; Chan, H.M.; Kubota, M.; Murata, K. Relationships between trace element concentrations in chorionic tissue of placenta and umbilical cord tissue: Potential use as indicators for prenatal exposure. Environ. Int. 2013, 60, 106–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Moos, T.; Morgan, E.H. A morphological study of the developmentally regulated transport of iron into the brain. Dev. Neurosci. 2002, 24, 99–105. [Google Scholar] [CrossRef]
  178. Siddappa, A.J.; Rao, R.B.; Wobken, J.D.; Leibold, E.A.; Connor, J.R.; Georgieff, M.K. Developmental changes in the expression of iron regulatory proteins and iron transport proteins in the perinatal rat brain. J. Neurosci. Res. 2002, 68, 761–775. [Google Scholar] [CrossRef]
  179. Lonnerdal, B.; Georgieff, M.K.; Hernell, O. Developmental physiology of iron absorption, homeostasis, and metabolism in the healthy term infant. J. Pediatr. 2015, 167, S8–S14. [Google Scholar] [CrossRef] [Green Version]
  180. Hoet, P.H.; Bruske-Hohlfeld, I.; Salata, O.V. Nanoparticles—Known and unknown health risks. J. Nanobiotechnol. 2004, 2, 12. [Google Scholar] [CrossRef] [Green Version]
  181. Geiser, M.; Rothen-Rutishauser, B.; Kapp, N.; Schurch, S.; Kreyling, W.; Schulz, H.; Semmler, M.; Im Hof, V.; Heyder, J.; Gehr, P. Ultrafine particles cross cellular membranes by nonphagocytic mechanisms in lungs and in cultured cells. Environ. Health Perspect. 2005, 113, 1555–1560. [Google Scholar] [CrossRef] [Green Version]
  182. Lin, Y.; Hu, C.; Chen, A.; Feng, X.; Liang, H.; Yin, S.; Zhang, G.; Shao, L. Neurotoxicity of nanoparticles entering the brain via sensory nerve-to-brain pathways: Injuries and mechanisms. Arch. Toxicol. 2020, 94, 1479–1495. [Google Scholar] [CrossRef] [Green Version]
  183. Tjalve, H.; Henriksson, J. Uptake of metals in the brain via olfactory pathways. Neurotoxicology 1999, 20, 181–195. [Google Scholar]
  184. Wang, B.; Feng, W.Y.; Wang, M.; Shi, J.W.; Zhang, F.; Ouyang, H.; Zhao, Y.L.; Chai, Z.F.; Huang, Y.Y.; Xie, Y.N.; et al. Transport of intranasally instilled fine Fe2O3 particles into the brain: Micro-distribution, chemical states, and histopathological observation. Biol. Trace Elem. Res. 2007, 118, 233–243. [Google Scholar] [CrossRef]
  185. Petters, C.; Irrsack, E.; Koch, M.; Dringen, R. Uptake and metabolism of iron oxide nanoparticles in brain cells. Neurochem. Res. 2014, 39, 1648–1660. [Google Scholar] [CrossRef]
  186. Persson, E.; Henriksson, J.; Tjalve, H. Uptake of cobalt from the nasal mucosa into the brain via olfactory pathways in rats. Toxicol. Lett. 2003, 145, 19–27. [Google Scholar] [CrossRef]
  187. Sunderman, F.W., Jr. Nasal toxicity, carcinogenicity, and olfactory uptake of metals. Ann. Clin. Lab. Sci. 2001, 31, 3–24. [Google Scholar]
  188. Perl, D.; Good, P. Uptake of aluminium into central nervous system along nasal-olfactory pathways. Lancet 1987, 329, 1028. [Google Scholar] [CrossRef]
  189. Ibanez, C.; Suhard, D.; Tessier, C.; Delissen, O.; Lestaevel, P.; Dublineau, I.; Gourmelon, P. Intranasal exposure to uranium results in direct transfer to the brain along olfactory nerve bundles. Neuropathol. Appl. Neurobiol. 2014, 40, 477–488. [Google Scholar] [CrossRef]
  190. Lucchini, R.G.; Dorman, D.C.; Elder, A.; Veronesi, B. Neurological impacts from inhalation of pollutants and the nose-brain connection. Neurotoxicology 2012, 33, 838–841. [Google Scholar] [CrossRef] [Green Version]
  191. Oberdörster, G.; Sharp, Z.; Atudorei, V.; Elder, A.; Gelein, R.; Kreyling, W.; Cox, C. Translocation of inhaled ultrafine particles to the brain. Inhal. Toxicol. 2004, 16, 437–445. [Google Scholar] [CrossRef]
  192. Askri, D.; Ouni, S.; Galai, S.; Arnaud, J.; Chovelon, B.; Lehmann, S.G.; Sturm, N.; Sakly, M.; Seve, M.; Amara, S. Intranasal instillation of iron oxide nanoparticles induces inflammation and perturbation of trace elements and neurotransmitters, but not behavioral impairment in rats. Environ. Sci. Pollut. Res. Int. 2018. [Google Scholar] [CrossRef]
  193. Costa-Mallen, P.; Gatenby, C.; Friend, S.; Maravilla, K.R.; Hu, S.-C.; Cain, K.C.; Agarwal, P.; Anzai, Y. Brain iron concentrations in regions of interest and relation with serum iron levels in Parkinson disease. J. Neurol. Sci. 2017, 378, 38–44. [Google Scholar] [CrossRef] [PubMed]
  194. Jin, L.; Wang, J.; Zhao, L.; Jin, H.; Fei, G.; Zhang, Y.; Zeng, M.; Zhong, C. Decreased serum ceruloplasmin levels characteristically aggravate nigral iron deposition in Parkinson’s disease. Brain 2011, 134, 50–58. [Google Scholar] [CrossRef] [Green Version]
  195. Logroscino, G.; Marder, K.; Graziano, J.; Freyer, G.; Slavkovich, V.; LoIacono, N.; Cote, L.; Mayeux, R. Altered systemic iron metabolism in Parkinson’s disease. Neurology 1997, 49, 714–717. [Google Scholar] [CrossRef] [PubMed]
  196. Hunter, D.D.; Dey, R.D. Identification and neuropeptide content of trigeminal neurons innervating the rat nasal epithelium. Neuroscience 1998, 83, 591–599. [Google Scholar] [CrossRef]
  197. Oberdörster, G.; Oberdörster, E.; Oberdörster, J. Nanotoxicology: An emerging discipline evolving from studies of ultrafine particles. Environ. Health Perspect. 2005, 113, 823–839. [Google Scholar] [CrossRef]
  198. Hawkes, C.H.; Del Tredici, K.; Braak, H. Parkinson’s disease: A dual-hit hypothesis. Neuropathol. Appl. Neurobiol. 2007, 33, 599–614. [Google Scholar] [CrossRef] [PubMed]
  199. Chounlamountry, K.; Boyer, B.; Penalba, V.; François-Bellan, A.M.; Bosler, O.; Kessler, J.P.; Strube, C. Remodeling of glial coverage of glutamatergic synapses in the rat nucleus tractus solitarii after ozone inhalation. J. Neurochem. 2015, 134, 857–864. [Google Scholar] [CrossRef] [Green Version]
  200. Robinson, R.K.; Birrell, M.A.; Adcock, J.J.; Wortley, M.A.; Dubuis, E.D.; Chen, S.; McGilvery, C.M.; Hu, S.; Shaffer, M.S.P.; Bonvini, S.J.; et al. Mechanistic link between diesel exhaust particles and respiratory reflexes. J. Allergy Clin. Immunol. 2018, 141, 1074–1084.e1079. [Google Scholar] [CrossRef] [Green Version]
  201. Villarreal-Calderon, R.; Torres-Jardón, R.; Palacios-Moreno, J.; Osnaya, N.; Pérez-Guillé, B.; Maronpot, R.R.; Reed, W.; Zhu, H.; Calderón-Garcidueñas, L. Urban air pollution targets the dorsal vagal complex and dark chocolate offers neuroprotection. Int. J. Toxicol. 2010, 29, 604–615. [Google Scholar] [CrossRef] [PubMed]
  202. Baruah, J.K.; Rasool, C.G.; Bradley, W.G.; Munsat, T.L. Retrograde axonal transport of lead in rat sciatic nerve. Neurology 1981, 31, 612. [Google Scholar] [CrossRef] [PubMed]
  203. Morgan, J.; Bell, R.; Jones, A.L. Endogenous doesn’t always mean innocuous: A scoping review of iron toxicity by inhalation. J. Toxicol. Environ. Health B 2020, 23, 107–136. [Google Scholar] [CrossRef]
  204. Verkhlyutov, V.M.; Gapienko, G.V.; Ushakov, V.L.; Portnova, G.V.; Verkhlyutova, I.A.; Anisimov, N.V.; Pirogov, Y.A. MRI morphometry of the cerebral ventricles in patients with attention deficit hyperactivity disorder. Neurosci. Behav. Physiol. 2010, 40, 295–303. [Google Scholar] [CrossRef]
  205. Wang, J.; Jiang, T.; Cao, Q.; Wang, Y. Characterizing anatomic differences in boys with attention-deficit/hyperactivity disorder with the use of deformation-based morphometry. AJNR Am. J. Neuroradiol. 2007, 28, 543–547. [Google Scholar]
  206. Cheung, C.; Yu, K.; Fung, G.; Leung, M.; Wong, C.; Li, Q.; Sham, P.; Chua, S.; McAlonan, G. Autistic disorders and schizophrenia: Related or remote? An anatomical likelihood estimation. PLoS ONE 2010, 5, e12233. [Google Scholar] [CrossRef] [Green Version]
  207. Cuesta, M.J.; Lecumberri, P.; Cabada, T.; Moreno-Izco, L.; Ribeiro, M.; Lopez-Ilundain, J.M.; Peralta, V.; Lorente-Omenaca, R.; Sanchez-Torres, A.M.; Gomez, M. Basal ganglia and ventricle volume in first-episode psychosis. A family and clinical study. Psychiatry Res. Neuroimaging 2017, 269, 90–96. [Google Scholar] [CrossRef] [PubMed]
  208. Del Re, E.C.; Konishi, J.; Bouix, S.; Blokland, G.A.; Mesholam-Gately, R.I.; Goldstein, J.; Kubicki, M.; Wojcik, J.; Pasternak, O.; Seidman, L.J.; et al. Enlarged lateral ventricles inversely correlate with reduced corpus callosum central volume in first episode schizophrenia: Association with functional measures. Brain Imaging Behav. 2016, 10, 1264–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Milovanovic, N.; Damjanovic, A.; Puskas, L.; Milovanovic, S.; Barisic, J.; Malis, M.; Stankovic, G.; Rankovic, A.; Latas, M.; Filipovic, B.F.; et al. Reliability of the bicaudate parameter in the revealing of the enlarged lateral Ventricles in schizophrenia patients. Psychiatr. Danub. 2018, 30, 150–156. [Google Scholar] [CrossRef] [PubMed]
  210. Okada, N.; Fukunaga, M.; Yamashita, F.; Koshiyama, D.; Yamamori, H.; Ohi, K.; Yasuda, Y.; Fujimoto, M.; Watanabe, Y.; Yahata, N.; et al. Abnormal asymmetries in subcortical brain volume in schizophrenia. Mol. Psychiatry 2016, 21, 1460–1466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. van Erp, T.G.; Greve, D.N.; Rasmussen, J.; Turner, J.; Calhoun, V.D.; Young, S.; Mueller, B.; Brown, G.G.; McCarthy, G.; Glover, G.H.; et al. A multi-scanner study of subcortical brain volume abnormalities in schizophrenia. Psychiatry Res. 2014, 222, 10–16. [Google Scholar] [CrossRef] [Green Version]
  212. van Erp, T.G.; Hibar, D.P.; Rasmussen, J.M.; Glahn, D.C.; Pearlson, G.D.; Andreassen, O.A.; Agartz, I.; Westlye, L.T.; Haukvik, U.K.; Dale, A.M.; et al. Subcortical brain volume abnormalities in 2028 individuals with schizophrenia and 2540 healthy controls via the ENIGMA consortium. Mol. Psychiatry 2016, 21, 585. [Google Scholar] [CrossRef]
  213. Lange, N.; Travers, B.G.; Bigler, E.D.; Prigge, M.B.; Froehlich, A.L.; Nielsen, J.A.; Cariello, A.N.; Zielinski, B.A.; Anderson, J.S.; Fletcher, P.T.; et al. Longitudinal volumetric brain changes in autism spectrum disorder ages 6-35 years. Autism Res. 2015, 8, 82–93. [Google Scholar] [CrossRef]
  214. Aoki, Y.; Yoncheva, Y.N.; Chen, B.; Nath, T.; Sharp, D.; Lazar, M.; Velasco, P.; Milham, M.P.; Di Martino, A. Association of white matter structure with autism spectrum disorder and attention-deficit/hyperactivity disorder. JAMA Psychiatry 2017. [Google Scholar] [CrossRef]
  215. Di, X.; Azeez, A.; Li, X.; Haque, E.; Biswal, B.B. Disrupted focal white matter integrity in autism spectrum disorder: A voxel-based meta-analysis of diffusion tensor imaging studies. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 82, 242–248. [Google Scholar] [CrossRef] [PubMed]
  216. Fingher, N.; Dinstein, I.; Ben-Shachar, M.; Haar, S.; Dale, A.M.; Eyler, L.; Pierce, K.; Courchesne, E. Toddlers later diagnosed with autism exhibit multiple structural abnormalities in temporal corpus callosum fibers. Cortex 2017, 97, 291–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Karahanoglu, F.I.; Baran, B.; Nguyen, Q.T.H.; Meskaldji, D.E.; Yendiki, A.; Vangel, M.; Santangelo, S.L.; Manoach, D.S. Diffusion-weighted imaging evidence of altered white matter development from late childhood to early adulthood in autism spectrum disorder. Neuroimage Clin. 2018, 19, 840–847. [Google Scholar] [CrossRef] [PubMed]
  218. Albaugh, M.D.; Hudziak, J.J.; Ing, A.; Chaarani, B.; Barker, E.; Jia, T.; Lemaitre, H.; Watts, R.; Orr, C.; Spechler, P.A.; et al. White matter microstructure is associated with hyperactive/inattentive symptomatology and polygenic risk for attention-deficit/hyperactivity disorder in a population-based sample of adolescents. Neuropsychopharmacology 2019, 44, 1597–1603. [Google Scholar] [CrossRef]
  219. Cropley, V.L.; Klauser, P.; Lenroot, R.K.; Bruggemann, J.; Sundram, S.; Bousman, C.; Pereira, A.; Di Biase, M.A.; Weickert, T.W.; Weickert, C.S.; et al. Accelerated gray and white matter deterioration with age in schizophrenia. Am. J. Psychiatry 2017, 174, 286–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Levitt, J.J.; Nestor, P.G.; Levin, L.; Pelavin, P.; Lin, P.; Kubicki, M.; McCarley, R.W.; Shenton, M.E.; Rathi, Y. Reduced structural connectivity in frontostriatal white matter tracts in the associative loop in schizophrenia. Am. J. Psychiatry 2017, 174, 1102–1111. [Google Scholar] [CrossRef]
  221. Price, G.; Cercignani, M.; Parker, G.J.; Altmann, D.R.; Barnes, T.R.; Barker, G.J.; Joyce, E.M.; Ron, M.A. Abnormal brain connectivity in first-episode psychosis: A diffusion MRI tractography study of the corpus callosum. Neuroimage 2007, 35, 458–466. [Google Scholar] [CrossRef]
  222. Seal, M.L.; Yucel, M.; Fornito, A.; Wood, S.J.; Harrison, B.J.; Walterfang, M.; Pell, G.S.; Pantelis, C. Abnormal white matter microstructure in schizophrenia: A voxelwise analysis of axial and radial diffusivity. Schizophr. Res. 2008, 101, 106–110. [Google Scholar] [CrossRef]
  223. Siffredi, V.; Anderson, V.; McIlroy, A.; Wood, A.G.; Leventer, R.J.; Spencer-Smith, M.M. A neuropsychological profile for agenesis of the corpus callosum? Cognitive, academic, executive, social, and behavioral functioning in school-age children. J. Int. Neuropsychol. Soc. 2018, 24, 445–455. [Google Scholar] [CrossRef] [Green Version]
  224. Anderson, L.B.; Paul, L.K.; Brown, W.S. Emotional intelligence in agenesis of the corpus callosum. Arch. Clin. Neuropsychol. 2017, 32, 267–279. [Google Scholar] [CrossRef] [PubMed]
  225. Blackmon, K.; Ben-Avi, E.; Wang, X.; Pardoe, H.R.; Di Martino, A.; Halgren, E.; Devinsky, O.; Thesen, T.; Kuzniecky, R. Periventricular white matter abnormalities and restricted repetitive behavior in autism spectrum disorder. NeuroImage Clin. 2016, 10, 36–45. [Google Scholar] [CrossRef] [Green Version]
  226. Chen, B. Abnormal regions in functional connectivity of chronic schizophrenia. Int. J. Neurosci. 2018, 128, 1150–1156. [Google Scholar] [CrossRef]
  227. Frith, C. Is autism a disconnection disorder? Lancet Neurol. 2004, 3, 577. [Google Scholar] [CrossRef]
  228. Geschwind, D.H.; Levitt, P. Autism spectrum disorders: Developmental disconnection syndromes. Curr. Opin. Neurobiol. 2007, 17, 103–111. [Google Scholar] [CrossRef]
  229. Cai, W.; Chen, T.; Szegletes, L.; Supekar, K.; Menon, V. Aberrant time-varying cross-network interactions in children with attention-deficit/hyperactivity disorder and the relation to attention deficits. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2018, 3, 263–273. [Google Scholar] [CrossRef]
  230. Griffa, A.; Baumann, P.S.; Klauser, P.; Mullier, E.; Cleusix, M.; Jenni, R.; van den Heuvel, M.P.; Do, K.Q.; Conus, P.; Hagmann, P. Brain connectivity alterations in early psychosis: From clinical to neuroimaging staging. Transl. Psychiatry 2019, 9, 62. [Google Scholar] [CrossRef] [Green Version]
  231. Joo, S.W.; Yoon, W.; Shon, S.H.; Kim, H.; Cha, S.; Park, K.J.; Lee, J. Altered white matter connectivity in patients with schizophrenia: An investigation using public neuroimaging data from SchizConnect. PLoS ONE 2018, 13, e0205369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Zhang, W.; Li, S.; Wang, X.; Gong, Y.; Yao, L.; Xiao, Y.; Liu, J.; Keedy, S.K.; Gong, Q.; Sweeney, J.A.; et al. Abnormal dynamic functional connectivity between speech and auditory areas in schizophrenia patients with auditory hallucinations. Neuroimage Clin. 2018, 19, 918–924. [Google Scholar] [CrossRef] [PubMed]
  233. Zhu, F.; Liu, F.; Guo, W.; Chen, J.; Su, Q.; Zhang, Z.; Li, H.; Fan, X.; Zhao, J. Disrupted asymmetry of inter- and intra-hemispheric functional connectivity in patients with drug-naive, first-episode schizophrenia and their unaffected siblings. EBioMedicine 2018, 36, 429–435. [Google Scholar] [CrossRef] [Green Version]
  234. Cochran, D.M.; Sikoglu, E.M.; Hodge, S.M.; Edden, R.A.; Foley, A.; Kennedy, D.N.; Moore, C.M.; Frazier, J.A. Relationship among glutamine, gamma-aminobutyric acid, and social cognition in autism spectrum disorders. J. Child Adolesc. Psychopharmacol. 2015, 25, 314–322. [Google Scholar] [CrossRef] [Green Version]
  235. Drenthen, G.S.; Barendse, E.M.; Aldenkamp, A.P.; van Veenendaal, T.M.; Puts, N.A.; Edden, R.A.; Zinger, S.; Thoonen, G.; Hendriks, M.P.; Kessels, R.P.; et al. Altered neurotransmitter metabolism in adolescents with high-functioning autism. Psychiatry Res. 2016, 256, 44–49. [Google Scholar] [CrossRef] [Green Version]
  236. Ford, T.C.; Nibbs, R.; Crewther, D.P. Glutamate/GABA+ ratio is associated with the psychosocial domain of autistic and schizotypal traits. PLoS ONE 2017, 12, e0181961. [Google Scholar] [CrossRef] [Green Version]
  237. Rosch, K.S.; Mostofsky, S.H.; Nebel, M.B. ADHD-related sex differences in fronto-subcortical intrinsic functional connectivity and associations with delay discounting. J. Neurodev. Disord. 2018, 10, 34. [Google Scholar] [CrossRef]
  238. Zepf, F.D.; Bubenzer-Busch, S.; Runions, K.C.; Rao, P.; Wong, J.W.Y.; Mahfouda, S.; Morandini, H.A.E.; Stewart, R.M.; Moore, J.K.; Biskup, C.S.; et al. Functional connectivity of the vigilant-attention network in children and adolescents with attention-deficit/hyperactivity disorder. Brain Cogn. 2019, 131, 56–65. [Google Scholar] [CrossRef]
  239. Al-Otaish, H.; Al-Ayadhi, L.; Bjorklund, G.; Chirumbolo, S.; Urbina, M.A.; El-Ansary, A. Relationship between absolute and relative ratios of glutamate, glutamine and GABA and severity of autism spectrum disorder. Metab. Brain Dis. 2018. [Google Scholar] [CrossRef]
  240. Avissar, M.; Javitt, D. Mismatch negativity: A simple and useful biomarker of N-methyl-d-aspartate receptor (NMDAR)-type glutamate dysfunction in schizophrenia. Schizophr. Res. 2018, 191, 1–4. [Google Scholar] [CrossRef]
  241. Bauer, J.; Werner, A.; Kohl, W.; Kugel, H.; Shushakova, A.; Pedersen, A.; Ohrmann, P. Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. World J. Biol. Psychiatry 2018, 19, 538–546. [Google Scholar] [CrossRef]
  242. Chang, J.P.; Lane, H.Y.; Tsai, G.E. Attention deficit hyperactivity disorder and N-methyl-D-aspartate (NMDA) dysregulation. Curr. Pharm. Des. 2014, 20, 5180–5185. [Google Scholar] [CrossRef]
  243. Duarte, J.M.N.; Xin, L. Magnetic resonance spectroscopy in schizophrenia: Evidence for glutamatergic dysfunction and impaired energy metabolism. Neurochem. Res. 2019, 44, 102–116. [Google Scholar] [CrossRef] [Green Version]
  244. Hegarty, J.P., 2nd; Weber, D.J.; Cirstea, C.M.; Beversdorf, D.Q. Cerebro-cerebellar functional connectivity is associated with cerebellar excitation-inhibition balance in autism spectrum disorder. J. Autism Dev. Disord. 2018, 48, 3460–3473. [Google Scholar] [CrossRef]
  245. Hoftman, G.D.; Dienel, S.J.; Bazmi, H.H.; Zhang, Y.; Chen, K.; Lewis, D.A. Altered gradients of glutamate and gamma-aminobutyric acid transcripts in the cortical visuospatial working Memory network in schizophrenia. Biol. Psychiatry 2017. [Google Scholar] [CrossRef]
  246. Jelen, L.A.; King, S.; Horne, C.M.; Lythgoe, D.J.; Young, A.H.; Stone, J.M. Functional magnetic resonance spectroscopy in patients with schizophrenia and bipolar affective disorder: Glutamate dynamics in the anterior cingulate cortex during a working memory task. Eur. Neuropsychopharmacol. 2019, 29, 222–234. [Google Scholar] [CrossRef] [Green Version]
  247. Uno, Y.; Coyle, J.T. Glutamate hypothesis in schizophrenia. Psychiatry Clin. Neurosci. 2019, 73, 204–215. [Google Scholar] [CrossRef] [Green Version]
  248. Batool, S.; Raza, H.; Zaidi, J.; Riaz, S.; Hasan, S.; Syed, N.I. Synapse formation: From cellular and molecular mechanisms to neurodevelopmental and neurodegenerative disorders. J. Neurophysiol. 2019, 121, 1381–1397. [Google Scholar] [CrossRef]
  249. Bennett, M.R.; Lagopoulos, J. Neurodevelopmental sequelae associated with gray and white matter changes and their cellular basis: A comparison between autism spectrum disorder, ADHD and dyslexia. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 2015, 46, 132–143. [Google Scholar] [CrossRef]
  250. Bunney, B.G.; Potkin, S.G.; Bunney, W.E., Jr. New morphological and neuropathological findings in schizophrenia: A neurodevelopmental perspective. Clin. Neurosci. 1995, 3, 81–88. [Google Scholar]
  251. Connor, C.M.; Crawford, B.C.; Akbarian, S. White matter neuron alterations in schizophrenia and related disorders. Int. J. Dev. Neurosci. 2011, 29, 325–334. [Google Scholar] [CrossRef] [Green Version]
  252. Gilmore, J.H.; Jarskog, L.F.; Vadlamudi, S. Maternal poly I:C exposure during pregnancy regulates TNF alpha, BDNF, and NGF expression in neonatal brain and the maternal-fetal unit of the rat. J. Neuroimmunol. 2005, 159, 106–112. [Google Scholar] [CrossRef]
  253. Glantz, L.A.; Gilmore, J.H.; Lieberman, J.A.; Jarskog, L.F. Apoptotic mechanisms and the synaptic pathology of schizophrenia. Schizophr. Res. 2006, 81, 47–63. [Google Scholar] [CrossRef]
  254. Moretto, E.; Murru, L.; Martano, G.; Sassone, J.; Passafaro, M. Glutamatergic synapses in neurodevelopmental disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 84, 328–342. [Google Scholar] [CrossRef]
  255. Rankovic, M.; Zweckstetter, M. Upregulated levels and pathological aggregation of abnormally phosphorylated Tau-protein in children with neurodevelopmental disorders. Neurosci. Biobehav. Rev. 2019, 98, 1–9. [Google Scholar] [CrossRef]
  256. Vilalta, A.; Brown, G.C. Neurophagy, the phagocytosis of live neurons and synapses by glia, contributes to brain development and disease. FEBS J. 2018, 285, 3566–3575. [Google Scholar] [CrossRef] [Green Version]
  257. Wei, H.; Alberts, I.; Li, X. The apoptotic perspective of autism. Int. J. Dev. Neurosci. 2014, 36, 13–18. [Google Scholar] [CrossRef]
  258. King, B.H.; Lord, C. Is schizophrenia on the autism spectrum? Brain Res. 2011, 1380, 34–41. [Google Scholar] [CrossRef]
  259. Meyer, U.; Feldon, J.; Dammann, O. Schizophrenia and autism: Both shared and disorder-specific pathogenesis via perinatal inflammation? Pediatric Res. 2011, 69, 26R–33R. [Google Scholar] [CrossRef] [Green Version]
  260. Mitchell, R.H.; Goldstein, B.I. Inflammation in children and adolescents with neuropsychiatric disorders: A systematic review. J. Am. Acad. Child Adolesc. Psychiatry 2014, 53, 274–296. [Google Scholar] [CrossRef]
  261. Prata, J.; Santos, S.G.; Almeida, M.I.; Coelho, R.; Barbosa, M.A. Bridging autism spectrum disorders and schizophrenia through inflammation and biomarkers—Pre-clinical and clinical investigations. J. Neuroinflammation 2017, 14, 179. [Google Scholar] [CrossRef] [Green Version]
  262. Rogers, J.C.; De Brito, S.A. Cortical and subcortical gray matter volume in youths with conduct problems: A meta-analysis. JAMA Psychiatry 2016, 73, 64–72. [Google Scholar] [CrossRef]
  263. Smaga, I.; Niedzielska, E.; Gawlik, M.; Moniczewski, A.; Krzek, J.; Przegalinski, E.; Pera, J.; Filip, M. Oxidative stress as an etiological factor and a potential treatment target of psychiatric disorders. Part 2. Depression, anxiety, schizophrenia and autism. Pharmacol. Rep. 2015, 67, 569–580. [Google Scholar] [CrossRef]
  264. Takahashi, Y.; Yu, Z.; Sakai, M.; Tomita, H. Linking activation of microglia and peripheral monocytic cells to the pathophysiology of psychiatric disorders. Front. Cell. Neurosci. 2016, 10, 144. [Google Scholar] [CrossRef]
  265. Krzysztofik, K.; Otrębski, W. Measurement tools of autism syndrome severity and selected neurocognitive processes in individuals with ASD. Psychiatr. Pol. 2018, 52, 641–650. [Google Scholar] [CrossRef]
  266. Leffa, D.T.; Torres, I.L.S.; Rohde, L.A. A Review on the role of inflammation in attention-deficit/hyperactivity disorder. Neuroimmunomodulation 2018, 25, 328–333. [Google Scholar] [CrossRef]
  267. Maas, D.A.; Vallès, A.; Martens, G.J.M. Oxidative stress, prefrontal cortex hypomyelination and cognitive symptoms in schizophrenia. Transl. Psychiatry 2017, 7, e1171. [Google Scholar] [CrossRef] [Green Version]
  268. Nakagawa, Y.; Chiba, K. Involvement of neuroinflammation during brain development in social cognitive deficits in autism spectrum disorder and schizophrenia. J. Pharmacol. Exp. Ther. 2016, 358, 504–515. [Google Scholar] [CrossRef] [Green Version]
  269. Rommelse, N.N.; Geurts, H.M.; Franke, B.; Buitelaar, J.K.; Hartman, C.A. A review on cognitive and brain endophenotypes that may be common in autism spectrum disorder and attention-deficit/hyperactivity disorder and facilitate the search for pleiotropic genes. Neurosci. Biobehav. Rev. 2011, 35, 1363–1396. [Google Scholar] [CrossRef]
  270. Upthegrove, R.; Khandaker, G.M. Cytokines, oxidative stress and cellular markers of inflammation in schizophrenia. Curr. Top. Behav. Neurosci. 2020, 44, 49–66. [Google Scholar] [CrossRef]
  271. Waligora, A.; Waligora, S.; Kozarska, M.; Damasiewicz-Bodzek, A.; Gorczyca, P.; Tyrpien-Golder, K. Autism spectrum disorder (ASD)—Biomarkers of oxidative stress and methylation and transsulfuration cycle. Psychiatr. Pol. 2019, 53, 771–788. [Google Scholar] [CrossRef]
  272. Yui, K.; Kawasaki, Y.; Yamada, H.; Ogawa, S. Oxidative stress and nitric oxide in autism spectrum disorder and other neuropsychiatric disorders. CNS Neurol. Disord. Drug Targets 2016, 15, 587–596. [Google Scholar] [CrossRef]
  273. Andrews, G.; Pine, D.S.; Hobbs, M.J.; Anderson, T.M.; Sunderland, M. Neurodevelopmental disorders: Cluster 2 of the proposed meta-structure for DSM-V and ICD-11. Psychol. Med. 2009, 39, 2013–2023. [Google Scholar] [CrossRef] [Green Version]
  274. Guo, J.Y.; Ragland, J.D.; Carter, C.S. Memory and cognition in schizophrenia. Mol. Psychiatry 2019, 24, 633–642. [Google Scholar] [CrossRef]
  275. Ha, S.; Hu, H.; Roussos-Ross, D.; Haidong, K.; Roth, J.; Xu, X. The effects of air pollution on adverse birth outcomes. Environ. Res. 2014, 134, 198–204. [Google Scholar] [CrossRef] [Green Version]
  276. Hollis, F.; Kanellopoulos, A.K.; Bagni, C. Mitochondrial dysfunction in autism spectrum disorder: Clinical features and perspectives. Curr. Opin. Neurobiol. 2017, 45, 178–187. [Google Scholar] [CrossRef]
  277. Legido, A.; Jethva, R.; Goldenthal, M.J. Mitochondrial dysfunction in autism. Semin. Pediatr. Neurol. 2013, 20, 163–175. [Google Scholar] [CrossRef]
  278. Noreika, V.; Falter, C.M.; Rubia, K. Timing deficits in attention-deficit/hyperactivity disorder (ADHD): Evidence from neurocognitive and neuroimaging studies. Neuropsychologia 2013, 51, 235–266. [Google Scholar] [CrossRef]
  279. Robbins, T.W. Pharmacological treatment of cognitive deficits in nondementing mental health disorders. Dialogues Clin. Neurosci. 2019, 21, 301–308. [Google Scholar] [CrossRef] [Green Version]
  280. Sheffield, J.M.; Karcher, N.R.; Barch, D.M. Cognitive deficits in psychotic disorders: A lifespan perspective. Neuropsychol. Rev. 2018, 28, 509–533. [Google Scholar] [CrossRef]
  281. Sheppard, D.P.; Bruineberg, J.P.; Kretschmer-Trendowicz, A.; Altgassen, M. Prospective memory in autism: Theory and literature review. Clin. Neuropsychol. 2018, 32, 748–782. [Google Scholar] [CrossRef] [Green Version]
  282. Fernández, M.; Mollinedo-Gajate, I.; Peñagarikano, O. Neural circuits for social cognition: Implications for autism. Neuroscience 2018, 370, 148–162. [Google Scholar] [CrossRef]
  283. Marazziti, D.; Baroni, S.; Picchetti, M.; Landi, P.; Silvestri, S.; Vatteroni, E.; Catena Dell’Osso, M. Psychiatric disorders and mitochondrial dysfunctions. Eur. Rev. Med. Pharmacol. Sci. 2012, 16, 270–275. [Google Scholar]
  284. Mier, D.; Kirsch, P. Social-cognitive deficits in schizophrenia. Curr. Top. Behav. Neurosci. 2017, 30, 397–409. [Google Scholar] [CrossRef]
  285. Rajasekaran, A.; Venkatasubramanian, G.; Berk, M.; Debnath, M. Mitochondrial dysfunction in schizophrenia: Pathways, mechanisms and implications. Neurosci. Biobehav. Rev. 2015, 48, 10–21. [Google Scholar] [CrossRef] [PubMed]
  286. Rossignol, D.A.; Frye, R.E. Mitochondrial dysfunction in autism spectrum disorders: A systematic review and meta-analysis. Mol. Psychiatry 2012, 17, 290–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Sun, J.; Buys, N. Early executive function deficit in preterm children and its association with neurodevelopmental disorders in childhood: A literature review. Int. J. Adolesc. Med. Health 2012, 24, 291–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Uittenbogaard, M.; Chiaramello, A. Mitochondrial biogenesis: A therapeutic target for neurodevelopmental disorders and neurodegenerative diseases. Curr. Pharm. Des. 2014, 20, 5574–5593. [Google Scholar] [CrossRef] [Green Version]
  289. Yui, K.; Sato, A.; Imataka, G. Mitochondrial dysfunction and its relationship with mTOR signaling and oxidative damage in autism spectrum disorders. Mini Rev. Med. Chem. 2015, 15, 373–389. [Google Scholar] [CrossRef]
  290. Mahone, E.M.; Denckla, M.B. Attention-deficit/hyperactivity disorder: A historical neuropsychological perspective. J. Int. Neuropsychol. Soc. 2017, 23, 916–929. [Google Scholar] [CrossRef] [Green Version]
  291. Verma, P.; Singh, A.; Nthenge-Ngumbau, D.N.; Rajamma, U.; Sinha, S.; Mukhopadhyay, K.; Mohanakumar, K.P. Attention deficit-hyperactivity disorder suffers from mitochondrial dysfunction. BBA Clin. 2016, 6, 153–158. [Google Scholar] [CrossRef] [Green Version]
  292. Ouyang, M.; Cheng, H.; Mishra, V.; Gong, G.; Mosconi, M.W.; Sweeney, J.; Peng, Y.; Huang, H. Atypical age-dependent effects of autism on white matter microstructure in children of 2–7 years. Hum. Brain Mapp. 2016, 37, 819–832. [Google Scholar] [CrossRef] [Green Version]
  293. Turner, A.H.; Greenspan, K.S.; van Erp, T.G.M. Pallidum and lateral ventricle volume enlargement in autism spectrum disorder. Psychiatry Res. Neuroimaging 2016, 252, 40–45. [Google Scholar] [CrossRef] [Green Version]
  294. Gaffney, G.R.; Kuperman, S.; Tsai, L.Y.; Minchin, S. Forebrain structure in infantile autism. J. Am. Acad. Child Adolesc. Psychiatry 1989, 28, 534–537. [Google Scholar] [CrossRef]
  295. Piven, J.; Arndt, S.; Bailey, J.; Havercamp, S.; Andreasen, N.C.; Palmer, P. An MRI study of brain size in autism. Am. J. Psychiatry 1995, 152, 1145–1149. [Google Scholar]
  296. Bouziane, C.; Caan, M.W.A.; Tamminga, H.G.H.; Schrantee, A.; Bottelier, M.A.; de Ruiter, M.B.; Kooij, S.J.J.; Reneman, L. ADHD and maturation of brain white matter: A DTI study in medication naive children and adults. Neuroimage Clin. 2018, 17, 53–59. [Google Scholar] [CrossRef] [PubMed]
  297. Wu, W.; McAnulty, G.; Hamoda, H.M.; Sarill, K.; Karmacharya, S.; Gagoski, B.; Ning, L.; Grant, P.E.; Shenton, M.E.; Waber, D.P.; et al. Detecting microstructural white matter abnormalities of frontal pathways in children with ADHD using advanced diffusion models. Brain Imaging Behav. 2019. [Google Scholar] [CrossRef] [PubMed]
  298. Ertekin, T.; Acer, N.; Koseoglu, E.; Zararsiz, G.; Sonmez, A.; Gumus, K.; Kurtoglu, E. Total intracranial and lateral ventricle volumes measurement in Alzheimer’s disease: A methodological study. J. Clin. Neurosci. 2016, 34, 133–139. [Google Scholar] [CrossRef]
  299. Bartos, A.; Gregus, D.; Ibrahim, I.; Tintera, J. Brain volumes and their ratios in Alzheimer s disease on magnetic resonance imaging segmented using Freesurfer 6.0. Psychiatry Res. Neuroimaging 2019, 287, 70–74. [Google Scholar] [CrossRef] [PubMed]
  300. Marizzoni, M.; Ferrari, C.; Jovicich, J.; Albani, D.; Babiloni, C.; Cavaliere, L.; Didic, M.; Forloni, G.; Galluzzi, S.; Hoffmann, K.T.; et al. Predicting and tracking short term disease progression in amnestic mild cognitive impairment patients with prodromal Alzheimer’s disease: Structural brain biomarkers. J. Alzheimers Dis. 2019, 69, 3–14. [Google Scholar] [CrossRef] [Green Version]
  301. Yang, X.; Tan, M.Z.; Qiu, A. CSF and brain structural imaging markers of the Alzheimer’s pathological cascade. PLoS ONE 2012, 7, e47406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  302. Lewis, M.M.; Smith, A.B.; Styner, M.; Gu, H.; Poole, R.; Zhu, H.; Li, Y.; Barbero, X.; Gouttard, S.; McKeown, M.J.; et al. Asymmetrical lateral ventricular enlargement in Parkinson’s disease. Eur. J. Neurol. 2009, 16, 475–481. [Google Scholar] [CrossRef] [Green Version]
  303. Vazquez-Costa, J.F.; Carratala-Bosca, S.; Tembl, J.I.; Fornes-Ferrer, V.; Perez-Tur, J.; Marti-Bonmati, L.; Sevilla, T. The width of the third ventricle associates with cognition and behaviour in motor neuron disease. Acta Neurol. Scand. 2019, 139, 118–127. [Google Scholar] [CrossRef]
  304. Westeneng, H.J.; Verstraete, E.; Walhout, R.; Schmidt, R.; Hendrikse, J.; Veldink, J.H.; van den Heuvel, M.P.; van den Berg, L.H. Subcortical structures in amyotrophic lateral sclerosis. Neurobiol. Aging 2015, 36, 1075–1082. [Google Scholar] [CrossRef]
  305. Sinnecker, T.; Ruberte, E.; Schädelin, S.; Canova, V.; Amann, M.; Naegelin, Y.; Penner, I.K.; Müller, J.; Kuhle, J.; Décard, B.; et al. New and enlarging white matter lesions adjacent to the ventricle system and thalamic atrophy are independently associated with lateral ventricular enlargement in multiple sclerosis. J. Neurol. 2020, 267, 192–202. [Google Scholar] [CrossRef]
  306. Gaetano, L.; Häring, D.A.; Radue, E.W.; Mueller-Lenke, N.; Thakur, A.; Tomic, D.; Kappos, L.; Sprenger, T. Fingolimod effect on gray matter, thalamus, and white matter in patients with multiple sclerosis. Neurology 2018, 90, e1324–e1332. [Google Scholar] [CrossRef]
  307. Kim, J.H.; Cumming, P.; Son, Y.D.; Kim, H.K.; Joo, Y.H.; Kim, J.H. Altered connectivity between striatal and extrastriatal regions in patients with schizophrenia on maintenance antipsychotics: An [(18) F]fallypride PET and functional MRI study. Synapse 2018, 72, e22064. [Google Scholar] [CrossRef] [Green Version]
  308. Mayo, C.D.; Mazerolle, E.L.; Ritchie, L.; Fisk, J.D.; Gawryluk, J.R. Longitudinal changes in microstructural white matter metrics in Alzheimer’s disease. Neuroimage Clin. 2017, 13, 330–338. [Google Scholar] [CrossRef]
  309. Tang, X.; Qin, Y.; Zhu, W.; Miller, M.I. Surface-based vertexwise analysis of morphometry and microstructural integrity for white matter tracts in diffusion tensor imaging: With application to the corpus callosum in Alzheimer’s disease. Hum. Brain Mapp. 2017, 38, 1875–1893. [Google Scholar] [CrossRef] [Green Version]
  310. Tomimoto, H.; Lin, J.X.; Matsuo, A.; Ihara, M.; Ohtani, R.; Shibata, M.; Miki, Y.; Shibasaki, H. Different mechanisms of corpus callosum atrophy in Alzheimer’s disease and vascular dementia. J. Neurol. 2004, 251, 398–406. [Google Scholar] [CrossRef]
  311. Bledsoe, I.O.; Stebbins, G.T.; Merkitch, D.; Goldman, J.G. White matter abnormalities in the corpus callosum with cognitive impairment in Parkinson disease. Neurology 2018, 91, e2244–e2255. [Google Scholar] [CrossRef]
  312. Goldman, J.G.; Bledsoe, I.O.; Merkitch, D.; Dinh, V.; Bernard, B.; Stebbins, G.T. Corpus callosal atrophy and associations with cognitive impairment in Parkinson disease. Neurology 2017, 88, 1265–1272. [Google Scholar] [CrossRef] [Green Version]
  313. Gorges, M.; Muller, H.P.; Liepelt-Scarfone, I.; Storch, A.; Dodel, R.; Consortium, L.; Hilker-Roggendorf, R.; Berg, D.; Kunz, M.S.; Kalbe, E.; et al. Structural brain signature of cognitive decline in Parkinson’s disease: DTI-based evidence from the LANDSCAPE study. Ther. Adv. Neurol. Disord. 2019, 12, 1756286419843447. [Google Scholar] [CrossRef]
  314. Koirala, N.; Anwar, A.R.; Ciolac, D.; Glaser, M.; Pintea, B.; Deuschl, G.; Muthuraman, M.; Groppa, S. Alterations in white matter network and microstructural integrity differentiate Parkinson’s disease patients and healthy subjects. Front. Aging Neurosci. 2019, 11, 191. [Google Scholar] [CrossRef] [Green Version]
  315. Cai, M.T.; Zhang, Y.X.; Zheng, Y.; Fang, W.; Ding, M.P. Callosal lesions on magnetic resonance imaging with multiple sclerosis, neuromyelitis optica spectrum disorder and acute disseminated encephalomyelitis. Mult. Scler. Relat. Disord. 2019, 32, 41–45. [Google Scholar] [CrossRef]
  316. Garg, N.; Reddel, S.W.; Miller, D.H.; Chataway, J.; Riminton, D.S.; Barnett, Y.; Masters, L.; Barnett, M.H.; Hardy, T.A. The corpus callosum in the diagnosis of multiple sclerosis and other CNS demyelinating and inflammatory diseases. J. Neurol. Neurosurg. Psychiatry 2015, 86, 1374–1382. [Google Scholar] [CrossRef]
  317. Gonçalves, L.I.; Dos Passos, G.R.; Conzatti, L.P.; Burger, J.L.P.; Tomasi, G.H.; Zandoná, M.; Azambuja, L.S.; Gomes, I.; Franco, A.; Sato, D.K.; et al. Correlation between the corpus callosum index and brain atrophy, lesion load, and cognitive dysfunction in multiple sclerosis. Mult. Scler. Relat. Disord. 2018, 20, 154–158. [Google Scholar] [CrossRef]
  318. Granberg, T.; Bergendal, G.; Shams, S.; Aspelin, P.; Kristoffersen-Wiberg, M.; Fredrikson, S.; Martola, J. MRI-defined corpus callosal atrophy in multiple sclerosis: A comparison of volumetric measurements, corpus callosum area and index. J. Neuroimaging 2015, 25, 996–1001. [Google Scholar] [CrossRef]
  319. Gabel, M.C.; Broad, R.J.; Young, A.L.; Abrahams, S.; Bastin, M.E.; Menke, R.A.L.; Al-Chalabi, A.; Goldstein, L.H.; Tsermentseli, S.; Alexander, D.C.; et al. Evolution of white matter damage in amyotrophic lateral sclerosis. Ann. Clin. Transl. Neurol. 2020, 7, 722–732. [Google Scholar] [CrossRef]
  320. Hübers, A.; Böckler, B.; Abaei, A.; Rasche, V.; Lulé, D.; Ercan, E.; Doorenweerd, N.; Müller, H.P.; Dreyhaupt, J.; Kammer, T.; et al. Functional and structural impairment of transcallosal motor fibres in ALS: A study using transcranial magnetic stimulation, diffusion tensor imaging, and diffusion weighted spectroscopy. Brain Imaging Behav. 2020. [Google Scholar] [CrossRef]
  321. Tu, S.; Wang, C.; Menke, R.A.L.; Talbot, K.; Barnett, M.; Kiernan, M.C.; Turner, M.R. Regional callosal integrity and bilaterality of limb weakness in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener 2020. [Google Scholar] [CrossRef]
  322. Benarroch, E.E. Glutamatergic synaptic plasticity and dysfunction in Alzheimer disease: Emerging mechanisms. Neurology 2018, 91, 125–132. [Google Scholar] [CrossRef]
  323. Butterfield, D.A.; Pocernich, C.B. The glutamatergic system and Alzheimer’s disease: Therapeutic implications. CNS Drugs 2003, 17, 641–652. [Google Scholar] [CrossRef]
  324. Hynd, M.R.; Scott, H.L.; Dodd, P.R. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer’s disease. Neurochem. Int. 2004, 45, 583–595. [Google Scholar] [CrossRef]
  325. Ahmed, I.; Bose, S.K.; Pavese, N.; Ramlackhansingh, A.; Turkheimer, F.; Hotton, G.; Hammers, A.; Brooks, D.J. Glutamate NMDA receptor dysregulation in Parkinson’s disease with dyskinesias. Brain 2011, 134, 979–986. [Google Scholar] [CrossRef]
  326. Obal, I.; Majlath, Z.; Toldi, J.; Vecsei, L. Mental disturbances in Parkinson’s disease and related disorders: The role of excitotoxins. J. Parkinsons Dis. 2014, 4, 139–150. [Google Scholar] [CrossRef]
  327. Frigo, M.; Cogo, M.G.; Fusco, M.L.; Gardinetti, M.; Frigeni, B. Glutamate and multiple sclerosis. Curr. Med. Chem. 2012, 19, 1295–1299. [Google Scholar] [CrossRef]
  328. Levite, M. Glutamate, T cells and multiple sclerosis. J. Neural. Transm. 2017, 124, 775–798. [Google Scholar] [CrossRef]
  329. Macrez, R.; Stys, P.K.; Vivien, D.; Lipton, S.A.; Docagne, F. Mechanisms of glutamate toxicity in multiple sclerosis: Biomarker and therapeutic opportunities. Lancet Neurol. 2016, 15, 1089–1102. [Google Scholar] [CrossRef]
  330. Stojanovic, I.R.; Kostic, M.; Ljubisavljevic, S. The role of glutamate and its receptors in multiple sclerosis. J. Neural. Transm. 2014, 121, 945–955. [Google Scholar] [CrossRef]
  331. Fogarty, M.J. Driven to decay: Excitability and synaptic abnormalities in amyotrophic lateral sclerosis. Brain Res. Bull. 2018, 140, 318–333. [Google Scholar] [CrossRef]
  332. Holecek, V.; Rokyta, R. Possible etiology and treatment of amyotrophic lateral sclerosis. Neuroendocrinol. Lett. 2018, 38, 528–531. [Google Scholar]
  333. Shaw, P.J.; Ince, P.G. Glutamate, excitotoxicity and amyotrophic lateral sclerosis. J. Neurol. 1997, 244, S3–S14. [Google Scholar] [CrossRef]
  334. Øie, M.G.; Andersen, P.N.; Hovik, K.T.; Skogli, E.W.; Rund, B.R. Similar impairments shown on a neuropsychological test battery in adolescents with high-functioning autism and early onset schizophrenia: A two-year follow-up study. Cogn. Neuropsychiatry 2020, 25, 163–178. [Google Scholar] [CrossRef]
  335. Rabiee, A.; Vasaghi-Gharamaleki, B.; Samadi, S.A.; Amiri-Shavaki, Y.; Alaghband-Rad, J. Working memory deficits and its relationship to autism spectrum disorders. Iran. J. Med. Sci. 2020, 45, 100–109. [Google Scholar] [CrossRef]
  336. Seng, G.J.; Tseng, W.L.; Chiu, Y.N.; Tsai, W.C.; Wu, Y.Y.; Gau, S.S. Executive functions in youths with autism spectrum disorder and their unaffected siblings. Psychol. Med. 2020. [Google Scholar] [CrossRef]
  337. Valeri, G.; Casula, L.; Napoli, E.; Stievano, P.; Trimarco, B.; Vicari, S.; Scalisi, T.G. Executive functions and symptom severity in an Italian sample of intellectually able preschoolers with autism spectrum disorder. J. Autism Dev. Disord. 2020, 50, 3207–3215. [Google Scholar] [CrossRef] [Green Version]
  338. Teigset, C.M.; Mohn, C.; Rund, B.R. Perinatal complications and executive dysfunction in early-onset schizophrenia. BMC Psychiatry 2020, 20, 103. [Google Scholar] [CrossRef] [Green Version]
  339. Vahdani, B.; Armani Kian, A.; Esmaeilzadeh, A.; Zenoozian, S.; Yousefi, V.; Mazloomzadeh, S. Adjunctive raloxifene and isradipine improve cognitive functioning in patients with schizophrenia: A pilot study. J. Clin. Psychopharmacol. 2020. [Google Scholar] [CrossRef]
  340. Kaga, Y.; Ueda, R.; Tanaka, M.; Kita, Y.; Suzuki, K.; Okumura, Y.; Egashira, Y.; Shirakawa, Y.; Mitsuhashi, S.; Kitamura, Y.; et al. Executive dysfunction in medication-naïve children with ADHD: A multi-modal fNIRS and EEG study. Brain Dev. 2020, 42, 555–563. [Google Scholar] [CrossRef]
  341. Kim, M.J.; Park, H.Y.; Yoo, E.Y.; Kim, J.R. Effects of a cognitive-functional intervention method on improving executive function and self-directed learning in school-aged children with attention deficit hyperactivity disorder: A single-subject design study. Occup. Ther. Int. 2020, 2020, 1250801. [Google Scholar] [CrossRef]
  342. Taş Torun, Y.; Işik Taner, Y.; Güney, E.; İseri, E. Osmotic release oral system-methylphenidate hydrochloride (OROS-MPH) versus atomoxetine on executive function improvement and clinical effectiveness in ADHD: A randomized controlled trial. Appl. Neuropsychol. Child 2020. [Google Scholar] [CrossRef]
  343. Besag, F.M.C.; Vasey, M.J. Social cognition and psychopathology in childhood and adolescence. Epilepsy Behav. E&B 2019, 100, 106210. [Google Scholar] [CrossRef]
  344. Pallathra, A.A.; Cordero, L.; Wong, K.; Brodkin, E.S. Psychosocial interventions targeting social functioning in adults on the autism spectrum: A literature review. Curr. Psychiatry Rep. 2019, 21, 5. [Google Scholar] [CrossRef]
  345. Reyes, N.M.; Pickard, K.; Reaven, J. Emotion regulation: A treatment target for autism spectrum disorder. Bull. Menninger Clin. 2019, 83, 205–234. [Google Scholar] [CrossRef]
  346. Kimoto, S.; Makinodan, M.; Kishimoto, T. Neurobiology and treatment of social cognition in schizophrenia: Bridging the bed-bench gap. Neurobiol. Dis. 2019, 131, 104315. [Google Scholar] [CrossRef]
  347. Aman, M.G.; Farmer, C.A.; Hollway, J.; Arnold, L.E. Treatment of inattention, overactivity, and impulsiveness in autism spectrum disorders. Child Adolesc. Psychiatr. Clin. N. Am. 2008, 17, 713–738. [Google Scholar] [CrossRef] [Green Version]
  348. Skoretz, P.; Tang, C. Stimulants for impulsive violence in schizophrenia spectrum disordered women: A case series and brief review. CNS Spectr. 2016, 21, 445–449. [Google Scholar] [CrossRef]
  349. Patros, C.H.; Alderson, R.M.; Kasper, L.J.; Tarle, S.J.; Lea, S.E.; Hudec, K.L. Choice-impulsivity in children and adolescents with attention-deficit/hyperactivity disorder (ADHD): A meta-analytic review. Clin. Psychol. Rev. 2016, 43, 162–174. [Google Scholar] [CrossRef]
  350. Kirova, A.M.; Bays, R.B.; Lagalwar, S. Working memory and executive function decline across normal aging, mild cognitive impairment, and Alzheimer’s disease. Biomed. Res. Int. 2015, 2015, 748212. [Google Scholar] [CrossRef] [Green Version]
  351. Li, J.Q.; Tan, L.; Wang, H.F.; Tan, M.S.; Tan, L.; Xu, W.; Zhao, Q.F.; Wang, J.; Jiang, T.; Yu, J.T. Risk factors for predicting progression from mild cognitive impairment to Alzheimer’s disease: A systematic review and meta-analysis of cohort studies. J. Neurol. Neurosurg. Psychiatry 2016, 87, 476–484. [Google Scholar] [CrossRef] [Green Version]
  352. Ding, W.; Ding, L.J.; Li, F.F.; Han, Y.; Mu, L. Neurodegeneration and cognition in Parkinson’s disease: A review. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 2275–2281. [Google Scholar]
  353. Hoogland, J.; van Wanrooij, L.L.; Boel, J.A.; Goldman, J.G.; Stebbins, G.T.; Dalrymple-Alford, J.C.; Marras, C.; Adler, C.H.; Junque, C.; Pedersen, K.F.; et al. Detecting mild cognitive deficits in Parkinson’s disease: Comparison of neuropsychological tests. Mov. Disord. 2018, 33, 1750–1759. [Google Scholar] [CrossRef]
  354. Chiaravalloti, N.D.; DeLuca, J. Cognitive impairment in multiple sclerosis. Lancet Neurol. 2008, 7, 1139–1151. [Google Scholar] [CrossRef]
  355. Grzegorski, T.; Losy, J. Cognitive impairment in multiple sclerosis—A review of current knowledge and recent research. Rev. Neurosci. 2017, 28, 845–860. [Google Scholar] [CrossRef]
  356. Koutsouraki, E.; Kalatha, T.; Grosi, E.; Koukoulidis, T.; Michmizos, D. Cognitive decline in Multiple Sclerosis patients. Hell. J. Nucl. Med. 2019, 22, 75–81. [Google Scholar]
  357. Miller, E.; Morel, A.; Redlicka, J.; Miller, I.; Saluk, J. Pharmacological and non-pharmacological therapies of cognitive impairment in multiple sclerosis. Curr. Neuropharmacol. 2018, 16, 475–483. [Google Scholar] [CrossRef]
  358. Beeldman, E.; Raaphorst, J.; Klein Twennaar, M.; de Visser, M.; Schmand, B.A.; de Haan, R.J. The cognitive profile of ALS: A systematic review and meta-analysis update. J. Neurol. Neurosurg. Psychiatry 2016, 87, 611–619. [Google Scholar] [CrossRef] [Green Version]
  359. Goldstein, L.H.; Abrahams, S. Changes in cognition and behaviour in amyotrophic lateral sclerosis: Nature of impairment and implications for assessment. Lancet Neurol. 2013, 12, 368–380. [Google Scholar] [CrossRef]
  360. Irwin, D.; Lippa, C.F.; Swearer, J.M. Cognition and amyotrophic lateral sclerosis (ALS). Am. J. Alzheimers Dis. Other Demen. 2007, 22, 300–312. [Google Scholar] [CrossRef]
  361. Khin Khin, E.; Minor, D.; Holloway, A.; Pelleg, A. Decisional capacity in amyotrophic lateral sclerosis. J. Am. Acad. Psychiatry Law 2015, 43, 210–217. [Google Scholar]
  362. Phukan, J.; Pender, N.P.; Hardiman, O. Cognitive impairment in amyotrophic lateral sclerosis. Lancet Neurol. 2007, 6, 994–1003. [Google Scholar] [CrossRef]
  363. Vance, D.E.; Moore, B.S.; Farr, K.F.; Struzick, T. Procedural memory and emotional attachment in Alzheimer disease: Implications for meaningful and engaging activities. J. Neurosci. Nurs. 2008, 40, 96–102. [Google Scholar] [CrossRef]
  364. Palmeri, R.; Lo Buono, V.; Corallo, F.; Foti, M.; Di Lorenzo, G.; Bramanti, P.; Marino, S. Nonmotor symptoms in Parkinson disease: A descriptive review on social cognition ability. J. Geriatr. Psychiatry Neurol. 2017, 30, 109–121. [Google Scholar] [CrossRef]
  365. Sofologi, M.; Koutsouraki, E.; Tsolaki, M.; Tsolaki, A.; Koukoulidis, T.; Theofilidis, A.; Papantoniou, G.; Moraitou, D. Analyzing social cognition and understanding of social inferences in patients with multiple sclerosis. A comparative study. Hell. J. Nucl. Med. 2019, 22, 15–26. [Google Scholar]
  366. Dondu, A.; Sevincoka, L.; Akyol, A.; Tataroglu, C. Is obsessive-compulsive symptomatology a risk factor for Alzheimer-type dementia? Psychiatry Res. 2015, 225, 381–386. [Google Scholar] [CrossRef]
  367. Keszycki, R.M.; Fisher, D.W.; Dong, H. The hyperactivity-impulsivity-irritiability-disinhibition-aggression-agitation domain in Alzheimer’s disease: Current management and future directions. Front. Pharmacol. 2019, 10, 1109. [Google Scholar] [CrossRef] [Green Version]
  368. Jeon, N.; Bortolato, M. What drugs modify the risk of iatrogenic impulse-control disorders in Parkinson’s disease? A preliminary pharmacoepidemiologic study. PLoS ONE 2020, 15, e0227128. [Google Scholar] [CrossRef]
  369. O’Callaghan, C. The multifaceted nature of impulsivity in Parkinson’s disease. Brain 2019, 142, 3666–3669. [Google Scholar] [CrossRef]
  370. Toro, J.; Blanco, L.; Orozco-Cabal, L.F.; Díaz, C.; Reyes, S.; Burbano, L.; Cuéllar-Giraldo, D.F.; Duque, A.; Patiño, J.; Cortés, F. Impulsivity traits in patients with multiple sclerosis. Mult. Scler. Relat. Disord. 2018, 22, 148–152. [Google Scholar] [CrossRef]
  371. Schumann, C.M.; Amaral, D.G. Stereological analysis of amygdala neuron number in autism. J. Neurosci. 2006, 26, 7674. [Google Scholar] [CrossRef]
  372. Falkai, P.; Bogerts, B. Cell loss in the hippocampus of schizophrenics. Eur. Arch. Psychiatry Neurol. Sci. 1986, 236, 154–161. [Google Scholar] [CrossRef]
  373. Ardalan, M.; Chumak, T.; Vexler, Z.; Mallard, C. Sex-dependent effects of perinatal inflammation on the brain: Implication for neuro-psychiatric disorders. Int. J. Mol. Sci. 2019, 20, 2270. [Google Scholar] [CrossRef] [Green Version]
  374. Flinkkila, E.; Keski-Rahkonen, A.; Marttunen, M.; Raevuori, A. Prenatal inflammation, infections and mental disorders. Psychopathology 2016, 49, 317–333. [Google Scholar] [CrossRef]
  375. Cortelazzo, A.; De Felice, C.; Guerranti, R.; Signorini, C.; Leoncini, S.; Zollo, G.; Leoncini, R.; Timperio, A.M.; Zolla, L.; Ciccoli, L.; et al. Expression and oxidative modifications of plasma proteins in autism spectrum disorders: Interplay between inflammatory response and lipid peroxidation. Proteom. Clin. Appl. 2016, 10, 1103–1112. [Google Scholar] [CrossRef]
  376. El-Ansary, A.; Bjorklund, G.; Chirumbolo, S.; Alnakhli, O.M. Predictive value of selected biomarkers related to metabolism and oxidative stress in children with autism spectrum disorder. Metab. Brain Dis. 2017, 32, 1209–1221. [Google Scholar] [CrossRef] [PubMed]
  377. Frye, R.E.; James, S.J. Metabolic pathology of autism in relation to redox metabolism. Biomark. Med. 2014, 8, 321–330. [Google Scholar] [CrossRef] [Green Version]
  378. Instanes, J.T.; Halmøy, A.; Engeland, A.; Haavik, J.; Furu, K.; Klungsøyr, K. Attention-deficit/hyperactivity disorder in offspring of mothers with inflammatory and immune system diseases. Biol. Psychiatry 2017, 81, 452–459. [Google Scholar] [CrossRef] [Green Version]
  379. Eckert, A.; Marques, C.A.; Keil, U.; Schussel, K.; Muller, W.E. Increased apoptotic cell death in sporadic and genetic Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2003, 1010, 604–609. [Google Scholar] [CrossRef]
  380. Herrup, K. The contributions of unscheduled neuronal cell cycle events to the death of neurons in Alzheimer’s disease. Front. Biosci. (Elite Ed) 2012, 4, 2101–2109. [Google Scholar] [CrossRef]
  381. Michel, P.P.; Hirsch, E.C.; Hunot, S. Understanding dopaminergic cell death pathways in Parkinson disease. Neuron 2016, 90, 675–691. [Google Scholar] [CrossRef] [Green Version]
  382. Surmeier, D.J. Determinants of dopaminergic neuron loss in Parkinson’s disease. FEBS J. 2018, 285, 3657–3668. [Google Scholar] [CrossRef] [Green Version]
  383. Papadopoulos, D.; Dukes, S.; Patel, R.; Nicholas, R.; Vora, A.; Reynolds, R. Substantial archaeocortical atrophy and neuronal loss in multiple sclerosis. Brain Pathol. 2009, 19, 238–253. [Google Scholar] [CrossRef]
  384. Schirmer, L.; Albert, M.; Buss, A.; Schulz-Schaeffer, W.J.; Antel, J.P.; Brück, W.; Stadelmann, C. Substantial early, but nonprogressive neuronal loss in multiple sclerosis (MS) spinal cord. Ann. Neurol. 2009, 66, 698–704. [Google Scholar] [CrossRef]
  385. Wegner, C.; Esiri, M.M.; Chance, S.A.; Palace, J.; Matthews, P.M. Neocortical neuronal, synaptic, and glial loss in multiple sclerosis. Neurology 2006, 67, 960–967. [Google Scholar] [CrossRef] [PubMed]
  386. Iwai, Y.; Shibuya, K.; Misawa, S.; Sekiguchi, Y.; Watanabe, K.; Amino, H.; Kuwabara, S. Axonal dysfunction precedes motor neuronal death in amyotrophic lateral sclerosis. PLoS ONE 2016, 11, e0158596. [Google Scholar] [CrossRef]
  387. Macdonald, R.; Barnes, K.; Hastings, C.; Mortiboys, H. Mitochondrial abnormalities in Parkinson’s disease and Alzheimer’s disease: Can mitochondria be targeted therapeutically? Biochem. Soc. Trans. 2018, 46, 891–909. [Google Scholar] [CrossRef] [PubMed]
  388. Swerdlow, R.H. Mitochondria and mitochondrial cascades in Alzheimer’s disease. J. Alzheimers Dis. 2018, 62, 1403–1416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  389. Tobore, T.O. On the central role of mitochondria dysfunction and oxidative stress in Alzheimer’s disease. Neurol. Sci. 2019, 40, 1527–1540. [Google Scholar] [CrossRef]
  390. Bose, A.; Beal, M.F. Mitochondrial dysfunction in Parkinson’s disease. J. Neurochem. 2016, 139, 216–231. [Google Scholar] [CrossRef] [PubMed]
  391. Lin, K.J.; Lin, K.L.; Chen, S.D.; Liou, C.W.; Chuang, Y.C.; Lin, H.Y.; Lin, T.K. The overcrowded crossroads: Mitochondria, alpha-synuclein, and the endo-lysosomal system interaction in Parkinson’s disease. Int. J. Mol. Sci. 2019, 20, 5312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  392. Campbell, G.; Mahad, D.J. Mitochondrial dysfunction and axon degeneration in progressive multiple sclerosis. FEBS Lett. 2018, 592, 1113–1121. [Google Scholar] [CrossRef] [PubMed]
  393. Rajda, C.; Pukoli, D.; Bende, Z.; Majláth, Z.; Vécsei, L. Excitotoxins, mitochondrial and redox disturbances in multiple sclerosis. Int. J. Mol. Sci. 2017, 18, 353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Witte, M.E.; Mahad, D.J.; Lassmann, H.; van Horssen, J. Mitochondrial dysfunction contributes to neurodegeneration in multiple sclerosis. Trends Mol. Med. 2014, 20, 179–187. [Google Scholar] [CrossRef] [PubMed]
  395. Greco, V.; Longone, P.; Spalloni, A.; Pieroni, L.; Urbani, A. Crosstalk between oxidative stress and mitochondrial damage: Focus on amyotrophic lateral sclerosis. Adv. Exp. Med. Biol. 2019, 1158, 71–82. [Google Scholar] [CrossRef]
  396. Hervias, I.; Beal, M.F.; Manfredi, G. Mitochondrial dysfunction and amyotrophic lateral sclerosis. Muscle Nerve 2006, 33, 598–608. [Google Scholar] [CrossRef] [PubMed]
  397. Smith, E.F.; Shaw, P.J.; De Vos, K.J. The role of mitochondria in amyotrophic lateral sclerosis. Neurosci. Lett. 2019, 710, 132933. [Google Scholar] [CrossRef]
  398. Dani, M.; Wood, M.; Mizoguchi, R.; Fan, Z.; Walker, Z.; Morgan, R.; Hinz, R.; Biju, M.; Kuruvilla, T.; Brooks, D.J.; et al. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer’s disease. Brain 2018, 141, 2740–2754. [Google Scholar] [CrossRef] [PubMed]
  399. Fan, Z.; Brooks, D.J.; Okello, A.; Edison, P. An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain 2017, 140, 792–803. [Google Scholar] [CrossRef] [PubMed]
  400. Hansen, D.V.; Hanson, J.E.; Sheng, M. Microglia in Alzheimer’s disease. J. Cell Biol. 2018, 217, 459–472. [Google Scholar] [CrossRef] [PubMed]
  401. Regen, F.; Hellmann-Regen, J.; Costantini, E.; Reale, M. Neuroinflammation and Alzheimer’s Disease: Implications for microglial activation. Curr. Alzheimer Res. 2017, 14, 1140–1148. [Google Scholar] [CrossRef] [PubMed]
  402. Sarlus, H.; Heneka, M.T. Microglia in Alzheimer’s disease. J. Clin. Investig. 2017, 127, 3240–3249. [Google Scholar] [CrossRef] [PubMed]
  403. Ho, M.S. Microglia in Parkinson’s disease. Adv. Exp. Med. Biol. 2019, 1175, 335–353. [Google Scholar] [CrossRef]
  404. Sanchez-Guajardo, V.; Tentillier, N.; Romero-Ramos, M. The relation between alpha-synuclein and microglia in Parkinson’s disease: Recent developments. Neuroscience 2015, 302, 47–58. [Google Scholar] [CrossRef]
  405. Vivekanantham, S.; Shah, S.; Dewji, R.; Dewji, A.; Khatri, C.; Ologunde, R. Neuroinflammation in Parkinson’s disease: Role in neurodegeneration and tissue repair. Int. J. Neurosci. 2015, 125, 717–725. [Google Scholar] [CrossRef]
  406. Correale, J.; Gaitán, M.I.; Ysrraelit, M.C.; Fiol, M.P. Progressive multiple sclerosis: From pathogenic mechanisms to treatment. Brain 2017, 140, 527–546. [Google Scholar] [CrossRef] [Green Version]
  407. Matthews, P.M. Chronic inflammation in multiple sclerosis—Seeing what was always there. Nat. Rev. Neurol. 2019, 15, 582–593. [Google Scholar] [CrossRef]
  408. Patejdl, R.; Penner, I.K.; Noack, T.K.; Zettl, U.K. Multiple sclerosis and fatigue: A review on the contribution of inflammation and immune-mediated neurodegeneration. Autoimmun. Rev. 2016, 15, 210–220. [Google Scholar] [CrossRef] [PubMed]
  409. Voet, S.; Prinz, M.; van Loo, G. Microglia in central nervous system inflammation and multiple sclerosis pathology. Trends Mol. Med. 2019, 25, 112–123. [Google Scholar] [CrossRef]
  410. Evans, M.C.; Couch, Y.; Sibson, N.; Turner, M.R. Inflammation and neurovascular changes in amyotrophic lateral sclerosis. Mol. Cell Neurosci. 2013, 53, 34–41. [Google Scholar] [CrossRef] [PubMed]
  411. Lyon, M.S.; Wosiski-Kuhn, M.; Gillespie, R.; Caress, J.; Milligan, C. Inflammation, immunity, and amyotrophic lateral sclerosis: I. Etiology and pathology. Muscle Nerve 2019, 59, 10–22. [Google Scholar] [CrossRef] [Green Version]
  412. Thonhoff, J.R.; Simpson, E.P.; Appel, S.H. Neuroinflammatory mechanisms in amyotrophic lateral sclerosis pathogenesis. Curr. Opin. Neurol. 2018, 31, 635–639. [Google Scholar] [CrossRef] [PubMed]
  413. Ganguly, G.; Chakrabarti, S.; Chatterjee, U.; Saso, L. Proteinopathy, oxidative stress and mitochondrial dysfunction: Cross talk in Alzheimer’s disease and Parkinson’s disease. Drug Des. Dev. Ther. 2017, 11, 797–810. [Google Scholar] [CrossRef] [Green Version]
  414. Jiang, T.; Sun, Q.; Chen, S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson’s disease and Alzheimer’s disease. Prog. Neurobiol. 2016, 147, 1–19. [Google Scholar] [CrossRef] [PubMed]
  415. Umeno, A.; Biju, V.; Yoshida, Y. In vivo ROS production and use of oxidative stress-derived biomarkers to detect the onset of diseases such as Alzheimer’s disease, Parkinson’s disease, and diabetes. Free Radic. Res. 2017, 51, 413–427. [Google Scholar] [CrossRef]
  416. Tonnies, E.; Trushina, E. Oxidative stress, synaptic dysfunction, and Alzheimer’s disease. J. Alzheimers Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  417. Subramaniam, S.R.; Chesselet, M.F. Mitochondrial dysfunction and oxidative stress in Parkinson’s disease. Prog. Neurobiol. 2013, 106–107, 17–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  418. Ibitoye, R.; Kemp, K.; Rice, C.; Hares, K.; Scolding, N.; Wilkins, A. Oxidative stress-related biomarkers in multiple sclerosis: A review. Biomark. Med. 2016, 10, 889–902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  419. Lassmann, H.; van Horssen, J. Oxidative stress and its impact on neurons and glia in multiple sclerosis lesions. Biochim. Biophys. Acta 2016, 1862, 506–510. [Google Scholar] [CrossRef]
  420. Lassmann, H.; van Horssen, J.; Mahad, D. Progressive multiple sclerosis: Pathology and pathogenesis. Nat. Rev. Neurol. 2012, 8, 647–656. [Google Scholar] [CrossRef]
  421. Ohl, K.; Tenbrock, K.; Kipp, M. Oxidative stress in multiple sclerosis: Central and peripheral mode of action. Exp. Neurol. 2016, 277, 58–67. [Google Scholar] [CrossRef] [PubMed]
  422. Bozzo, F.; Mirra, A.; Carrì, M.T. Oxidative stress and mitochondrial damage in the pathogenesis of ALS: New perspectives. Neurosci. Lett. 2017, 636, 3–8. [Google Scholar] [CrossRef]
  423. D’Amico, E.; Factor-Litvak, P.; Santella, R.M.; Mitsumoto, H. Clinical perspective on oxidative stress in sporadic amyotrophic lateral sclerosis. Free Radic. Biol. Med. 2013, 65, 509–527. [Google Scholar] [CrossRef] [Green Version]
  424. Ault, A.P.; Peters, T.M.; Sawvel, E.J.; Casuccio, G.S.; Willis, R.D.; Norris, G.A.; Grassian, V.H. Single-particle SEM-EDX analysis of iron-containing coarse particulate matter in an urban environment: Sources and distribution of iron within Cleveland, Ohio. Environ. Sci. Technol. 2012, 46, 4331–4339. [Google Scholar] [CrossRef]
  425. Wang, B.; Wang, Q.; Chen, H.; Zhou, X.; Wang, H.; Wang, H.; Zhang, J.; Feng, W. Size-dependent translocation pattern, chemical and biological transformation of nano- and submicron-sized ferric oxide particles in the central nervous system. J. Nanosci. Nanotechnol. 2016, 16, 5553–5561. [Google Scholar] [CrossRef] [PubMed]
  426. Sutunkova, M.P.; Katsnelson, B.A.; Privalova, L.I.; Gurvich, V.B.; Konysheva, L.K.; Shur, V.Y.; Shishkina, E.V.; Minigalieva, I.A.; Solovjeva, S.N.; Grebenkina, S.V.; et al. On the contribution of the phagocytosis and the solubilization to the iron oxide nanoparticles retention in and elimination from lungs under long-term inhalation exposure. Toxicology 2016, 363–364, 19–28. [Google Scholar] [CrossRef] [PubMed]
  427. Hopkins, L.E.; Laing, E.A.; Peake, J.L.; Uyeminami, D.; Mack, S.M.; Li, X.; Smiley-Jewell, S.; Pinkerton, K.E. Repeated iron-soot exposure and nose-to-brain transport of inhaled ultrafine particles. Toxicol. Pathol. 2018, 46, 75–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  428. Maher, B.A.; Ahmed, I.A.; Karloukovski, V.; MacLaren, D.A.; Foulds, P.G.; Allsop, D.; Mann, D.M.; Torres-Jardon, R.; Calderon-Garciduenas, L. Magnetite pollution nanoparticles in the human brain. Proc. Natl. Acad. Sci. USA 2016, 113, 10797–10801. [Google Scholar] [CrossRef] [Green Version]
  429. Wu, J.; Ding, T.; Sun, J. Neurotoxic potential of iron oxide nanoparticles in the rat brain striatum and hippocampus. Neurotoxicology 2013, 34, 243–253. [Google Scholar] [CrossRef]
  430. Lubczynska, M.J.; Sunyer, J.; Tiemeier, H.; Porta, D.; Kasper-Sonnenberg, M.; Jaddoe, V.W.V.; Basagana, X.; Dalmau-Bueno, A.; Forastiere, F.; Wittsiepe, J.; et al. Exposure to elemental composition of outdoor PM2.5 at birth and cognitive and psychomotor function in childhood in four European birth cohorts. Environ. Int. 2017, 109, 170–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  431. Chen, J.H.; Singh, N.; Tay, H.; Walczyk, T. Imbalance of iron influx and efflux causes brain iron accumulation over time in the healthy adult rat. Metallomics 2014, 6, 1417–1426. [Google Scholar] [CrossRef]
  432. Banks, W.A.; Kastin, A.J.; Fasold, M.B.; Barrera, C.M.; Augereau, G. Studies of the slow bidirectional transport of iron and transferrin across the blood-brain barrier. Brain Res. Bull. 1988, 21, 881–885. [Google Scholar] [CrossRef]
  433. Dallman, P.R.; Spirito, R.A. Brain iron in the rat: Extremely slow turnover in normal rats may explain long-lasting effects of early iron deficiency. J. Nutr. 1977, 107, 1075–1081. [Google Scholar] [CrossRef] [PubMed]
  434. Chenery, S.R.N.; Sarkar, S.K.; Chatterjee, M.; Marriott, A.L.; Watts, M.J. Heavy metals in urban road dusts from Kolkata and Bengaluru, India: Implications for human health. Environ. Geochem. Health 2020, 42, 2627–2643. [Google Scholar] [CrossRef]
  435. Zhang, L.; Bai, R.; Liu, Y.; Meng, L.; Li, B.; Wang, L.; Xu, L.; Le Guyader, L.; Chen, C. The dose-dependent toxicological effects and potential perturbation on the neurotransmitter secretion in brain following intranasal instillation of copper nanoparticles. Nanotoxicology 2012, 6, 562–575. [Google Scholar] [CrossRef] [PubMed]
  436. Bai, R.; Zhang, L.; Liu, Y.; Li, B.; Wang, L.; Wang, P.; Autrup, H.; Beer, C.; Chen, C. Integrated analytical techniques with high sensitivity for studying brain translocation and potential impairment induced by intranasally instilled copper nanoparticles. Toxicol. Lett. 2014, 226, 70–80. [Google Scholar] [CrossRef] [PubMed]
  437. Liu, Y.; Gao, Y.; Liu, Y.; Li, B.; Chen, C.; Wu, G. Oxidative stress and acute changes in murine brain tissues after nasal instillation of copper particles with different sizes. J. Nanosci. Nanotechnol. 2014, 14, 4534–4540. [Google Scholar] [CrossRef]
  438. Alemany, S.; Vilor-Tejedor, N.; Bustamante, M.; Álvarez-Pedrerol, M.; Rivas, I.; Forns, J.; Querol, X.; Pujol, J.; Sunyer, J. Interaction between airborne copper exposure and ATP7B polymorphisms on inattentiveness in scholar children. Int. J. Hyg. Environ. Health 2017, 220, 51–56. [Google Scholar] [CrossRef]
  439. Persson, E.; Henriksson, J.; Tallkvist, J.; Rouleau, C.; Tjalve, H. Transport and subcellular distribution of intranasally administered zinc in the olfactory system of rats and pikes. Toxicology 2003, 191, 97–108. [Google Scholar] [CrossRef]
  440. Kao, Y.Y.; Cheng, T.J.; Yang, D.M.; Wang, C.T.; Chiung, Y.M.; Liu, P.S. Demonstration of an olfactory bulb-brain translocation pathway for ZnO nanoparticles in rodent cells in vitro and in vivo. J. Mol. Neurosci. 2012, 48, 464–471. [Google Scholar] [CrossRef]
  441. Liu, H.; Yang, H.; Fang, Y.; Li, K.; Tian, L.; Liu, X.; Zhang, W.; Tan, Y.; Lai, W.; Bian, L.; et al. Neurotoxicity and biomarkers of zinc oxide nanoparticles in main functional brain regions and dopaminergic neurons. Sci. Total Environ. 2020, 705, 135809. [Google Scholar] [CrossRef]
  442. Dorman, D.C.; Brenneman, K.A.; McElveen, A.M.; Lynch, S.E.; Roberts, K.C.; Wong, B.A. Olfactory transport: A direct route of delivery of inhaled manganese phosphate to the rat brain. J. Toxicol. Environ. Health Part A 2002, 65, 1493–1511. [Google Scholar] [CrossRef]
  443. Lewis, J.; Bench, G.; Myers, O.; Tinner, B.; Staines, W.; Barr, E.; Divine, K.K.; Barrington, W.; Karlsson, J. Trigeminal uptake and clearance of inhaled manganese chloride in rats and mice. Neurotoxicology 2005, 26, 113–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  444. Colin-Barenque, L.; Souza-Gallardo, L.M.; Fortoul, T.I. Toxic effects of inhaled manganese on the olfactory bulb: An ultrastructural approach in mice. J. Electron. Microsc. 2011, 60, 73–78. [Google Scholar] [CrossRef] [PubMed]
  445. Moberly, A.H.; Czarnecki, L.A.; Pottackal, J.; Rubinstein, T.; Turkel, D.J.; Kass, M.D.; McGann, J.P. Intranasal exposure to manganese disrupts neurotransmitter release from glutamatergic synapses in the central nervous system in vivo. Neurotoxicology 2012, 33, 996–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  446. Normandin, L.; Carrier, G.; Gardiner, P.F.; Kennedy, G.; Hazell, A.S.; Mergler, D.; Butterworth, R.F.; Philippe, S.; Zayed, J. Assessment of bioaccumulation, neuropathology, and neurobehavior following subchronic (90 days) inhalation in Sprague-Dawley rats exposed to manganese phosphate. Toxicol. Appl. Pharmacol. 2002, 183, 135–145. [Google Scholar] [CrossRef] [PubMed]
  447. Tapin, D.; Kennedy, G.; Lambert, J.; Zayed, J. Bioaccumulation and locomotor effects of manganese sulfate in Sprague-Dawley rats following subchronic (90 days) inhalation exposure. Toxicol. Appl. Pharmacol. 2006, 211, 166–174. [Google Scholar] [CrossRef]
  448. Dobson, A.W.; Weber, S.; Dorman, D.C.; Lash, L.K.; Erikson, K.M.; Aschner, M. Oxidative stress is induced in the rat brain following repeated inhalation exposure to manganese sulfate. Biol. Trace Elem. Res. 2003, 93, 113–126. [Google Scholar] [CrossRef]
  449. Taylor, M.D.; Erikson, K.M.; Dobson, A.W.; Fitsanakis, V.A.; Dorman, D.C.; Aschner, M. Effects of inhaled manganese on biomarkers of oxidative stress in the rat brain. Neurotoxicology 2006, 27, 788–797. [Google Scholar] [CrossRef] [PubMed]
  450. Erikson, K.M.; Dorman, D.C.; Lash, L.H.; Dobson, A.W.; Aschner, M. Airborne manganese exposure differentially affects end points of oxidative stress in an age- and sex-dependent manner. Biol. Trace Elem. Res. 2004, 100, 49–62. [Google Scholar] [CrossRef] [Green Version]
  451. Dorman, D.C.; McManus, B.E.; Marshall, M.W.; James, R.A.; Struve, M.F. Old age and gender influence the pharmacokinetics of inhaled manganese sulfate and manganese phosphate in rats. Toxicol. Appl. Pharmacol. 2004, 197, 113–124. [Google Scholar] [CrossRef] [PubMed]
  452. Erikson, K.M.; Dorman, D.C.; Fitsanakis, V.; Lash, L.H.; Aschner, M. Alterations of oxidative stress biomarkers due to in utero and neonatal exposures of airborne manganese. Biol. Trace Elem. Res. 2006, 111, 199–215. [Google Scholar] [CrossRef] [Green Version]
  453. Erikson, K.M.; Dorman, D.C.; Lash, L.H.; Aschner, M. Persistent alterations in biomarkers of oxidative stress resulting from combined in utero and neonatal manganese inhalation. Biol. Trace Elem. Res. 2005, 104, 151–163. [Google Scholar] [CrossRef]
  454. Foster, M.L.; Rao, D.B.; Francher, T.; Traver, S.; Dorman, D.C. Olfactory toxicity in rats following manganese chloride nasal instillation: A pilot study. Neurotoxicology 2018, 64, 284–290. [Google Scholar] [CrossRef]
  455. Saputra, D.; Chang, J.; Lee, B.J.; Yoon, J.H.; Kim, J.; Lee, K. Short-term manganese inhalation decreases brain dopamine transporter levels without disrupting motor skills in rats. J. Toxicol. Sci. 2016, 41, 391–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  456. Sanchez-Betancourt, J.; Anaya-Martinez, V.; Gutierrez-Valdez, A.L.; Ordonez-Librado, J.L.; Montiel-Flores, E.; Espinosa-Villanueva, J.; Reynoso-Erazo, L.; Avila-Costa, M.R. Manganese mixture inhalation is a reliable Parkinson disease model in rats. Neurotoxicology 2012, 33, 1346–1355. [Google Scholar] [CrossRef] [PubMed]
  457. Elder, A.; Gelein, R.; Silva, V.; Feikert, T.; Opanashuk, L.; Carter, J.; Potter, R.; Maynard, A.; Ito, Y.; Finkelstein, J.; et al. Translocation of inhaled ultrafine manganese oxide particles to the central nervous system. Environ. Health Perspect. 2006, 114, 1172–1178. [Google Scholar] [CrossRef] [PubMed]
  458. Dorman, D.C.; Struve, M.F.; Wong, B.A.; Dye, J.A.; Robertson, I.D. Correlation of brain magnetic resonance imaging changes with pallidal manganese concentrations in rhesus monkeys following subchronic manganese inhalation. Toxicol. Sci. 2006, 92, 219–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  459. Erikson, K.M.; Dorman, D.C.; Lash, L.H.; Aschner, M. Manganese inhalation by rhesus monkeys is associated with brain regional changes in biomarkers of neurotoxicity. Toxicol. Sci. 2007, 97, 459–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  460. Erikson, K.M.; Dorman, D.C.; Lash, L.H.; Aschner, M. Duration of airborne-manganese exposure in rhesus monkeys is associated with brain regional changes in biomarkers of neurotoxicity. Neurotoxicology 2008, 29, 377–385. [Google Scholar] [CrossRef] [Green Version]
  461. Carvalho, C.F.; Menezes-Filho, J.A.; de Matos, V.P.; Bessa, J.R.; Coelho-Santos, J.; Viana, G.F.; Argollo, N.; Abreu, N. Elevated airborne manganese and low executive function in school-aged children in Brazil. Neurotoxicology 2014, 45, 301–308. [Google Scholar] [CrossRef] [PubMed]
  462. Ingenbleek, Y.; Kimura, H. Nutritional essentiality of sulfur in health and disease. Nutr. Rev. 2013, 71, 413–432. [Google Scholar] [CrossRef]
  463. Yargicoglu, P.; Agar, A.; Gumuslu, S.; Bilmen, S.; Oguz, Y. Age-related alterations in antioxidant enzymes, lipid peroxide levels, and somatosensory-evoked potentials: Effect of sulfur dioxide. Arch. Environ. Contam. Toxicol. 1999, 37, 554–560. [Google Scholar] [CrossRef]
  464. Kilic, D. The effects of ageing and sulfur dioxide inhalation exposure on visual-evoked potentials, antioxidant enzyme systems, and lipid-peroxidation levels of the brain and eye. Neurotoxicol. Teratol. 2003, 25, 587–598. [Google Scholar] [CrossRef]
  465. Meng, Z.; Zhang, B. Oxidative damage of sulfur dioxide inhalation on brains and livers of mice. Environ. Toxicol. Pharmacol. 2003, 13, 1–8. [Google Scholar] [CrossRef]
  466. Meng, Z. Oxidative damage of sulfur dioxide on various organs of mice: Sulfur dioxide is a systemic oxidative damage agent. Inhal. Toxicol. 2003, 15, 181–195. [Google Scholar] [CrossRef] [PubMed]
  467. Wu, D.; Meng, Z. Effect of sulfur dioxide inhalation on the glutathione redox system in mice and protective role of sea buckthorn seed oil. Arch. Environ. Contam. Toxicol. 2003, 45, 423–428. [Google Scholar] [CrossRef] [PubMed]
  468. Meng, Z.; Li, R.; Zhang, X. Levels of sulfite in three organs from mice exposed to sulfur [corrected] dioxide. Inhal. Toxicol. 2005, 17, 309–313. [Google Scholar] [CrossRef]
  469. Meng, Z.; Liu, Y. Cell morphological ultrastructural changes in various organs from mice exposed by inhalation to sulfur dioxide. Inhal. Toxicol. 2007, 19, 543–551. [Google Scholar] [CrossRef] [PubMed]
  470. Qin, G.; Wang, J.; Huo, Y.; Yan, H.; Jiang, C.; Zhou, J.; Wang, X.; Sang, N. Sulfur dioxide inhalation stimulated mitochondrial biogenesis in rat brains. Toxicology 2012, 300, 67–74. [Google Scholar] [CrossRef]
  471. Yun, Y.; Yao, G.; Yue, H.; Guo, L.; Qin, G.; Li, G.; Sang, N. SO(2) inhalation causes synaptic injury in rat hippocampus via its derivatives in vivo. Chemosphere 2013, 93, 2426–2432. [Google Scholar] [CrossRef]
  472. Yao, G.; Yun, Y.; Sang, N. Differential effects between one week and four weeks exposure to same mass of SO on synaptic plasticity in rat hippocampus. Environ. Toxicol. 2014. [Google Scholar] [CrossRef]
  473. Slikker, W., Jr.; Andersen, M.E.; Bogdanffy, M.S.; Bus, J.S.; Cohen, S.D.; Conolly, R.B.; David, R.M.; Doerrer, N.G.; Dorman, D.C.; Gaylor, D.W.; et al. Dose-dependent transitions in mechanisms of toxicity. Toxicol. Appl. Pharmacol. 2004, 201, 203–225. [Google Scholar] [CrossRef]
  474. Baisch, B.L.; Corson, N.M.; Wade-Mercer, P.; Gelein, R.; Kennell, A.J.; Oberdörster, G.; Elder, A. Equivalent titanium dioxide nanoparticle deposition by intratracheal instillation and whole body inhalation: The effect of dose rate on acute respiratory tract inflammation. Part. Fibre Toxicol. 2014, 11, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  475. Yao, G.; Yue, H.; Yun, Y.; Sang, N. Chronic SO inhalation above environmental standard impairs neuronal behavior and represses glutamate receptor gene expression and memory-related kinase activation via neuroinflammation in rats. Environ. Res. 2014, 137C, 85–93. [Google Scholar] [CrossRef]
  476. Igbokwe, I.O.; Igwenagu, E.; Igbokwe, N.A. Aluminium toxicosis: A review of toxic actions and effects. Interdiscip. Toxicol. 2019, 12, 45–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  477. Klotz, K.; Weistenhöfer, W.; Neff, F.; Hartwig, A.; van Thriel, C.; Drexler, H. The health effects of aluminum exposure. Dtsch. Arztebl. Int. 2017, 114, 653–659. [Google Scholar] [CrossRef] [Green Version]
  478. Röllin, H.B.; Theodorou, P.; Kilroe-Smith, T.A. Deposition of aluminium in tissues of rabbits exposed to inhalation of low concentrations of Al2O3 dust. Br. J. Ind. Med. 1991, 48, 389–391. [Google Scholar] [CrossRef] [Green Version]
  479. Kwon, J.T.; Seo, G.B.; Jo, E.; Lee, M.; Kim, H.M.; Shim, I.; Lee, B.W.; Yoon, B.I.; Kim, P.; Choi, K. Aluminum nanoparticles induce ERK and p38MAPK activation in rat brain. Toxicol. Res. 2013, 29, 181–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  480. Zhang, X.; Xu, Y.; Zhou, L.; Zhang, C.; Meng, Q.; Wu, S.; Wang, S.; Ding, Z.; Chen, X.; Li, X.; et al. Sex-dependent depression-like behavior induced by respiratory administration of aluminum oxide nanoparticles. Int. J. Environ. Res. Public Health 2015, 12, 15692–15705. [Google Scholar] [CrossRef]
  481. Murugadoss, S.; Lison, D.; Godderis, L.; Van Den Brule, S.; Mast, J.; Brassinne, F.; Sebaihi, N.; Hoet, P.H. Toxicology of silica nanoparticles: An update. Arch. Toxicol. 2017, 91, 2967–3010. [Google Scholar] [CrossRef]
  482. Parveen, A.; Rizvi, S.H.; Sushma; Mahdi, F.; Ahmad, I.; Singh, P.P.; Mahdi, A.A. Intranasal exposure to silica nanoparticles induces alterations in pro-inflammatory environment of rat brain. Toxicol. Ind. Health 2017, 33, 119–132. [Google Scholar] [CrossRef]
  483. Lebedova, J.; Novakova, Z.; Vecera, Z.; Buchtova, M.; Dumkova, J.; Docekal, B.; Blahova, L.; Mikuska, P.; Misek, I.; Hampl, A.; et al. Impact of acute and subchronic inhalation exposure to PbO nanoparticles on mice. Nanotoxicology 2018, 12, 290–304. [Google Scholar] [CrossRef]
  484. Blahova, L.; Novakova, Z.; Vecera, Z.; Vrlikova, L.; Docekal, B.; Dumkova, J.; Krumal, K.; Mikuska, P.; Buchtova, M.; Hampl, A.; et al. The effects of nano-sized PbO on biomarkers of membrane disruption and DNA damage in a sub-chronic inhalation study on mice. Nanotoxicology 2020, 14, 214–231. [Google Scholar] [CrossRef]
  485. Sutunkova, M.P.; Solovyeva, S.N.; Chernyshov, I.N.; Klinova, S.V.; Gurvich, V.B.; Shur, V.Y.; Shishkina, E.V.; Zubarev, I.V.; Privalova, L.I.; Katsnelson, B.A. Manifestation of systemic toxicity in rats after a short-time inhalation of lead oxide nanoparticles. Int. J. Mol. Sci. 2020, 21, 690. [Google Scholar] [CrossRef] [Green Version]
  486. Ahsan, S.M.; Rao, C.M.; Ahmad, M.F. Nanoparticle-protein interaction: The significance and role of protein corona. Adv. Exp. Med. Biol. 2018, 1048, 175–198. [Google Scholar] [CrossRef]
  487. Tenzer, S.; Docter, D.; Rosfa, S.; Wlodarski, A.; Kuharev, J.; Rekik, A.; Knauer, S.K.; Bantz, C.; Nawroth, T.; Bier, C.; et al. Nanoparticle size is a critical physicochemical determinant of the human blood plasma corona: A comprehensive quantitative proteomic analysis. ACS Nano 2011, 5, 7155–7167. [Google Scholar] [CrossRef] [PubMed]
  488. Maiorano, G.; Sabella, S.; Sorce, B.; Brunetti, V.; Malvindi, M.A.; Cingolani, R.; Pompa, P.P. Effects of cell culture media on the dynamic formation of protein-nanoparticle complexes and influence on the cellular response. ACS Nano 2010, 4, 7481–7491. [Google Scholar] [CrossRef] [PubMed]
  489. Nagayama, S.; Ogawara, K.; Fukuoka, Y.; Higaki, K.; Kimura, T. Time-dependent changes in opsonin amount associated on nanoparticles alter their hepatic uptake characteristics. Int. J. Pharm. 2007, 342, 215–221. [Google Scholar] [CrossRef]
  490. Cox, A.; Andreozzi, P.; Dal Magro, R.; Fiordaliso, F.; Corbelli, A.; Talamini, L.; Chinello, C.; Raimondo, F.; Magni, F.; Tringali, M.; et al. Evolution of nanoparticle protein corona across the blood–brain barrier. ACS Nano 2018, 12, 7292–7300. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Maps of the United States from the first National Emissions Inventory PM2.5 measured at ambient monitoring sites across the U.S. It depicts the total emissions (ton/year) as well as the ug/m3/day of PM2.5, and the trace elements and metals found in AP (air pollution), including Fe, Al, Si, S, Zn, Pb, and Cu. Levels of Fe, Al, Si, and S were among the highest, with lower exposure levels for Zn, Pb, and Cu. Modified from Reff et al., 2009 [83].
Figure 1. Maps of the United States from the first National Emissions Inventory PM2.5 measured at ambient monitoring sites across the U.S. It depicts the total emissions (ton/year) as well as the ug/m3/day of PM2.5, and the trace elements and metals found in AP (air pollution), including Fe, Al, Si, S, Zn, Pb, and Cu. Levels of Fe, Al, Si, and S were among the highest, with lower exposure levels for Zn, Pb, and Cu. Modified from Reff et al., 2009 [83].
Atmosphere 11 01098 g001
Figure 2. Tree-maps of trace elements from elements from ambient UFP exposures analyzed via XRF analysis of filters from postnatal (left; Rochester, NY) or gestational exposures (right; Tuxedo NY). Modified from Cory-Slechta et al., 2019 [92].
Figure 2. Tree-maps of trace elements from elements from ambient UFP exposures analyzed via XRF analysis of filters from postnatal (left; Rochester, NY) or gestational exposures (right; Tuxedo NY). Modified from Cory-Slechta et al., 2019 [92].
Atmosphere 11 01098 g002
Figure 3. Sagittal brain sections representative postnatal air (left) vs. UFP-exposed (right) males depicting metals as indicated and analyzed by ICP-MS. Modified from Cory-Slechta et al., 2019 [92].
Figure 3. Sagittal brain sections representative postnatal air (left) vs. UFP-exposed (right) males depicting metals as indicated and analyzed by ICP-MS. Modified from Cory-Slechta et al., 2019 [92].
Atmosphere 11 01098 g003
Figure 4. Schematic of brain with arrows depicting origins of the brain pathology—i.e., brain stem and olfactory bulb in Parkinson’s disease based on extensive autopsies. Corresponding routes allow ultrafine particle air pollution to enter the brain while bypassing the blood brain barrier.
Figure 4. Schematic of brain with arrows depicting origins of the brain pathology—i.e., brain stem and olfactory bulb in Parkinson’s disease based on extensive autopsies. Corresponding routes allow ultrafine particle air pollution to enter the brain while bypassing the blood brain barrier.
Atmosphere 11 01098 g004
Table 1. Shared characterristics of neurodevelopment disorders and neurodegenerative diseases and current understanding of the impact of inhaled nanoparticulate metals in reproducing these characteristics.
Table 1. Shared characterristics of neurodevelopment disorders and neurodegenerative diseases and current understanding of the impact of inhaled nanoparticulate metals in reproducing these characteristics.
CharacteristicNDDsNDGDsFeCuZnMnSPbAlSiCa
VentriculomegalyASD, SCZ, ADHDAD, PD, MS, ALS
Altered MyelinationASD, SCZ, ADHDAD, PD, MS, ALS
Interhemispheric DisconnectionASD, SCZ, ADHDAD, PD, MS, ALS
Glutamatergic DysfunctionASD, SCZ, ADHDAD, PD, MS, ALS
Neuronal Cell DeathASD, SCZAD, PD, MS, ALS
Inflammation/Microglial ActivationASD, SCZ, ADHDAD, PD, MS, ALS
Oxidative StressASD, SCZ, ADHDAD, PD, MS, ALS
Cognitive/Executive DeficitsASD, SCZ, ADHDAD, PD, MS, ALS
Mitochondrial DysfunctionASD, SCZ, ADHDAD, PD, MS, ALS
Social Behavioral DeficitsASD, SCZ, ADHDAD, PD, MS, ALS
ImpulsivityASD, SCZ, ADHDAD, PD, MS
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cory-Slechta, D.A.; Sobolewski, M.; Oberdörster, G. Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases. Atmosphere 2020, 11, 1098. https://doi.org/10.3390/atmos11101098

AMA Style

Cory-Slechta DA, Sobolewski M, Oberdörster G. Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases. Atmosphere. 2020; 11(10):1098. https://doi.org/10.3390/atmos11101098

Chicago/Turabian Style

Cory-Slechta, Deborah A., Marissa Sobolewski, and Günter Oberdörster. 2020. "Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases" Atmosphere 11, no. 10: 1098. https://doi.org/10.3390/atmos11101098

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

Article Metrics

Back to TopTop