Next Article in Journal
Cysteine Is the Only Universally Affected and Disfavored Proteomic Amino Acid under Oxidative Conditions in Animals
Previous Article in Journal
Understanding the Factors That Influence the Antioxidant Activity of Manganosalen Complexes with Neuroprotective Effects
Previous Article in Special Issue
The Roles of Neutrophil-Derived Myeloperoxidase (MPO) in Diseases: The New Progress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Crosstalk between Oxidative Stress and Inflammation Caused by Noise and Air Pollution—Implications for Neurodegenerative Diseases

by
Marin Kuntić
1,2,
Omar Hahad
1,2,
Thomas Münzel
1,2,*,† and
Andreas Daiber
1,2,*,†
1
Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
2
German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2024, 13(3), 266; https://doi.org/10.3390/antiox13030266
Submission received: 16 January 2024 / Revised: 14 February 2024 / Accepted: 19 February 2024 / Published: 22 February 2024

Abstract

:
Neurodegenerative diseases are often referred to as diseases of old age, and with the aging population, they are gaining scientific and medical interest. Environmental stressors, most notably traffic noise and air pollution, have recently come to the forefront, and have emerged as disease risk factors. The evidence for a connection between environmental risk factors and neurodegenerative disease is growing. In this review, the most common neurodegenerative diseases and their epidemiological association with traffic noise and air pollution are presented. Also, the most important mechanisms involved in neurodegenerative disease development, oxidative stress, and neuroinflammation are highlighted. An overview of the in vivo findings will provide a mechanistic link between noise, air pollution, and neurodegenerative pathology. Finally, the importance of the direct and indirect pathways, by which noise and air pollution cause cerebral damage, is discussed. More high-quality data are still needed from both epidemiological and basic science studies in order to better understand the causal connection between neurodegenerative diseases and environmental risk factors.

1. Introduction

1.1. Air and Noise Pollution—Overview

As the world is becoming more and more industrialized, and medicine and sanitation are becoming better in combating communicable diseases, the scientific community is focusing on risk factors of the physical environment, such as air and noise pollution [1,2]. Air pollution is a broad term used to describe the toxic gases, liquids, and solids derived from natural and artificial sources present in the atmosphere [3]. According to the Global Burden of Disease study, air pollution ranked 4th among the leading causes of global morbidity and mortality for both males and females [4]. The global mortality burden from air pollution was assessed by multiple models and recently reached 8.7 million to 10.2 million annual deaths, accounting for almost 20% of all deaths worldwide [5,6]. Solid constituents of air pollution, commonly known as particulate matter (PM), recently gained significant interest because of its prevalence in developing and industrialized countries, complementing gaseous air pollution, like ozone and nitric oxides, which have been studied for many decades [3,7]. Apart from the composition of PM, its size is a major determinant of its toxicity for human organ systems [8,9].
Traffic noise has become a prominent modern risk factor for human disease, making the famous words of the Nobel Prize laureate Robert Koch prophetic: “One day man will have to fight noise as relentlessly as plague and cholera” [10]. The World Health Organization (WHO) estimates that at least 1.6 million healthy life years are lost yearly due to traffic-related noise in Western Europe alone, and that 40% of people in the European Union live in areas where traffic-related noise exceeds the recommended thresholds [11]. Although traffic noise is recognized as a prominent risk factor, it was not included in the latest report of the Global Burden of Disease study (only occupational noise was mentioned there, rather at the bottom of the ranking [12]).
In 2005, Christopher P. Wild coined the term “exposome” in order to describe all of the environmental exposures that influence human physiology over the duration of a lifespan [13]. This term does not only include external stressors such as climate, pathogens, UV radiation, noise, and air pollution but also sociological factors such as lifestyle and socioeconomic status, psychological factors such as anxiety and depression, and daily surroundings, such as urban environment and green spaces [14,15,16]. Exposome research is important, as it provides an important link between disease onset and progression and human endeavors. It provides a rational basis for the statement “Genetics load the gun, but environment pulls the trigger” [17,18] and the estimation that 2/3 of all chronic diseases have environmental instead of genetic causes [19].

1.2. Neurodegenerative Disease—Overview

Advances in medicine and hygiene during the past century have significantly contributed to eradicating communicable diseases. Today, due to the aging population, non-communicable diseases such as cardiovascular diseases (CVD) and neurodegenerative diseases, together with cancer, represent the major contributors to the global disease burden [20]. Neurodegenerative diseases are usually referred to as diseases of old age, as they are most prevalent in the elderly population [21,22]. It is estimated that 50 million people globally suffer from dementia and that this number will increase to 130 million by 2050 [23,24]. The most common neurodegenerative diseases are Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), and they will be the focus of this review. Other neurodegenerative diseases, like multiple sclerosis, Huntington’s disease, multiple system atrophy, and frontotemporal dementia, are also relatively common, and will be addressed where appropriate.
AD is the most common type of dementia and the most common neurodegenerative disease, with a prevalence of almost two-thirds of all dementia cases [25]. As some reports predict that dementia will almost double by the year 2050 [26], strategies to prevent and alleviate AD will become essential for reducing the disease burden. AD is characterized by loss of memory and general cognitive decline, resulting in aggression, confusion, problems with attention and language, mood-swings, and sleep impairment [27,28,29]. The prognosis for AD patients is poor with only 5–9 years of life left after diagnosis [30]. A diagnosis of AD severity usually follows a classic dementia seven-stage progression model, starting with no impairment and moving through stages of cognitive decline, all the way to late-stage dementia [31]. The disease is often identified late, when patients notice an ongoing cognitive impairment. The presence of AD as a subtype of dementia was previously only possible by post-mortem autopsy, as clear atrophy of the frontal and temporal cortices is visible [32]. Modern diagnostic tools, including magnetic resonance imaging (MRI), and blood and spinal fluid biomarkers, improved early diagnosis of AD [33,34]. There are several mechanistic concepts of AD pathophysiology, and the most widely accepted one is the aggregation of the extracellular amyloid beta (Aβ) protein plaques and the formation of the intracellular neurofibrillary tangles (NFTs) of tau protein [35]. These protein plaques inhibit synapse communication, promote inflammation through microglia activation, and impair blood flow in the brain (cerebral amyloid angiopathy) [36].
PD is a syndrome with different underlying causes and variable symptoms. Still, it is overall characterized by the loss of the motor neuron function, resulting in tremors, rigidity, slowness and difficulty in movement, and impairment in posture [37]. There are currently over 10 million people living with PD worldwide, and the number of newly diagnosed patients is rising every year [38]. Unlike other neurodegenerative diseases, PD patients can live with the symptoms for a long time, as the prodromal period is substantially longer than in other types of neurodegenerative diseases [39]. Only a small percentage (5–10%) of all PD cases are inherited, although results from the genome-wide association study have identified 90 independent genome-wide significant risk signals [40]. Most of the PD cases occur sporadically (90–95%). Symptoms of PD are a result of the death of neurons in the substantia nigra, a region of the midbrain that supplies dopamine to the basal ganglia [41]. The pathology of PD is defined by Lewy bodies, an accumulation of α-synuclein protein inside neurons [42]. Misfolded α-synuclein was also found in AD patients, but it is still not recognized as a major cause of AD pathology, as it is for the PD pathology [43]. Lewy bodies consist not only of α-synuclein aggregates but also contain lipids in the form of membrane fragments, mitochondria, and vesicular structures [44]. PD is usually treated with dopamine replacement, which suppresses the symptoms, but no cure is available [45].
ALS, also known as Lou Gehrig’s disease, results in the progressive loss of motor neurons in the cerebral cortex, brainstem, and spinal cord [46]. ALS patients suffer from muscle wasting, weakness, and paralysis, leading to death by respiratory failure [47]. ALS is relatively rare in comparison with AD and PD, with incidence rate of 10–15 cases per 100,000 capita [48]. The progression of ALS is rapid, with death occurring 2–3 years after diagnosis [49]. Only 10% of all cases are familial, but mutations in up to 40 gene mutations have been associated with ALS [50]. Due to such a wide variety of different possible genetic causes, there is no known cure for ALS, only therapies that alleviate symptoms, such as antioxidant therapies and treatments for dysphagia and respiratory failure [51]. More than 20 years ago, mutation of the superoxide dismutase (SOD1) gene was identified as the major cause of the familial ALS [52], but other genes, such as fused in sarcoma (FUS), transacting response DNA-binding protein (TARDBP), and chromosome 9 open reading frame 72 (C9ORF72), were also shown to be accountable for the large percentage of familial ALS cases [53]. The pathology of ALS also includes aggregations of ubiquitinated proteins in the cytoplasm of degenerating motor neurons and surrounding oligodendrocytes [54].

2. Role of Oxidative Stress and Inflammation in Neurodegenerative Diseases

Neurodegenerative diseases are most often characterized by the accumulation of protein aggregates in either the intracellular or the extracellular space of the neuronal system [55]. This accumulation of protein aggregates is accompanied by neuroinflammation [56,57,58] and oxidative stress [27,59,60]. Oxidative stress is a pathophysiological state in which the amount of reactive oxygen and nitrogen species (RONS) produced exceeds the capacity of the system to remove them successfully or to repair the resulting oxidative damage [61,62]. This imbalance happens due to many factors but is ubiquitous in almost all human diseases [63,64]. Inflammation of the nervous tissue is a hallmark of neurodegenerative diseases [56]. Inflammation and oxidative stress are almost inseparable and exhibit a crosstalk [65] that has been explored deeply for many years in both human and animal models [57,66]. Immune cells that infiltrate tissue produce large amounts of RONS as a host defense mechanism, and oxidative stress in tissues is a signal for the recruitment of pro-inflammatory immune cells [67]. Therefore, exploring oxidative stress and inflammation in neurodegenerative pathophysiology as an intertwined pathomechanism is important. The general interplay between neuroinflammation and oxidative stress is shown in Figure 1. It is important to keep in mind that neuroinflammation is not the only source of cerebral oxidative stress, and that other prominent sources like the mitochondria and the dopamine metabolism can significantly contribute to neurodegeneration [68,69]. The RONS that we talk about in the subsequent sections, are mostly those produced from NADPH oxidases during inflammation and by mitochondria upon stress challenges (superoxide and hydrogen peroxide) or direct physicochemical effects from air pollutants (e.g., nano-sized combustion-derived particles). The term RONS also comprises the major product of superoxide and nitric oxide, peroxynitrite. Besides these most important RONS, there may be some minor contribution by hydroxyl radicals that are directly formed from transition metal reactions with peroxides or other redox-active chemicals on the surface of air pollution particles. More details on the species formed and their sources can be found in recent overviews [3,70].

2.1. Neurodegenerative Disease and Oxidative Stress

Although the brain constitutes only around 2% of the total body mass, it is responsible for consuming 20% of the available oxygen [71]. The high oxygen consumption is needed to produce ATP in order to maintain the neuronal membrane potential through Na+ and K+ gradients [72]. Mitochondrial respiratory chain, responsible for most oxygen consumption, can leak electrons to reduce molecular oxygen and form superoxide anion radical (O2) [73]. Although there are mechanisms to eliminate the O2 from the cell via enzymatic catalysis by SOD enzymes, the kinetically favorable reaction of O2 with nitric oxide (NO) will ultimately lead to peroxynitrite (ONOO) formation before O2 can be eliminated [74]. NO is produced by nitric oxide synthases (NOS) that are expressed as different isoforms, e.g., by endothelial cells (eNOS) and neuronal cells (nNOS), or can be induced (iNOS) in immune cells, such as macrophages and microglia, but also other cell types, such as astrocytes [75,76]. Free radicals are important at physiological concentrations as they can act as cellular signaling molecules. Still, during oxidative stress, they are produced at high levels and can damage lipids, proteins, and nucleic acids. As the cerebral tissue is rich in functional lipids it is especially susceptible to oxidative stress damage [77].
During many years of AD research, oxidative stress markers have often been associated with AD pathology, especially by using post-mortem brain material of AD patients. In these brain samples, researchers identified elevated activity of glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase, pointing to the increased demand for reducing equivalents in the form of NADPH for brain peroxide metabolism [78]. Classical markers of lipid peroxidation, such as malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and F2-isoprostanes were also observed in AD patient brains [79,80]. It was even proposed that these lipid peroxides are responsible for the observed AD protein aggregates by creating protein adducts [81]. Evidence of protein and nucleic acid damage from oxidative stress in AD patients was envisaged by protein carbonylation and an increase in 8-hydroxyguanosine (8-OHG) levels in brain homogenates [82,83]. Increased 3-nitrotyrosine (3-NT) levels, a marker of ONOO-mediated damage, were observed in AD patient brains and cerebral blood vessels, leading to the hypothesis that Aβ plaques can alter iNOS and eNOS expression [84,85,86]. The AD-associated oxidative stress is also accompanied by lower reduced glutathione levels (mostly measured by a diminished GSH/GSSG ratio) and reduced expression of antioxidant enzymes, such as SOD enzymes, catalase (CAT), and glutathione peroxidases (GPx) [87,88,89]. Most of the antioxidant therapies used to combat AD progression failed, and many of the drugs used to reduce cognitive decline did not provide solid evidence of oxidative stress reduction, pointing to the possibility that the oxidative stress is a symptom of AD pathology rather than a cause [90].
Similarly to AD, markers of oxidative stress were also identified in PD patients, but they were mostly localized in the substantia nigra [41]. Increased levels of MDA, 4-HNE, and acrolein-modified proteins were all observed in the substantia nigra, frontal cortex, and brainstem but not in other brain regions, such as the caudate nucleus and putamen [91,92,93,94,95]. This is important as 4-HNE adducts with α-synuclein protein were shown to promote aggregation, a hallmark of PD [96]. Also, nitration of α-synuclein was suggested as an oxidative mechanism promoting aggregate formation [97,98]. Other markers of oxidative stress, such as protein carbonyls, 8-OHG and 8-OHdG were also identified in substantia nigra, pointing to direct protein and nucleic acid oxidation [99,100]. Interestingly, some research shows that protein carbonyls might come from L-DOPA, a therapeutic drug used to treat dopamine deficiency in PD, as it can have pro-oxidant properties via hydroxyl radicals [101]. L-DOPA could also provide the substrates for mitochondrial monoamine oxidases, e.g., dopamine and other catecholamines, with subsequent hydrogen peroxide formation, which was therapeutically exploited using monoamine oxidase inhibitors in PD patients [102]. Another prominent feature of dopamine is its ability to undergo auto-oxidation, or metal-oxidation reactions, resulting in formation of dopamine quinones [103]. The short-lived dopamine-o-quinone is created together with O2, and can undergo further oxidation to form aminochrome [104]. Both dopamine-o-quinone and aminochrome are highly reactive and form protein adducts on the cysteine, lysine, and tyrosine residues, causing specific damage to mitochondria and thereby worsening the PD pathology. An increase in oxidative stress in PD patients is also accompanied by a decrease in GSH and an increase in SOD levels, specifically in substantia nigra [41,105,106]. The involvement of mitochondria was also observed in PD patients with a reduction in respiratory chain complexes activity [107,108], and inhibition of complex I, e.g., by rotenone, was previously shown to cause Parkinsonism [109], which was even used as an animal model of PD. Therapies with coenzyme Q10, an important molecule for the function of the mitochondrial respiratory chain, showed limited but positive outcomes on the progression of PD [110], although later meta-analysis showed no improvement compared to placebo [111].
ALS is tightly related to oxidative stress as one of the major mutations associated with familial ALS is on the SOD1 gene [53]. A connection between the impaired function of SOD1, peroxynitrite formation, and ALS was recognized early [52]. The mutated gene can produce misfolded proteins that create aggregates and lose their catalytic function, rendering the enzyme unable to clear O2, thus producing oxidative stress [112,113]. Other mutated genes identified in ALS patients can also contribute to oxidative stress. The protein encoded by the TARDBP gene is known to affect the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, an important antioxidant response mechanism [114,115]. The protein encoded by the FUS gene is involved in the repair of DNA-double strand breaks, and C9ORF72 gene mutation can produce dipeptide repeat proteins that are responsible for the interruption of mitochondrial membrane potential and loss of DNA repair [116,117]. Like in AD and PD patients, markers of oxidative damage of proteins, lipids, and nucleic acids were found in ALS patients. 4-HNE and 3-NT were observed to be increased in the cerebrospinal fluid of ALS patients together with 8-OHdG [118,119,120,121]. Although the levels of oxidative stress markers were not associated with the severity of the ALS progression, some studies have directly shown increases in oxidative stress in certain brain regions of ALS patients [122,123]. Similarly to the AD and PD, almost all antioxidant therapies have failed to produce a meaningful improvement of symptoms or prolonged life [124]. Interestingly, the ALS drug edaravone, which was proposed to confer its pharmacological action through free radical and peroxynitrite scavenging, was shown to improve patient conditions, but only in a group with a high vital capacity and early-stage disease diagnosis [125].

2.2. Neurodegenerative Diseases and Inflammation

Inflammation is a hallmark of all neurodegenerative diseases as the accumulation of misfolded proteins into plaques can activate pathogen-associated and damage-associated molecular patterns (PAMPs and DAMPs) leading to sterile inflammation [126,127]. Some of the most prominent receptors tasked with these pattern recognitions in the nervous system are the Toll-like receptors (TLRs), which are present in microglia and astrocytes [128]. Activated TLRs mediate the release of both pro- and anti-inflammatory cytokines, which can either help with the repair of tissue damage and resolution of inflammation in the acute setting or promote further damage and oxidative stress in a chronic inflammatory setting [129,130]. The TLR signaling activates transcription factors, like activator protein 1 (AP-1) and nuclear factor ‘kappa-light-chain-enhancer’ of activated B-cells (NF-κB), that transcribe genes for pro-inflammatory cytokines [131]. Another class of danger sensing receptors, the pattern recognition receptors (PRRs), are located in the cytoplasm where they can be activated by the misfolded protein accumulation. These cytoplasmic PRRs are often called the inflammasome (most notably the nucleotide-binding domain and leucine-rich repeat-containing receptors family pyrin domain containing 3 (NLRP3)). They can activate pro-inflammatory pathways, e.g., via IL-1β [56,132]. Since the presence of misfolded protein aggregates produces a response where the microglia try to clear the cell debris through M2 polarization and release of anti-inflammatory cytokines, the long-lasting presence of protein plaques and the failure of microglia to effectively clear them lead to pro-inflammatory M1 polarization [133]. These long-lasting protein plaques are a hallmark of AD and PD, providing ideal conditions for chronic inflammation development. Activated microglia express iNOS that can, together with the activation of the (phagocytic) NADPH oxidase, produce peroxynitrite anion that nitrates proteins [134]. This was indeed observed in the brains of AD patients, pointing to a strong immune cell activation [135,136]. It was previously observed that Aβ could initiate the NF-κB sterile inflammation pathway though through the activation of a cluster of differentiation 36 (CD36), further promoting TLR4 and TLR6 activity [137]. CD36 can also promote Aβ aggregation and induce NLRP3 inflammasome, increasing IL-1β release [138]. IL-1β can later increase the activity of p38 mitogen-activated protein kinases (p38-MAPK) and glycogen synthase kinase 3 (GSK-3), promoting the phosphorylation of the tau protein, another hallmark of AD [139]. In PD, the presence of α-synuclein can also directly stimulate the NLRP3 inflammasome [140,141]. Importantly, ROS derived from mitochondria [142] or from NADPH oxidase [65] can activate the NLRP3 inflammasome by redox-dependent pathways, including thioredoxin-interacting protein (TXNIP) as a central redox switch [143].
The incidence of multiple sclerosis is similar to that of ALS, but multiple sclerosis is considered the most common type of chronic inflammatory disease of the CNS [56]. Multiple sclerosis is characterized by demyelination of neurons, inhibiting signal transduction [144]. Damaged myelin sheets are taken up and degraded by activated resident macrophages, creating a multiple sclerosis lesion [145]. These lesions typically consist of CD3+/CD8+ T lymphocytes and of CD68-positive microglia, and oxidized lipids [146,147]. In multiple sclerosis, macrophages and other circulating immune cells can cross the blood–brain barrier, creating an adaptive immune response, further damaging the neuronal tissue and promoting inflammation [148].
ALS is a systemic disease that attacks the motor neurons. Thus, it is outside of the brain tissue itself, making the adaptive immune response a bigger contributor to the overall inflammation burden [149]. Many circulating markers of inflammation were observed in ALS patients. Still, individually, these markers often fail to provide a good measurement for the severity and the progression of the disease [150]. Most of the many mutated genes in ALS are not directly related to the immune system but can indirectly increase inflammation, mostly through the NF-κB pathway. Transactive response DNA-binding protein of 43 kDa (TDP-43) is observed to form plaques in sporadic ALS patients after phosphorylation in the cytoplasm [151]. The TDP-43 is a known activator of p65, a subunit of NF-κB that promotes pro-inflammatory cytokine release [152]. Another activator of NF-κB is the FUS protein, known to be mutated in familial ALS [153]. The FUS protein, together with SOD1 and C9orf72, which carry known ALS-associated mutations, are more detrimental, and induce a stronger inflammatory response when expressed in glia cells than in neuronal cells, pointing to an important role of resident macrophages in ALS neurodegeneration [154,155,156,157].
It is important to remember that oxidative stress can induce (sterile) inflammation or exacerbate inflammatory pathways at various levels (see Figure 2 for an overview). Hydrogen peroxide and peroxynitrite cause redox modifications in central pro-inflammatory molecules such as high mobility group box 1 (HMGB1) protein, inflammasome constituent NLRP3, NF-κB, and the process of NETosis (reviewed in [65]). Also, many of the up-stream regulators of these master switches of inflammation are redox-regulated as exemplified by reversible and irreversible sulfoxidation, S-nitros(yl)ation, S-glutathionylation, and protein tyrosine nitration of p38, p50, p65, S100A9, thioredoxin (Trx), IkB, and TXNIP (reviewed in [65,158]). Of note, NOX-2 activation, per se, is indispensable for the activation, recruitment, and infiltration of myelomonocytic cells [159,160] and T-cells [161]. In addition, the migration of monocytes/recruitment of macrophages is controlled by the cellular redox potential [162], and potential redox regulatory mechanisms involve redox modifications of MAPK phosphatase 1 [163] and Slingshot-1L-binding protein 14-3-3zeta [164] as well as changes in the S-glutathionylation pattern [165]. By this adverse redox signaling of ROS formed by damaged mitochondria or by activated phagocytic cells, inflammatory reactions may be initiated and aggravated. Conversely, inflammation causes cell activation, which is always associated with ROS formation by cellular ROS sources. Specifically, activated phagocytic cells infiltrating into the tissue can induce an oxidative burst by substantial superoxide formation from phagocytic NADPH oxidase. This crosstalk between oxidative stress and inflammation may create a vicious circle [65,158], leading to substantial tissue damage and neurodegeneration when happening in the brain.
Since microglia play a central role in neuroinflammation, it is important to consider them in the light of novel findings associating their function with neurodegenerative diseases [166]. Advances in single cell RNA sequencing revealed that the traditional M1/M2 polarization paradigm is not detailed enough to explain the intricate differences in specific microglia gene expressions associated with neurodegenerative diseases [167]. Microglia responding to Aβ have a distinct expression profile envisaged by upregulation of APOE, TREM2, GPNMB, and are CD163-positive [168,169,170]. A similar specific gene expression was found for phosphorylated tau protein, like GRID2 [168], and for α-synuclein, like IL1B, GPNMB, and HSP90AA1, in PD patients [171].
Astrocytes are part of the glymphatic system that is responsible for clearing protein plaques, such as Aβ [172], but they are also involved in neurodegenerative disease-induced neuroinflammation [173]. Similarly to microglia, astrocytes express neurodegenerative disease-associated genes that tend to promote inflammatory processes, like CD44, GFAP, and HSPB1, observed in PD, Huntington’s disease, and AD [171,174,175,176,177].

3. Effects of Noise and Air Pollution on Inflammation and Oxidative Stress in Neurodegenerative Disease—Mechanistic Insights from Animal Studies

The intricate link between oxidative stress and neuroinflammation can mostly be explained through microglia activation. Microglia as resident macrophages of cerebral tissues can be both, stimulated to produce ROS, and sense ROS as secondary messengers to promote pro-inflammatory behavior [178]. Many in vitro and in vivo studies have shown that air and noise pollution can affect neuroinflammation and oxidative stress, thereby potentially influencing the onset and progression of neurodegenerative diseases. In this chapter, we will focus on the preclinical research showing mechanistic links between neuroinflammation and oxidative stress in response to air and noise pollution.

3.1. Air Pollution

Air pollution is now an established risk factor in many diseases, including neurodegenerative diseases [4]. Many of the components of air pollution were shown to be associated with neuroinflammation and oxidative stress. Particulate matter (PM), the solid component of air pollution, induced neuroinflammation in mouse models [179,180,181]. In these studies, neuroinflammation was mostly observed through the increase in pro-inflammatory cytokines in the brain tissue, such as TNF-α, IL-1α/β, and IL-6, and the activation of NF-κB. Exposure of mice to ultrafine PM, with a diameter of less than 0.1 µm, also showed an increase in neuroinflammation through the upregulation of TNF-α, iNOS [182], cytokines/chemokines IL-1β, IL-6, TGF-β, CCL5, and CXCL1 in the hippocampus [183]. This upregulation of pro-inflammatory cytokines was accompanied by increased oxidative stress, envisaged by higher levels of 4-HNE and 3-NT-positive proteins. Exposure to PM was also associated with known pathomechanisms of neurodegenerative diseases, also including mitochondrial dysfunction and reactive oxygen species formation [7]. Studies in mice and rats showed that ambient PM exposure can increase tau phosphorylation [184,185], elevate beta-amyloid precursor protein (β-APP) and Aβ(42) oligomers [186,187], and increase plaque deposition [188,189]. PM exposure also resulted in cognitive decline, measured as learning impairment and a decrease in memory retention [190,191,192]. Ozone, another prominent gaseous component of air pollution, was observed to induce neuroinflammation and oxidative stress. A study in rats observed increased NF-κB and cytochrome c and COX-2 levels in substantia nigra, after ozone exposure, indicating potential implications for PD [193]. Another study from the same group also found a p53-dependent stress response and a decrease in dopaminergic neurons in the substantia nigra, together with an increase in circulating markers of lipid peroxidation [194]. Implications for AD were also found in an ex vivo study on rats where microglia showed proinflammatory activation, and Aβ(42) aggregation was increased in response to ozone exposure [195]. Aβ(42) accumulation was also observed in the mitochondria of the rat hippocampus after ozone exposure for 60 and 90 days [196].
In addition to exposure to individual air pollution components, some studies examined the influence of concentrated air or naturally occurring air pollution. Pioneering studies on dogs in Mexico showed that high amounts of air pollution increase levels of NF-κB and iNOS in multiple brain regions, together with disruption in blood–brain barrier, degradation of cortical neurons, and accumulation of neurofibrillary tangles, and Aβ(42) in the olfactory bulb and frontal cortex [197,198]. The authors also demonstrated that administration of the anti-inflammatory drug nimesulide improved 3-NT levels, pointing to reduced contribution from iNOS, whereas no changes in other parameters, including AD pathology, were observed [199]. In addition to concentrated ambient air pollution, exposure studies with diesel exhaust PM and full vehicle emissions showed similar microglia activation and an increase in neuroinflammation and oxidative stress in multiple brain regions [200,201,202]. Interestingly, there are some indications that alternative fuel emissions, like those from biodiesel, do not initiate neuroinflammation but rather activate microglia in a different manner, causing no neuronal damage [203], although the evidence provided by these studies is still preliminary. In animal models of neurodegenerative diseases, like AD [184,189,190,204] and PD [205,206], air pollution was shown to aggravate the progression and severity of the disease.
Air pollution has two potential pathways by which it can reach the neuronal system: a direct pathway where components of air pollution can reach brain tissue directly, and an indirect pathway where the influence of air pollution components (or associated toxins) in other organs (mostly lung as a first point of entry) gains a systemic character that finally reaches brain tissue [207]. In the direct pathway, air pollution-derived PM can enter the brain through the olfactory bulb [208], or ultrafine (nano-sized) PM can cross the air–blood barrier in the lung and enter circulation, later reaching the brain [209,210]. In our research, we have indirectly observed the translocation of ambient air pollution-derived PM into the circulation, as the exposure of mice resulted in an impairment of microvascular function and an increase in oxidative stress in cerebral and retinal arterioles [211]. In the indirect pathway, air pollution components cause oxidative stress and inflammation in the lung, which then spreads secondary messengers through pro-inflammatory cytokines and pro-oxidants to the systemic circulation, interrupting the blood–brain barrier integrity and inducing microglia activation in the brain [212,213,214]. Our observations align with these pathways as well, as many circulating cytokines were elevated in mice after PM exposure [211]. In addition to circulating cytokines, a non-cytokine-dependent pathway can result in microglia activation and neuroinflammation, most notably through the neurohormonal stress response by activation of the sympathetic nervous system and hypothalamus–pituitary–adrenal (HPA) axis [195,215]. An overview of the mechanisms by which air pollution affects neuroinflammation and oxidative stress is shown in Figure 3.

3.2. Transportation Noise

Traffic noise has only recently received the mechanistic background to better understand the epidemiological data on associations of noise with disease [70]. Like air pollution, noise also has two pathways by which it affects human health: the direct pathway, where loud noise causes inner ear damage and hearing loss as well as sleep deprivation, and the indirect pathway, where noise at lower sound pressure levels impairs daily activities, induces annoyance and disturbs sleep, causing elevated stress [216]. It is through the indirect pathway that most people are exposed to noise. This noise annoyance can trigger HPA axis signaling and sympathetic nervous system activation, leading to the release of glucocorticoids and catecholamines, dysregulation of the circadian rhythm, and increased inflammation and oxidative stress [74,217,218,219]. All of this can then initiate cerebral inflammation and promote the progression of neurodegenerative diseases [220]. An overview of the mechanisms by which noise affects neuroinflammation and oxidative stress is shown in Figure 4.
Fewer data exist on the effects of noise on neurodegenerative diseases than on the effects of air pollution. Nevertheless, some animal studies have shown that noise exposure is related to neuroinflammation, oxidative stress, and neurodegenerative disease pathology [221]. Neuronal inflammation was also shown in our studies in mice, where iNOS, CD68, and IL-6 were elevated after 4 days of noise exposure (mean sound pressure level of 72 dBA) [222]. This neuronal inflammation was accompanied by elevation in oxidative stress derived from phagocytic NADPH oxidase (NOX2), pointing to an immune system activation. Evidence for a systemic inflammation was observed as well [223]. Another well-established effect of noise exposure is the increase in tau phosphorylation, as observed in multiple studies [224,225,226]. Noise exposure was also shown to disturb the AMPK-mTOR pathway, impeding the autophagosome–lysosome fusion and resulting in autophagosome aggregation [226], another hallmark of AD pathology [227].
Studies in animal models of neurodegenerative disease also show a detrimental impact on disease progression. In a senescence-accelerated mouse prone 8 (SAMP8) model, the exposure to high sound pressure noise caused increased Aβ(42) accumulation and higher levels of tau phosphorylation [228]. This worsening of AD pathology was accompanied by upregulation of AD-associated genes such as Arc, Egr1, Egr2, Fos, Nauk1, and Per2. The impact of noise exposure on AD pathology was confirmed by other studies using the SAMP8 model [229,230]. A study conducted in the APP/PS1 Tg mouse model of AD showed reduced hippocampal tight junction protein levels. It increased circulating pro-inflammatory markers in the noise-exposed group [231]. In a study on gestational noise exposure, APPNL-G-F/NL-G-F mice showed an activation of the HPA axis, impairment in spatial learning, and Aβ(42) deposition [232]. Interestingly, the effects were more pronounced in female than male mice. In addition to the indirect pathway of noise-induced detrimental effects, a study applying noise with high sound pressure levels found associations of noise with an increase in c-Fos, c-Myc, and β-APP, pointing to the importance of inner ear damage and hearing loss in the progression of AD [233]. The importance of hearing loss for the propagation of AD-like pathophysiology was also confirmed by other noise exposure studies with high sound pressure levels [234,235,236].

4. Environmental Stressors and Neurodegenerative Disease—Evidence from Epidemiological Studies

Environmental stressors, such as transportation noise and air pollution, have previously been linked to neurodegenerative diseases [207,237,238]. Although the molecular mechanisms are not fully understood, the epidemiological data from both large and small cohorts provide insights into the association between environmental stressors and neurodegenerative diseases. Animal data on the relationship between the onset of neurodegenerative diseases and noise/PM exposure are mostly correlative in nature and large clinical cohorts or epidemiological studies are scarce. Therefore, the present topic is still under debate, and high-quality human research is urgently needed.

4.1. Air Pollution and Neurodegenerative Diseases

The available evidence regarding the potential association between air pollution and PD is constrained, and the outcomes lack a consistent consensus. A recent comprehensive meta-analysis, encompassing a total of 21 studies, investigated the connection between long-term exposure to air pollution, second-hand smoke, and the onset of PD. The analysis yielded predominantly non-significant findings with only marginal elevations in risk per 10 μg/m3 increments in PM2.5 (relative risk [RR] 1.08, 95% confidence interval [CI] 0.98–1.19), NO2 (RR 1.03, 95% CI 0.99–1.07), O3 (RR 1.01, 95% CI 1.00–1.02), and CO (RR 1.32, 95% CI 0.82–2.11) [239]. Strikingly, exposure to second-hand smoke was paradoxically associated with a significantly decreased risk of developing PD.
Consequently, another meta-analysis, encompassing 15 studies, revealed slightly elevated risks for the incidence of PD following long-term air pollution exposure, with RRs of 1.06 (95% CI 0.99–1.14) for PM2.5, 1.01 (95% CI 0.98–1.03) for NO2, 1.01 (95% CI 1.00–1.02) for O3, and 1.34 (95% CI 0.85–2.10) for CO. Furthermore, there was a RR 1.03 (95% CI 1.01–1.05) for hospital admission due to PD in response to short-term exposure to PM2.5, although there was considerable heterogeneity among the studies [240].
A more robust effect estimate for the risk of PD emerged from a meta-analysis involving 10 studies, with RRs of 1.06 (95% CI 1.04–1.09) for NOx, 1.65 (95% CI 1.10–2.48) for CO, 1.01 (95% CI 1.00–1.03) for NO2, and 1.01 (95% CI 1.00–1.02) for O3. However, it is important to note that this analysis was associated with a high risk of bias [241].
The potential role of PM2.5 as a determining factor in the development of dementia has been examined in two recent meta-analyses. In the first analysis, which incorporated data from four cohort studies conducted in Canada, Taiwan, the UK, and the US and included over 12 million elderly individuals aged 50 years or older, a noteworthy threefold increase in the risk of dementia was observed (hazard ratio [HR] 3.26, 95% CI 1.20–5.31) for each 10 μg/m3 increase in long-term PM2.5 exposure [242]. Subgroup analyses within this study further unveiled an almost fivefold elevated risk for AD (HR 4.82, 95% CI 2.28–7.36).
A comprehensive meta-analysis, encompassing 80 studies from 26 different countries, aimed to investigate the impact of PM2.5 on various cerebrovascular and neurological disorders [243]. The findings of this analysis revealed that long-term exposure to PM2.5 was associated with an increased overall risk of dementia (odds ratio [OR] 1.16, 95% CI 1.07–1.26), with a particular emphasis on AD (OR 3.26, 95% CI 0.84–12.74). Additionally, increased risks were observed for autism spectrum disorder (OR 1.68, 95% CI 1.20–2.34), PD (OR 1.34, 95% CI 1.04–1.73), and stroke.
Interestingly, recent investigations have highlighted the significant role of cardiovascular diseases in modifying and mediating the association between air pollutants and dementia risk [244,245]. In a prospective study from Sweden, it was observed that the presence or development of heart failure, ischemic heart disease, and stroke (which collectively explained 49.4% of air pollution-related dementia cases) appeared to amplify the link between long-term exposure to PM2.5 and NOx and the risk of dementia. This phenomenon is likely due to shared pathophysiological pathways through which air pollutants exert adverse effects on cardiovascular and neurological systems. The study’s authors concluded that as cardiovascular diseases accelerate cognitive decline and anticipate the onset of dementia, exposure to air pollution may indirectly exert detrimental effects on cognition, by pathways associated with cardiovascular disease, without the pollutants necessarily reaching the brain.
Substantiating this perspective, a prospective study from Italy demonstrated that the neurological effects of air pollution are notably associated with vascular damage. This was exemplified by positive long-term associations between NOx, NO2, PM2.5, and PM10 exposure and the risk of vascular dementia. In contrast, the relationships with AD and senile dementia were less evident [246]. Consistently with these findings, after accounting for potential confounding factors and other air pollutants, a Taiwanese study established associations between PM10, CO, and NO2 exposure and vascular dementia [247]. These relationships were further confirmed by additional studies conducted in Sweden and Canada, which indicated that exposure to multiple air pollutants may elevate the risk of vascular dementia and AD [248,249,250,251].
Notably, as traffic is a primary source of air pollution in urbanized areas and is also associated with increased noise pollution, research endeavors have explored the combined effects of air and noise pollution on the risk of dementia. However, these investigations did neither uncover any substantial evidence linking road traffic noise exposure with dementia risk, nor did they identify any significant interactions between noise and air pollutants that would modulate the risk of dementia [252,253].
In a cohort of 161,808 postmenopausal women aged 50–79 years at baseline, a nested case–control study of 256 ALS deaths and 2486 matched controls was performed [254]. Overall, there was only suggestive evidence for an association between PM10–2.5 and ALS deaths. More conclusive evidence was found in a case–control study involving 51 sporadic ALS cases and 51 matched controls [255]. It was demonstrated that residential exposure to aromatic solvents was associated with a higher risk of ALS among cases than controls (OR 5.03, 95% CI 1.29–19.53). Likewise, in a cohort from the Netherlands, the risk of ALS was higher in subjects with increased exposure to PM2.5 (OR 1.67, 95% CI 1.27–2.18), NO2 (OR 1.74, 95% CI 1.32–2.30), and NOx (OR 1.38, 95% CI 1.07–1.77) [256]. Further studies support the hypothesis of an association between air pollution and ALS risk [257,258,259,260].
There is not much quality literature on the association between multiple sclerosis and air pollution. A study from Iran found an association of PM10 and SO2 with higher risk of multiple sclerosis [261]. A study from Lombardy in northern Italy found a significant association between PM10 and hospital admissions for multiple sclerosis with an increase of 42% (RR 1.42, 95% CI 1.39–1.45) on the days preceded by one week with PM10 levels in the highest quartile [262]. Relapse in multiple sclerosis patients was also found to be associated with air pollution [263].

4.2. Transportation Noise and Neurodegenerative Diseases

In a study comprising 5227 participants aged 65 years or older from the Chicago Health and Aging Project, residential noise exposure was linked to a 36% higher likelihood of prevalent mild cognitive impairment (OR 1.36, 95% CI 1.15–1.62) and a 29% higher likelihood of AD (OR 1.29, 95% CI 1.08–1.55) for every 10 dB(A) increase in noise levels [264]. Noise exposure was also associated with lower global cognitive performance, particularly in perceptual speed, although it did not consistently lead to cognitive decline.
Similarly, data from the German Heinz Nixdorf Recall study indicated that exposure to traffic-related noise was linked to decreased global cognitive scores and an increase in mild cognitive impairment [265]. Notably, these associations were more pronounced in former and current smokers, suggesting that lifestyle risk factors may potentiate the adverse cognitive effects of noise exposure [266]. Importantly, the study also revealed that air pollution and road traffic noise exposure may interact synergistically to impact cognitive function negatively [267].
In another study involving 288 elderly women from the German SALIA study, road traffic noise exposure was associated with impaired total cognition and the constructional praxis domain, and these effects remained significant even after adjusting for air pollution exposure [268]. The Irish Longitudinal Study on Aging found that road traffic noise exposure hurts executive function [269]. Additionally, in a study involving 1612 elderly Mexican-American participants from Sacramento, there was suggestive evidence that traffic noise exposure increased the risk of dementia and cognitive impairment [270]. Interestingly, a subsequent study demonstrated that metabolic dysfunction, such as hyperglycemia or low HDL-cholesterol, could modify the influence of traffic-related air pollution and noise exposure on these outcomes [271].
Linares et al. reported a short-term association between traffic noise and dementia-related emergency hospital admissions risk in Spain [272]. In contrast, a study by Andersson et al. did not find any significant effect of road traffic noise exposure, independently or in conjunction with traffic-related air pollution, on the risk of dementia in a cohort of 1721 subjects [252]. In a larger study of 130,978 subjects in London, the relationship between night-time traffic noise exposure and the incidence of dementia became statistically insignificant when multiple air pollutants were considered in the analysis [253].
Likewise, in a large Canadian study involving approximately 678,000 individuals, road proximity and air pollution were positively associated with the risk of dementia and PD, while noise exposure showed no such relationship with these outcomes [273]. Most recently, a nationwide study in Denmark that included nearly 2 million adults aged 60 years or older examined the impact of road traffic and railway noise exposure on the incidence of dementia. The results indicated that both road traffic noise and railway noise exposure were associated with an increased risk of AD. Notably, only road traffic noise exposure, but not railway noise, was linked to a higher risk of vascular dementia [274]. Data from the Danish Nurse Cohort study also suggested an increased risk of death from dementia in response to road traffic noise (HR 1.12, 95% CI 0.90–1.38) [275].
There is a general lack of studies addressing other neurodegenerative diseases, such as multiple system atrophy, frontotemporal dementia, and Huntington’s disease. It is mostly the low prevalence of these diseases that does not provide enough patients for respective exposure–disease association studies or renders them underpowered. A good example of this is a study from UK that found a trend in frontotemporal dementia and air pollution exposure, however, without reaching significance due to the low number of patients [276]. In addition, the major genetic component of Huntington’s disease rather speaks against an association with environmental risk factors.

5. Conclusions

Environmental stressors such as noise and air pollution gain more and more attention in the modern urbanized and industrialized world. The fact that the human population is aging rapidly due to better healthcare, nutrition, and sanitation and that neurodegenerative diseases are considered ailments of the elderly, warrants a better understanding of the impact of environmental stressors on neurodegenerative diseases. This review has provided an overview of the most common neurodegenerative diseases and their pathomechanisms. We also highlighted epidemiological evidence for an association of noise and air pollution exposure to neurodegenerative diseases and mechanistic data connecting neurodegenerative pathophysiology to noise and air pollution exposure through neuroinflammation and oxidative stress. Importantly, inflammation and oxidative stress can promote each other in a vicious cycle (Figure 2), further progressing neurodegenerative diseases. More high-quality research is urgently needed, as more mechanistic and epidemiological data could drive better treatment and preventive strategies. Finally, more and better mechanistic insight and clinical evidence would also force better legislative interventions by implementing stricter limits for noise levels and air pollution concentrations. So far, the mitigation of adverse health effects by noise is mostly based on lowering the exposure levels, e.g., by speed limits, noise-isolating windows, lower noise-emitting car engines and tires, train breaks, and optimized aircraft starting/landing protocols. With respect to air pollution, lowering the legal limits is the preferred strategy, e.g., by better filtration systems for combustion emissions, traffic bans in city centers, and the regulation of home heating strategies. In addition, personal measures are popular such as face masks and household air filter systems. A detailed list of mitigation measures against noise and air pollution can be found in two recent review articles [1,2].

Author Contributions

All authors were involved in conceptualization, writing—original draft preparation, review and editing, and visualization. All authors have read and agreed to the published version of the manuscript.

Funding

We gratefully acknowledge financial support through vascular biology research grants from the Foundation Heart of Mainz (to all authors). T.M. is a principal investigator and M.K., O.H., and A.D. are (Young) Scientists of the DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany. Continuous structural funding by the DZHK is highly appreciated. The research of this study was also supported by European COST Action CA20121: Bench to bedside transition for pharmacological regulation of NRF2 in non-communicable diseases (BenBedPhar). Webpage: https://benbedphar.org/about-benbedphar/ (last accessed on 13 February 2024). The work was also supported by the environmental network EXPOHEALTH funded by the state Rhineland-Palatinate, Germany.

Conflicts of Interest

The authors declare no conflicts of interest with the contents of this review.

References

  1. Hahad, O.; Rajagopalan, S.; Lelieveld, J.; Sorensen, M.; Frenis, K.; Daiber, A.; Basner, M.; Nieuwenhuijsen, M.; Brook, R.D.; Munzel, T. Noise and Air Pollution as Risk Factors for Hypertension: Part I-Epidemiology. Hypertension 2023, 80, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  2. Hahad, O.; Rajagopalan, S.; Lelieveld, J.; Sorensen, M.; Kuntic, M.; Daiber, A.; Basner, M.; Nieuwenhuijsen, M.; Brook, R.D.; Munzel, T. Noise and Air Pollution as Risk Factors for Hypertension: Part II-Pathophysiologic Insight. Hypertension 2023, 80, 1384–1392. [Google Scholar] [CrossRef]
  3. Munzel, T.; Gori, T.; Al-Kindi, S.; Deanfield, J.; Lelieveld, J.; Daiber, A.; Rajagopalan, S. Effects of gaseous and solid constituents of air pollution on endothelial function. Eur. Heart J. 2018, 39, 3543–3550. [Google Scholar] [CrossRef] [PubMed]
  4. Diseases, G.B.D.; Injuries, C. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef]
  5. Lelieveld, J.; Klingmuller, K.; Pozzer, A.; Poschl, U.; Fnais, M.; Daiber, A.; Munzel, T. Cardiovascular disease burden from ambient air pollution in Europe reassessed using novel hazard ratio functions. Eur. Heart J. 2019, 40, 1590–1596. [Google Scholar] [CrossRef]
  6. Vohra, K.; Vodonos, A.; Schwartz, J.; Marais, E.A.; Sulprizio, M.P.; Mickley, L.J. Global mortality from outdoor fine particle pollution generated by fossil fuel combustion: Results from GEOS-Chem. Environ. Res. 2021, 195, 110754. [Google Scholar] [CrossRef] [PubMed]
  7. Daiber, A.; Kuntic, M.; Hahad, O.; Delogu, L.G.; Rohrbach, S.; Di Lisa, F.; Schulz, R.; Munzel, T. Effects of air pollution particles (ultrafine and fine particulate matter) on mitochondrial function and oxidative stress—Implications for cardiovascular and neurodegenerative diseases. Arch. Biochem. Biophys. 2020, 696, 108662. [Google Scholar] [CrossRef] [PubMed]
  8. Pinkerton, K.E.; Green, F.H.; Saiki, C.; Vallyathan, V.; Plopper, C.G.; Gopal, V.; Hung, D.; Bahne, E.B.; Lin, S.S.; Menache, M.G.; et al. Distribution of particulate matter and tissue remodeling in the human lung. Environ. Health Perspect. 2000, 108, 1063–1069. [Google Scholar] [CrossRef]
  9. Kreyling, W.G.; Semmler-Behnke, M.; Moller, W. Ultrafine particle-lung interactions: Does size matter? J. Aerosol. Med. 2006, 19, 74–83. [Google Scholar] [CrossRef]
  10. Steven, S.; Oelze, M.; Hausding, M.; Roohani, S.; Kashani, F.; Kroller-Schon, S.; Helmstadter, J.; Jansen, T.; Baum, C.; Iglarz, M.; et al. The Endothelin Receptor Antagonist Macitentan Improves Isosorbide-5-Mononitrate (ISMN) and Isosorbide Dinitrate (ISDN) Induced Endothelial Dysfunction, Oxidative Stress, and Vascular Inflammation. Oxid. Med. Cell Longev. 2018, 2018, 7845629. [Google Scholar] [CrossRef]
  11. World Health Organization. WHO—Noise. Available online: https://www.who.int/europe/news-room/fact-sheets/item/noise (accessed on 13 February 2024).
  12. Collaborators, G.B.D.R.F. Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390, 1345–1422. [Google Scholar] [CrossRef]
  13. Wild, C.P. Complementing the genome with an “exposome”: The outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1847–1850. [Google Scholar] [CrossRef]
  14. Sainani, K. Taking on the exposome—Bringing bioinformatics tools to the environmental side of the health equation. Biomed. Comput. Rev. 2016, 2016, 14–21. [Google Scholar]
  15. Vrijheid, M. The exposome: A new paradigm to study the impact of environment on health. Thorax 2014, 69, 876–878. [Google Scholar] [CrossRef]
  16. Hahad, O.; Al-Kindi, S.; Lelieveld, J.; Munzel, T.; Daiber, A. Supporting and implementing the beneficial parts of the exposome: The environment can be the problem, but it can also be the solution. Int. J. Hydrogen Environ. Health 2024, 255, 114290. [Google Scholar] [CrossRef]
  17. Olden, K.; Wilson, S. Environmental health and genomics: Visions and implications. Nat. Rev. Genet. 2000, 1, 149–153. [Google Scholar] [CrossRef]
  18. Munzel, T.; Sorensen, M.; Lelieveld, J.; Hahad, O.; Al-Kindi, S.; Nieuwenhuijsen, M.; Giles-Corti, B.; Daiber, A.; Rajagopalan, S. Heart healthy cities: Genetics loads the gun but the environment pulls the trigger. Eur. Heart J. 2021, 42, 2422–2438. [Google Scholar] [CrossRef] [PubMed]
  19. Munzel, T.; Sorensen, M.; Hahad, O.; Nieuwenhuijsen, M.; Daiber, A. The contribution of the exposome to the burden of cardiovascular disease. Nat. Rev. Cardiol. 2023, 20, 651–669. [Google Scholar] [CrossRef] [PubMed]
  20. Collaborators, G.B.D.C.o.D. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef]
  21. Assoc, A.s. 2018 Alzheimer’s Disease Facts and Figures (vol 14, pg 367, 2018). Alzheimers Dement. 2018, 14, 701. [Google Scholar] [CrossRef]
  22. Hahad, O.; Frenis, K.; Kuntic, M.; Daiber, A.; Munzel, T. Accelerated Aging and Age-Related Diseases (CVD and Neurological) Due to Air Pollution and Traffic Noise Exposure. Int. J. Mol. Sci. 2021, 22, 2419. [Google Scholar] [CrossRef]
  23. Collaborators, G.B.D.D. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88–106. [Google Scholar] [CrossRef]
  24. Prince, M.; Wimo, A.; Guerchet, M.; Ali, G.-C.; Wu, Y.-T.; Prina, M. World Alzheimer Report 2015—The Global Impact of Dementia. Doctoral Dissertation, Alzheimer’s Disease International, London, UK, 2015. [Google Scholar]
  25. Rasmussen, J.; Langerman, H. Alzheimer’s Disease—Why We Need Early Diagnosis. Degener. Neurol. Neuromuscul. Dis. 2019, 9, 123–130. [Google Scholar] [CrossRef]
  26. Alzheimer Europe. Dementia in Europe Yearbook 2019: Estimating the Prevalence of Dementia in Europe. Available online: https://www.alzheimer-europe.org/sites/default/files/alzheimer_europe_dementia_in_europe_yearbook_2019.pdf (accessed on 13 February 2024).
  27. Niedzielska, E.; Smaga, I.; Gawlik, M.; Moniczewski, A.; Stankowicz, P.; Pera, J.; Filip, M. Oxidative Stress in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 4094–4125. [Google Scholar] [CrossRef]
  28. Peter-Derex, L.; Yammine, P.; Bastuji, H.; Croisile, B. Sleep and Alzheimer’s disease. Sleep Med. Rev. 2015, 19, 29–38. [Google Scholar] [CrossRef]
  29. Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chetelat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
  30. Wolfson, C.; Wolfson, D.B.; Asgharian, M.; M’Lan, C.E.; Ostbye, T.; Rockwood, K.; Hogan, D.B.; Clinical Progression of Dementia Study, G. A reevaluation of the duration of survival after the onset of dementia. N. Engl. J. Med. 2001, 344, 1111–1116. [Google Scholar] [CrossRef]
  31. Reisberg, B.; Ferris, S.H.; de Leon, M.J.; Crook, T. The Global Deterioration Scale for assessment of primary degenerative dementia. Am. J. Psychiatry 1982, 139, 1136–1139. [Google Scholar] [CrossRef] [PubMed]
  32. Perl, D.P. Neuropathology of Alzheimer’s disease. Mt. Sinai. J. Med. 2010, 77, 32–42. [Google Scholar] [CrossRef] [PubMed]
  33. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed]
  34. Tiiman, A.; Jelic, V.; Jarvet, J.; Jaremo, P.; Bogdanovic, N.; Rigler, R.; Terenius, L.; Graslund, A.; Vukojevic, V. Amyloidogenic Nanoplaques in Blood Serum of Patients with Alzheimer’s Disease Revealed by Time-Resolved Thioflavin T Fluorescence Intensity Fluctuation Analysis. J. Alzheimers. Dis. 2019, 68, 571–582. [Google Scholar] [CrossRef] [PubMed]
  35. Kumar, A.; Singh, A.; Ekavali. A review on Alzheimer’s disease pathophysiology and its management: An update. Pharmacol. Rep. 2015, 67, 195–203. [Google Scholar] [CrossRef]
  36. Dal Pra, I.; Chiarini, A.; Gui, L.; Chakravarthy, B.; Pacchiana, R.; Gardenal, E.; Whitfield, J.F.; Armato, U. Do astrocytes collaborate with neurons in spreading the “infectious” abeta and Tau drivers of Alzheimer’s disease? Neuroscientist 2015, 21, 9–29. [Google Scholar] [CrossRef] [PubMed]
  37. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef] [PubMed]
  38. Parkinson’s Foundation. Who Has Parkinson’s? Available online: https://www.parkinson.org/understanding-parkinsons/statistics (accessed on 13 February 2024).
  39. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  40. Nalls, M.A.; Blauwendraat, C.; Vallerga, C.L.; Heilbron, K.; Bandres-Ciga, S.; Chang, D.; Tan, M.; Kia, D.A.; Noyce, A.J.; Xue, A.; et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: A meta-analysis of genome-wide association studies. Lancet Neurol. 2019, 18, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  41. Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell 2019, 18, e13031. [Google Scholar] [CrossRef] [PubMed]
  42. Goedert, M.; Spillantini, M.G.; Del Tredici, K.; Braak, H. 100 years of Lewy pathology. Nat. Rev. Neurol. 2013, 9, 13–24. [Google Scholar] [CrossRef]
  43. Twohig, D.; Nielsen, H.M. alpha-synuclein in the pathophysiology of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 23. [Google Scholar] [CrossRef]
  44. Shahmoradian, S.H.; Lewis, A.J.; Genoud, C.; Hench, J.; Moors, T.E.; Navarro, P.P.; Castano-Diez, D.; Schweighauser, G.; Graff-Meyer, A.; Goldie, K.N.; et al. Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes. Nat. Neurosci. 2019, 22, 1099–1109. [Google Scholar] [CrossRef]
  45. National Insitute on Aging. Parkinson’s Disease: Causes, Symptoms, and Treatments. Available online: https://www.nia.nih.gov/health/parkinsons-disease (accessed on 13 February 2024).
  46. Hardiman, O.; Al-Chalabi, A.; Chio, A.; Corr, E.M.; Logroscino, G.; Robberecht, W.; Shaw, P.J.; Simmons, Z.; van den Berg, L.H. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 2017, 3, 17071. [Google Scholar] [CrossRef]
  47. Parakh, S.; Spencer, D.M.; Halloran, M.A.; Soo, K.Y.; Atkin, J.D. Redox regulation in amyotrophic lateral sclerosis. Oxid. Med. Cell Longev. 2013, 2013, 408681. [Google Scholar] [CrossRef]
  48. Logroscino, G.; Urso, D.; Tortelli, R. The challenge of amyotrophic lateral sclerosis descriptive epidemiology: To estimate low incidence rates across complex phenotypes in different geographic areas. Curr. Opin. Neurol. 2022, 35, 678–685. [Google Scholar] [CrossRef]
  49. Hardiman, O.; van den Berg, L.H.; Kiernan, M.C. Clinical diagnosis and management of amyotrophic lateral sclerosis. Nat. Rev. Neurol. 2011, 7, 639–649. [Google Scholar] [CrossRef]
  50. Suzuki, N.; Nishiyama, A.; Warita, H.; Aoki, M. Genetics of amyotrophic lateral sclerosis: Seeking therapeutic targets in the era of gene therapy. J. Hum. Genet. 2023, 68, 131–152. [Google Scholar] [CrossRef]
  51. Gibson, S.B.; Downie, J.M.; Tsetsou, S.; Feusier, J.E.; Figueroa, K.P.; Bromberg, M.B.; Jorde, L.B.; Pulst, S.M. The evolving genetic risk for sporadic ALS. Neurology 2017, 89, 226–233. [Google Scholar] [CrossRef] [PubMed]
  52. Beckman, J.S.; Carson, M.; Smith, C.D.; Koppenol, W.H. ALS, SOD and peroxynitrite. Nature 1993, 364, 584. [Google Scholar] [CrossRef] [PubMed]
  53. Goutman, S.A.; Hardiman, O.; Al-Chalabi, A.; Chio, A.; Savelieff, M.G.; Kiernan, M.C.; Feldman, E.L. Emerging insights into the complex genetics and pathophysiology of amyotrophic lateral sclerosis. Lancet Neurol. 2022, 21, 465–479. [Google Scholar] [CrossRef]
  54. Blokhuis, A.M.; Groen, E.J.; Koppers, M.; van den Berg, L.H.; Pasterkamp, R.J. Protein aggregation in amyotrophic lateral sclerosis. Acta Neuropathol. 2013, 125, 777–794. [Google Scholar] [CrossRef]
  55. Peng, C.; Trojanowski, J.Q.; Lee, V.M. Protein transmission in neurodegenerative disease. Nat. Rev. Neurol. 2020, 16, 199–212. [Google Scholar] [CrossRef] [PubMed]
  56. Voet, S.; Srinivasan, S.; Lamkanfi, M.; van Loo, G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol. Med. 2019, 11, 248. [Google Scholar] [CrossRef]
  57. Michalska, P.; Leon, R. When It Comes to an End: Oxidative Stress Crosstalk with Protein Aggregation and Neuroinflammation Induce Neurodegeneration. Antioxidants 2020, 9, 740. [Google Scholar] [CrossRef]
  58. Katsumoto, A.; Takeuchi, H.; Takahashi, K.; Tanaka, F. Microglia in Alzheimer’s Disease: Risk Factors and Inflammation. Front. Neurol. 2018, 9, 978. [Google Scholar] [CrossRef] [PubMed]
  59. Ischiropoulos, H.; Beckman, J.S. Oxidative stress and nitration in neurodegeneration: Cause, effect, or association? J. Clin. Investig. 2003, 111, 163–169. [Google Scholar] [CrossRef]
  60. Tonnies, E.; Trushina, E. Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef]
  61. Steven, S.; Frenis, K.; Oelze, M.; Kalinovic, S.; Kuntic, M.; Bayo Jimenez, M.T.; Vujacic-Mirski, K.; Helmstadter, J.; Kroller-Schon, S.; Munzel, T.; et al. Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. Oxid. Med. Cell Longev. 2019, 2019, 7092151. [Google Scholar] [CrossRef]
  62. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  63. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef] [PubMed]
  64. Daiber, A.; Hahad, O.; Andreadou, I.; Steven, S.; Daub, S.; Munzel, T. Redox-related biomarkers in human cardiovascular disease—classical footprints and beyond. Redox Biol. 2021, 42, 101875. [Google Scholar] [CrossRef] [PubMed]
  65. Wenzel, P.; Kossmann, S.; Munzel, T.; Daiber, A. Redox regulation of cardiovascular inflammation—Immunomodulatory function of mitochondrial and Nox-derived reactive oxygen and nitrogen species. Free Radic. Biol. Med. 2017, 109, 48–60. [Google Scholar] [CrossRef] [PubMed]
  66. Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
  67. Daiber, A.; Steven, S.; Vujacic-Mirski, K.; Kalinovic, S.; Oelze, M.; Di Lisa, F.; Munzel, T. Regulation of Vascular Function and Inflammation via Cross Talk of Reactive Oxygen and Nitrogen Species from Mitochondria or NADPH Oxidase-Implications for Diabetes Progression. Int. J. Mol. Sci. 2020, 21, 3405. [Google Scholar] [CrossRef] [PubMed]
  68. Ahlskog, J.E. Challenging conventional wisdom: The etiologic role of dopamine oxidative stress in Parkinson’s disease. Mov. Disord. 2005, 20, 271–282. [Google Scholar] [CrossRef] [PubMed]
  69. Bader, V.; Winklhofer, K.F. Mitochondria at the interface between neurodegeneration and neuroinflammation. Semin. Cell Dev. Biol. 2020, 99, 163–171. [Google Scholar] [CrossRef] [PubMed]
  70. Sorensen, M.; Pershagen, G.; Thacher, J.D.; Lanki, T.; Wicki, B.; Roosli, M.; Vienneau, D.; Cantuaria, M.L.; Schmidt, J.H.; Aasvang, G.M.; et al. Health position paper and redox perspectives—Disease burden by transportation noise. Redox Biol. 2024, 69, 102995. [Google Scholar] [CrossRef]
  71. Rink, C.; Khanna, S. Significance of brain tissue oxygenation and the arachidonic acid cascade in stroke. Antioxid. Redox Signal. 2011, 14, 1889–1903. [Google Scholar] [CrossRef] [PubMed]
  72. Hallermann, S.; de Kock, C.P.; Stuart, G.J.; Kole, M.H. State and location dependence of action potential metabolic cost in cortical pyramidal neurons. Nat. Neurosci. 2012, 15, 1007–1014. [Google Scholar] [CrossRef] [PubMed]
  73. Popa-Wagner, A.; Mitran, S.; Sivanesan, S.; Chang, E.; Buga, A.M. ROS and brain diseases: The good, the bad, and the ugly. Oxid. Med. Cell Longev. 2013, 2013, 963520. [Google Scholar] [CrossRef] [PubMed]
  74. Daiber, A.; Kroller-Schon, S.; Oelze, M.; Hahad, O.; Li, H.; Schulz, R.; Steven, S.; Munzel, T. Oxidative stress and inflammation contribute to traffic noise-induced vascular and cerebral dysfunction via uncoupling of nitric oxide synthases. Redox Biol. 2020, 34, 101506. [Google Scholar] [CrossRef]
  75. Simic, G.; Lucassen, P.J.; Krsnik, Z.; Kruslin, B.; Kostovic, I.; Winblad, B.; Bogdanovi. nNOS expression in reactive astrocytes correlates with increased cell death related DNA damage in the hippocampus and entorhinal cortex in Alzheimer’s disease. Exp. Neurol. 2000, 165, 12–26. [Google Scholar] [CrossRef]
  76. Saha, R.N.; Pahan, K. Signals for the induction of nitric oxide synthase in astrocytes. Neurochem. Int. 2006, 49, 154–163. [Google Scholar] [CrossRef] [PubMed]
  77. Adibhatla, R.M.; Hatcher, J.F. Lipid oxidation and peroxidation in CNS health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2010, 12, 125–169. [Google Scholar] [CrossRef] [PubMed]
  78. Martins, R.N.; Harper, C.G.; Stokes, G.B.; Masters, C.L. Increased cerebral glucose-6-phosphate dehydrogenase activity in Alzheimer’s disease may reflect oxidative stress. J. Neurochem. 1986, 46, 1042–1045. [Google Scholar] [CrossRef] [PubMed]
  79. Sultana, R.; Butterfield, D.A. Role of oxidative stress in the progression of Alzheimer’s disease. J. Alzheimers Dis. 2010, 19, 341–353. [Google Scholar] [CrossRef] [PubMed]
  80. Ringman, J.M.; Frautschy, S.A.; Teng, E.; Begum, A.N.; Bardens, J.; Beigi, M.; Gylys, K.H.; Badmaev, V.; Heath, D.D.; Apostolova, L.G.; et al. Oral curcumin for Alzheimer’s disease: Tolerability and efficacy in a 24-week randomized, double blind, placebo-controlled study. Alzheimers Res. Ther. 2012, 4, 43. [Google Scholar] [CrossRef]
  81. Sutherland, G.T.; Chami, B.; Youssef, P.; Witting, P.K. Oxidative stress in Alzheimer’s disease: Primary villain or physiological by-product? Redox. Rep. 2013, 18, 134–141. [Google Scholar] [CrossRef]
  82. Ansari, M.A.; Scheff, S.W. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J. Neuropathol. Exp. Neurol. 2010, 69, 155–167. [Google Scholar] [CrossRef]
  83. Markesbery, W.R.; Lovell, M.A. DNA oxidation in Alzheimer’s disease. Antioxid. Redox Signal. 2006, 8, 2039–2045. [Google Scholar] [CrossRef]
  84. Luth, H.J.; Munch, G.; Arendt, T. Aberrant expression of NOS isoforms in Alzheimer’s disease is structurally related to nitrotyrosine formation. Brain Res. 2002, 953, 135–143. [Google Scholar] [CrossRef]
  85. Luth, H.J.; Holzer, M.; Gartner, U.; Staufenbiel, M.; Arendt, T. Expression of endothelial and inducible NOS-isoforms is increased in Alzheimer’s disease, in APP23 transgenic mice and after experimental brain lesion in rat: Evidence for an induction by amyloid pathology. Brain Res. 2001, 913, 57–67. [Google Scholar] [CrossRef] [PubMed]
  86. Massaad, C.A. Neuronal and vascular oxidative stress in Alzheimer’s disease. Curr. Neuropharmacol. 2011, 9, 662–673. [Google Scholar] [CrossRef]
  87. Casado, A.; Encarnacion Lopez-Fernandez, M.; Concepcion Casado, M.; de La Torre, R. Lipid peroxidation and antioxidant enzyme activities in vascular and Alzheimer dementias. Neurochem. Res. 2008, 33, 450–458. [Google Scholar] [CrossRef]
  88. Marcus, D.L.; Thomas, C.; Rodriguez, C.; Simberkoff, K.; Tsai, J.S.; Strafaci, J.A.; Freedman, M.L. Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer’s disease. Exp. Neurol. 1998, 150, 40–44. [Google Scholar] [CrossRef]
  89. Sultana, R.; Piroddi, M.; Galli, F.; Butterfield, D.A. Protein levels and activity of some antioxidant enzymes in hippocampus of subjects with amnestic mild cognitive impairment. Neurochem. Res. 2008, 33, 2540–2546. [Google Scholar] [CrossRef] [PubMed]
  90. Ferreira, M.E.; de Vasconcelos, A.S.; da Costa Vilhena, T.; da Silva, T.L.; da Silva Barbosa, A.; Gomes, A.R.; Dolabela, M.F.; Percario, S. Oxidative Stress in Alzheimer’s Disease: Should We Keep Trying Antioxidant Therapies? Cell Mol. Neurobiol. 2015, 35, 595–614. [Google Scholar] [CrossRef] [PubMed]
  91. Dexter, D.T.; Carter, C.J.; Wells, F.R.; Javoy-Agid, F.; Agid, Y.; Lees, A.; Jenner, P.; Marsden, C.D. Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease. J. Neurochem. 1989, 52, 381–389. [Google Scholar] [CrossRef] [PubMed]
  92. Mythri, R.B.; Venkateshappa, C.; Harish, G.; Mahadevan, A.; Muthane, U.B.; Yasha, T.C.; Srinivas Bharath, M.M.; Shankar, S.K. Evaluation of markers of oxidative stress, antioxidant function and astrocytic proliferation in the striatum and frontal cortex of Parkinson’s disease brains. Neurochem. Res. 2011, 36, 1452–1463. [Google Scholar] [CrossRef] [PubMed]
  93. Castellani, R.J.; Perry, G.; Siedlak, S.L.; Nunomura, A.; Shimohama, S.; Zhang, J.; Montine, T.; Sayre, L.M.; Smith, M.A. Hydroxynonenal adducts indicate a role for lipid peroxidation in neocortical and brainstem Lewy bodies in humans. Neurosci. Lett. 2002, 319, 25–28. [Google Scholar] [CrossRef] [PubMed]
  94. Yoritaka, A.; Hattori, N.; Uchida, K.; Tanaka, M.; Stadtman, E.R.; Mizuno, Y. Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. Proc. Natl. Acad. Sci. USA 1996, 93, 2696–2701. [Google Scholar] [CrossRef]
  95. Shamoto-Nagai, M.; Maruyama, W.; Hashizume, Y.; Yoshida, M.; Osawa, T.; Riederer, P.; Naoi, M. In parkinsonian substantia nigra, alpha-synuclein is modified by acrolein, a lipid-peroxidation product, and accumulates in the dopamine neurons with inhibition of proteasome activity. J. Neural. Transm. 2007, 114, 1559–1567. [Google Scholar] [CrossRef] [PubMed]
  96. Andersen, C.; Gronnemose, A.L.; Pedersen, J.N.; Nowak, J.S.; Christiansen, G.; Nielsen, J.; Mulder, F.A.A.; Otzen, D.E.; Jorgensen, T.J.D. Lipid Peroxidation Products HNE and ONE Promote and Stabilize Alpha-Synuclein Oligomers by Chemical Modifications. Biochemistry 2021, 60, 3644–3658. [Google Scholar] [CrossRef]
  97. Schildknecht, S.; Gerding, H.R.; Karreman, C.; Drescher, M.; Lashuel, H.A.; Outeiro, T.F.; Di Monte, D.A.; Leist, M. Oxidative and nitrative alpha-synuclein modifications and proteostatic stress: Implications for disease mechanisms and interventions in synucleinopathies. J. Neurochem. 2013, 125, 491–511. [Google Scholar] [CrossRef]
  98. Przedborski, S.; Chen, Q.; Vila, M.; Giasson, B.I.; Djaldatti, R.; Vukosavic, S.; Souza, J.M.; Jackson-Lewis, V.; Lee, V.M.; Ischiropoulos, H. Oxidative post-translational modifications of alpha-synuclein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson’s disease. J. Neurochem. 2001, 76, 637–640. [Google Scholar] [CrossRef]
  99. Zhang, J.; Perry, G.; Smith, M.A.; Robertson, D.; Olson, S.J.; Graham, D.G.; Montine, T.J. Parkinson’s disease is associated with oxidative damage to cytoplasmic DNA and RNA in substantia nigra neurons. Am. J. Pathol. 1999, 154, 1423–1429. [Google Scholar] [CrossRef]
  100. Alam, Z.I.; Daniel, S.E.; Lees, A.J.; Marsden, D.C.; Jenner, P.; Halliwell, B. A generalised increase in protein carbonyls in the brain in Parkinson’s but not incidental Lewy body disease. J. Neurochem. 1997, 69, 1326–1329. [Google Scholar] [CrossRef] [PubMed]
  101. Ogata, M.; Kaneya, D.; Shin-Ya, K.; Li, L.; Abe, Y.; Katoh, H.; Seki, S.; Seki, Y.; Gonda, R.; Urano, S.; et al. Trapping effect of eugenol on hydroxyl radicals induced by L-DOPA in vitro. Chem. Pharm. Bull 2005, 53, 1167–1170. [Google Scholar] [CrossRef] [PubMed]
  102. Group, P.D.M.C.; Gray, R.; Ives, N.; Rick, C.; Patel, S.; Gray, A.; Jenkinson, C.; McIntosh, E.; Wheatley, K.; Williams, A.; et al. Long-term effectiveness of dopamine agonists and monoamine oxidase B inhibitors compared with levodopa as initial treatment for Parkinson’s disease (PD MED): A large, open-label, pragmatic randomised trial. Lancet 2014, 384, 1196–1205. [Google Scholar] [CrossRef]
  103. Zhou, Z.D.; Yi, L.X.; Wang, D.Q.; Lim, T.M.; Tan, E.K. Role of dopamine in the pathophysiology of Parkinson’s disease. Transl. Neurodegener. 2023, 12, 44. [Google Scholar] [CrossRef]
  104. Zhou, Z.D.; Lim, T.M. Roles of glutathione (GSH) in dopamine (DA) oxidation studied by improved tandem HPLC plus ESI-MS. Neurochem. Res. 2009, 34, 316–326. [Google Scholar] [CrossRef]
  105. Sian, J.; Dexter, D.T.; Lees, A.J.; Daniel, S.; Agid, Y.; Javoy-Agid, F.; Jenner, P.; Marsden, C.D. Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia. Ann. Neurol. 1994, 36, 348–355. [Google Scholar] [CrossRef] [PubMed]
  106. Marttila, R.J.; Lorentz, H.; Rinne, U.K. Oxygen toxicity protecting enzymes in Parkinson’s disease. Increase of superoxide dismutase-like activity in the substantia nigra and basal nucleus. J. Neurol. Sci. 1988, 86, 321–331. [Google Scholar] [CrossRef]
  107. Grunewald, A.; Kumar, K.R.; Sue, C.M. New insights into the complex role of mitochondria in Parkinson’s disease. Prog. Neurobiol. 2019, 177, 73–93. [Google Scholar] [CrossRef] [PubMed]
  108. Haas, R.H.; Nasirian, F.; Nakano, K.; Ward, D.; Pay, M.; Hill, R.; Shults, C.W. Low platelet mitochondrial complex I and complex II/III activity in early untreated Parkinson’s disease. Ann. Neurol. 1995, 37, 714–722. [Google Scholar] [CrossRef] [PubMed]
  109. Richardson, J.R.; Caudle, W.M.; Guillot, T.S.; Watson, J.L.; Nakamaru-Ogiso, E.; Seo, B.B.; Sherer, T.B.; Greenamyre, J.T.; Yagi, T.; Matsuno-Yagi, A.; et al. Obligatory role for complex I inhibition in the dopaminergic neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Toxicol. Sci. 2007, 95, 196–204. [Google Scholar] [CrossRef] [PubMed]
  110. Shults, C.W.; Oakes, D.; Kieburtz, K.; Beal, M.F.; Haas, R.; Plumb, S.; Juncos, J.L.; Nutt, J.; Shoulson, I.; Carter, J.; et al. Effects of coenzyme Q10 in early Parkinson disease: Evidence of slowing of the functional decline. Arch. Neurol. 2002, 59, 1541–1550. [Google Scholar] [CrossRef] [PubMed]
  111. Zhu, Z.G.; Sun, M.X.; Zhang, W.L.; Wang, W.W.; Jin, Y.M.; Xie, C.L. The efficacy and safety of coenzyme Q10 in Parkinson’s disease: A meta-analysis of randomized controlled trials. Neurol. Sci. 2017, 38, 215–224. [Google Scholar] [CrossRef] [PubMed]
  112. Pasinelli, P.; Belford, M.E.; Lennon, N.; Bacskai, B.J.; Hyman, B.T.; Trotti, D.; Brown, R.H., Jr. Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind and aggregate with Bcl-2 in spinal cord mitochondria. Neuron 2004, 43, 19–30. [Google Scholar] [CrossRef]
  113. Forsberg, K.; Jonsson, P.A.; Andersen, P.M.; Bergemalm, D.; Graffmo, K.S.; Hultdin, M.; Jacobsson, J.; Rosquist, R.; Marklund, S.L.; Brannstrom, T. Novel antibodies reveal inclusions containing non-native SOD1 in sporadic ALS patients. PLoS ONE 2010, 5, e11552. [Google Scholar] [CrossRef]
  114. Tian, Y.P.; Che, F.Y.; Su, Q.P.; Lu, Y.C.; You, C.P.; Huang, L.M.; Wang, S.G.; Wang, L.; Yu, J.X. Effects of mutant TDP-43 on the Nrf2/ARE pathway and protein expression of MafK and JDP2 in NSC-34 cells. Genet. Mol. Res. 2017, 16, gmr16029638. [Google Scholar] [CrossRef]
  115. Duan, W.; Li, X.; Shi, J.; Guo, Y.; Li, Z.; Li, C. Mutant TAR DNA-binding protein-43 induces oxidative injury in motor neuron-like cell. Neuroscience 2010, 169, 1621–1629. [Google Scholar] [CrossRef]
  116. Andrade, N.S.; Ramic, M.; Esanov, R.; Liu, W.; Rybin, M.J.; Gaidosh, G.; Abdallah, A.; Del’Olio, S.; Huff, T.C.; Chee, N.T.; et al. Dipeptide repeat proteins inhibit homology-directed DNA double strand break repair in C9ORF72 ALS/FTD. Mol. Neurodegener. 2020, 15, 13. [Google Scholar] [CrossRef]
  117. Deng, Q.; Holler, C.J.; Taylor, G.; Hudson, K.F.; Watkins, W.; Gearing, M.; Ito, D.; Murray, M.E.; Dickson, D.W.; Seyfried, N.T.; et al. FUS is phosphorylated by DNA-PK and accumulates in the cytoplasm after DNA damage. J. Neurosci. 2014, 34, 7802–7813. [Google Scholar] [CrossRef]
  118. Tohgi, H.; Abe, T.; Yamazaki, K.; Murata, T.; Ishizaki, E.; Isobe, C. Remarkable increase in cerebrospinal fluid 3-nitrotyrosine in patients with sporadic amyotrophic lateral sclerosis. Ann. Neurol. 1999, 46, 129–131. [Google Scholar] [CrossRef] [PubMed]
  119. Smith, R.G.; Henry, Y.K.; Mattson, M.P.; Appel, S.H. Presence of 4-hydroxynonenal in cerebrospinal fluid of patients with sporadic amyotrophic lateral sclerosis. Ann. Neurol. 1998, 44, 696–699. [Google Scholar] [CrossRef]
  120. Bogdanov, M.; Brown, R.H.; Matson, W.; Smart, R.; Hayden, D.; O’Donnell, H.; Flint Beal, M.; Cudkowicz, M. Increased oxidative damage to DNA in ALS patients. Free Radic. Biol. Med. 2000, 29, 652–658. [Google Scholar] [CrossRef]
  121. Ihara, Y.; Nobukuni, K.; Takata, H.; Hayabara, T. Oxidative stress and metal content in blood and cerebrospinal fluid of amyotrophic lateral sclerosis patients with and without a Cu, Zn-superoxide dismutase mutation. Neurol. Res. 2005, 27, 105–108. [Google Scholar] [CrossRef]
  122. Ikawa, M.; Okazawa, H.; Tsujikawa, T.; Matsunaga, A.; Yamamura, O.; Mori, T.; Hamano, T.; Kiyono, Y.; Nakamoto, Y.; Yoneda, M. Increased oxidative stress is related to disease severity in the ALS motor cortex: A PET study. Neurology 2015, 84, 2033–2039. [Google Scholar] [CrossRef]
  123. 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] [PubMed]
  124. Cunha-Oliveira, T.; Montezinho, L.; Mendes, C.; Firuzi, O.; Saso, L.; Oliveira, P.J.; Silva, F.S.G. Oxidative Stress in Amyotrophic Lateral Sclerosis: Pathophysiology and Opportunities for Pharmacological Intervention. Oxid. Med. Cell Longev. 2020, 2020, 5021694. [Google Scholar] [CrossRef] [PubMed]
  125. Abe, K.; Itoyama, Y.; Sobue, G.; Tsuji, S.; Aoki, M.; Doyu, M.; Hamada, C.; Kondo, K.; Yoneoka, T.; Akimoto, M.; et al. Confirmatory double-blind, parallel-group, placebo-controlled study of efficacy and safety of edaravone (MCI-186) in amyotrophic lateral sclerosis patients. Amyotroph Lateral Scler Front. Degener 2014, 15, 610–617. [Google Scholar] [CrossRef]
  126. Liu, S.; Liu, Y.; Hao, W.; Wolf, L.; Kiliaan, A.J.; Penke, B.; Rube, C.E.; Walter, J.; Heneka, M.T.; Hartmann, T.; et al. TLR2 is a primary receptor for Alzheimer’s amyloid beta peptide to trigger neuroinflammatory activation. J. Immunol. 2012, 188, 1098–1107. [Google Scholar] [CrossRef]
  127. Amor, S.; Puentes, F.; Baker, D.; van der Valk, P. Inflammation in neurodegenerative diseases. Immunology 2010, 129, 154–169. [Google Scholar] [CrossRef] [PubMed]
  128. Bsibsi, M.; Ravid, R.; Gveric, D.; van Noort, J.M. Broad expression of Toll-like receptors in the human central nervous system. J. Neuropathol. Exp. Neurol. 2002, 61, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
  129. van Noort, J.M.; Bsibsi, M. Toll-like receptors in the CNS: Implications for neurodegeneration and repair. Prog. Brain Res. 2009, 175, 139–148. [Google Scholar] [CrossRef]
  130. Hsieh, H.L.; Yang, C.M. Role of redox signaling in neuroinflammation and neurodegenerative diseases. Biomed. Res. Int. 2013, 2013, 484613. [Google Scholar] [CrossRef]
  131. Akira, S.; Takeda, K. Toll-like receptor signalling. Nat. Rev. Immunol. 2004, 4, 499–511. [Google Scholar] [CrossRef]
  132. Lamkanfi, M.; Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157, 1013–1022. [Google Scholar] [CrossRef] [PubMed]
  133. Cherry, J.D.; Olschowka, J.A.; O’Banion, M.K. Neuroinflammation and M2 microglia: The good, the bad, and the inflamed. J. Neuroinflammation 2014, 11, 98. [Google Scholar] [CrossRef]
  134. Daiber, A.; Di Lisa, F.; Oelze, M.; Kroller-Schon, S.; Steven, S.; Schulz, E.; Munzel, T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br. J. Pharmacol. 2017, 174, 1670–1689. [Google Scholar] [CrossRef]
  135. Hensley, K.; Maidt, M.L.; Yu, Z.; Sang, H.; Markesbery, W.R.; Floyd, R.A. Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation. J. Neurosci. 1998, 18, 8126–8132. [Google Scholar] [CrossRef]
  136. Castegna, A.; Thongboonkerd, V.; Klein, J.B.; Lynn, B.; Markesbery, W.R.; Butterfield, D.A. Proteomic identification of nitrated proteins in Alzheimer’s disease brain. J. Neurochem. 2003, 85, 1394–1401. [Google Scholar] [CrossRef]
  137. Stewart, C.R.; Stuart, L.M.; Wilkinson, K.; van Gils, J.M.; Deng, J.; Halle, A.; Rayner, K.J.; Boyer, L.; Zhong, R.; Frazier, W.A.; et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 2010, 11, 155–161. [Google Scholar] [CrossRef] [PubMed]
  138. Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef] [PubMed]
  139. Kitazawa, M.; Cheng, D.; Tsukamoto, M.R.; Koike, M.A.; Wes, P.D.; Vasilevko, V.; Cribbs, D.H.; LaFerla, F.M. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal beta-catenin pathway function in an Alzheimer’s disease model. J. Immunol. 2011, 187, 6539–6549. [Google Scholar] [CrossRef] [PubMed]
  140. Zhou, Y.; Lu, M.; Du, R.H.; Qiao, C.; Jiang, C.Y.; Zhang, K.Z.; Ding, J.H.; Hu, G. MicroRNA-7 targets Nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson’s disease. Mol. Neurodegener 2016, 11, 28. [Google Scholar] [CrossRef] [PubMed]
  141. Codolo, G.; Plotegher, N.; Pozzobon, T.; Brucale, M.; Tessari, I.; Bubacco, L.; de Bernard, M. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS ONE 2013, 8, e55375. [Google Scholar] [CrossRef] [PubMed]
  142. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef]
  143. Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 2010, 11, 136–140. [Google Scholar] [CrossRef]
  144. Baecher-Allan, C.; Kaskow, B.J.; Weiner, H.L. Multiple Sclerosis: Mechanisms and Immunotherapy. Neuron 2018, 97, 742–768. [Google Scholar] [CrossRef]
  145. Lassmann, H. Review: The architecture of inflammatory demyelinating lesions: Implications for studies on pathogenesis. Neuropathol. Appl. Neurobiol. 2011, 37, 698–710. [Google Scholar] [CrossRef]
  146. Babbe, H.; Roers, A.; Waisman, A.; Lassmann, H.; Goebels, N.; Hohlfeld, R.; Friese, M.; Schroder, R.; Deckert, M.; Schmidt, S.; et al. Clonal expansions of CD8+ T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction. J. Exp. Med. 2000, 192, 393–404. [Google Scholar] [CrossRef]
  147. Marik, C.; Felts, P.A.; Bauer, J.; Lassmann, H.; Smith, K.J. Lesion genesis in a subset of patients with multiple sclerosis: A role for innate immunity? Brain 2007, 130, 2800–2815. [Google Scholar] [CrossRef]
  148. Lassmann, H.; van Horssen, J. The molecular basis of neurodegeneration in multiple sclerosis. FEBS Lett. 2011, 585, 3715–3723. [Google Scholar] [CrossRef] [PubMed]
  149. Beland, L.C.; Markovinovic, A.; Jakovac, H.; De Marchi, F.; Bilic, E.; Mazzini, L.; Kriz, J.; Munitic, I. Immunity in amyotrophic lateral sclerosis: Blurred lines between excessive inflammation and inefficient immune responses. Brain Commun. 2020, 2, fcaa124. [Google Scholar] [CrossRef]
  150. Staats, K.A.; Borchelt, D.R.; Tansey, M.G.; Wymer, J. Blood-based biomarkers of inflammation in amyotrophic lateral sclerosis. Mol. Neurodegener 2022, 17, 11. [Google Scholar] [CrossRef] [PubMed]
  151. Correia, A.S.; Patel, P.; Dutta, K.; Julien, J.P. Inflammation Induces TDP-43 Mislocalization and Aggregation. PLoS ONE 2015, 10, e0140248. [Google Scholar] [CrossRef]
  152. Swarup, V.; Phaneuf, D.; Dupre, N.; Petri, S.; Strong, M.; Kriz, J.; Julien, J.P. Deregulation of TDP-43 in amyotrophic lateral sclerosis triggers nuclear factor kappaB-mediated pathogenic pathways. J. Exp. Med. 2011, 208, 2429–2447. [Google Scholar] [CrossRef] [PubMed]
  153. Kia, A.; McAvoy, K.; Krishnamurthy, K.; Trotti, D.; Pasinelli, P. Astrocytes expressing ALS-linked mutant FUS induce motor neuron death through release of tumor necrosis factor-alpha. Glia 2018, 66, 1016–1033. [Google Scholar] [CrossRef]
  154. Brettschneider, J.; Toledo, J.B.; Van Deerlin, V.M.; Elman, L.; McCluskey, L.; Lee, V.M.; Trojanowski, J.Q. Microglial activation correlates with disease progression and upper motor neuron clinical symptoms in amyotrophic lateral sclerosis. PLoS ONE 2012, 7, e39216. [Google Scholar] [CrossRef]
  155. Lino, M.M.; Schneider, C.; Caroni, P. Accumulation of SOD1 mutants in postnatal motoneurons does not cause motoneuron pathology or motoneuron disease. J. Neurosci. 2002, 22, 4825–4832. [Google Scholar] [CrossRef]
  156. Clement, A.M.; Nguyen, M.D.; Roberts, E.A.; Garcia, M.L.; Boillee, S.; Rule, M.; McMahon, A.P.; Doucette, W.; Siwek, D.; Ferrante, R.J.; et al. Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 2003, 302, 113–117. [Google Scholar] [CrossRef]
  157. Spiller, K.J.; Restrepo, C.R.; Khan, T.; Dominique, M.A.; Fang, T.C.; Canter, R.G.; Roberts, C.J.; Miller, K.R.; Ransohoff, R.M.; Trojanowski, J.Q.; et al. Microglia-mediated recovery from ALS-relevant motor neuron degeneration in a mouse model of TDP-43 proteinopathy. Nat. Neurosci. 2018, 21, 329–340. [Google Scholar] [CrossRef] [PubMed]
  158. Lei, Y.; Wang, K.; Deng, L.; Chen, Y.; Nice, E.C.; Huang, C. Redox regulation of inflammation: Old elements, a new story. Med. Res. Rev. 2015, 35, 306–340. [Google Scholar] [CrossRef] [PubMed]
  159. Wenzel, P.; Knorr, M.; Kossmann, S.; Stratmann, J.; Hausding, M.; Schuhmacher, S.; Karbach, S.H.; Schwenk, M.; Yogev, N.; Schulz, E.; et al. Lysozyme M-positive monocytes mediate angiotensin II-induced arterial hypertension and vascular dysfunction. Circulation 2011, 124, 1370–1381. [Google Scholar] [CrossRef] [PubMed]
  160. Kossmann, S.; Schwenk, M.; Hausding, M.; Karbach, S.H.; Schmidgen, M.I.; Brandt, M.; Knorr, M.; Hu, H.; Kroller-Schon, S.; Schonfelder, T.; et al. Angiotensin II-induced vascular dysfunction depends on interferon-gamma-driven immune cell recruitment and mutual activation of monocytes and NK-cells. Arterioscler Thromb. Vasc. Biol. 2013, 33, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
  161. Guzik, T.J.; Hoch, N.E.; Brown, K.A.; McCann, L.A.; Rahman, A.; Dikalov, S.; Goronzy, J.; Weyand, C.; Harrison, D.G. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J. Exp. Med. 2007, 204, 2449–2460. [Google Scholar] [CrossRef] [PubMed]
  162. Qiao, M.; Zhao, Q.; Lee, C.F.; Tannock, L.R.; Smart, E.J.; LeBaron, R.G.; Phelix, C.F.; Rangel, Y.; Asmis, R. Thiol oxidative stress induced by metabolic disorders amplifies macrophage chemotactic responses and accelerates atherogenesis and kidney injury in LDL receptor-deficient mice. Arterioscler Thromb Vasc. Biol. 2009, 29, 1779–1786. [Google Scholar] [CrossRef]
  163. Kim, H.S.; Ullevig, S.L.; Zamora, D.; Lee, C.F.; Asmis, R. Redox regulation of MAPK phosphatase 1 controls monocyte migration and macrophage recruitment. Proc. Natl. Acad. Sci. USA 2012, 109, E2803–E2812. [Google Scholar] [CrossRef]
  164. Kim, H.S.; Ullevig, S.L.; Nguyen, H.N.; Vanegas, D.; Asmis, R. Redox regulation of 14-3-3zeta controls monocyte migration. Arterioscler Thromb Vasc. Biol. 2014, 34, 1514–1521. [Google Scholar] [CrossRef]
  165. Ullevig, S.; Kim, H.S.; Asmis, R. S-glutathionylation in monocyte and macrophage (dys)function. Int. J. Mol. Sci. 2013, 14, 15212–15232. [Google Scholar] [CrossRef]
  166. Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target Ther. 2023, 8, 359. [Google Scholar] [CrossRef]
  167. Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef]
  168. Gerrits, E.; Brouwer, N.; Kooistra, S.M.; Woodbury, M.E.; Vermeiren, Y.; Lambourne, M.; Mulder, J.; Kummer, M.; Moller, T.; Biber, K.; et al. Distinct amyloid-beta and tau-associated microglia profiles in Alzheimer’s disease. Acta Neuropathol. 2021, 141, 681–696. [Google Scholar] [CrossRef]
  169. Smith, A.M.; Davey, K.; Tsartsalis, S.; Khozoie, C.; Fancy, N.; Tang, S.S.; Liaptsi, E.; Weinert, M.; McGarry, A.; Muirhead, R.C.J.; et al. Diverse human astrocyte and microglial transcriptional responses to Alzheimer’s pathology. Acta Neuropathol. 2022, 143, 75–91. [Google Scholar] [CrossRef]
  170. Nguyen, A.T.; Wang, K.; Hu, G.; Wang, X.; Miao, Z.; Azevedo, J.A.; Suh, E.; Van Deerlin, V.M.; Choi, D.; Roeder, K.; et al. APOE and TREM2 regulate amyloid-responsive microglia in Alzheimer’s disease. Acta Neuropathol. 2020, 140, 477–493. [Google Scholar] [CrossRef] [PubMed]
  171. Smajic, S.; Prada-Medina, C.A.; Landoulsi, Z.; Ghelfi, J.; Delcambre, S.; Dietrich, C.; Jarazo, J.; Henck, J.; Balachandran, S.; Pachchek, S.; et al. Single-cell sequencing of human midbrain reveals glial activation and a Parkinson-specific neuronal state. Brain 2022, 145, 964–978. [Google Scholar] [CrossRef] [PubMed]
  172. Kylkilahti, T.M.; Berends, E.; Ramos, M.; Shanbhag, N.C.; Toger, J.; Markenroth Bloch, K.; Lundgaard, I. Achieving brain clearance and preventing neurodegenerative diseases-A glymphatic perspective. J. Cereb Blood Flow. Metab. 2021, 41, 2137–2149. [Google Scholar] [CrossRef] [PubMed]
  173. Brandebura, A.N.; Paumier, A.; Onur, T.S.; Allen, N.J. Astrocyte contribution to dysfunction, risk and progression in neurodegenerative disorders. Nat. Rev. Neurosci. 2023, 24, 23–39. [Google Scholar] [CrossRef] [PubMed]
  174. Al-Dalahmah, O.; Sosunov, A.A.; Shaik, A.; Ofori, K.; Liu, Y.; Vonsattel, J.P.; Adorjan, I.; Menon, V.; Goldman, J.E. Single-nucleus RNA-seq identifies Huntington disease astrocyte states. Acta Neuropathol. Commun. 2020, 8, 19. [Google Scholar] [CrossRef] [PubMed]
  175. Habib, N.; McCabe, C.; Medina, S.; Varshavsky, M.; Kitsberg, D.; Dvir-Szternfeld, R.; Green, G.; Dionne, D.; Nguyen, L.; Marshall, J.L.; et al. Disease-associated astrocytes in Alzheimer’s disease and aging. Nat. Neurosci. 2020, 23, 701–706. [Google Scholar] [CrossRef] [PubMed]
  176. Lau, S.F.; Cao, H.; Fu, A.K.Y.; Ip, N.Y. Single-nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2020, 117, 25800–25809. [Google Scholar] [CrossRef] [PubMed]
  177. Leng, K.; Li, E.; Eser, R.; Piergies, A.; Sit, R.; Tan, M.; Neff, N.; Li, S.H.; Rodriguez, R.D.; Suemoto, C.K.; et al. Molecular characterization of selectively vulnerable neurons in Alzheimer’s disease. Nat. Neurosci. 2021, 24, 276–287. [Google Scholar] [CrossRef] [PubMed]
  178. Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef] [PubMed]
  179. Campbell, A.; Araujo, J.A.; Li, H.; Sioutas, C.; Kleinman, M. Particulate matter induced enhancement of inflammatory markers in the brains of apolipoprotein E knockout mice. J. Nanosci. Nanotechnol. 2009, 9, 5099–5104. [Google Scholar] [CrossRef] [PubMed]
  180. Campbell, A.; Oldham, M.; Becaria, A.; Bondy, S.C.; Meacher, D.; Sioutas, C.; Misra, C.; Mendez, L.B.; Kleinman, M. Particulate matter in polluted air may increase biomarkers of inflammation in mouse brain. Neurotoxicology 2005, 26, 133–140. [Google Scholar] [CrossRef] [PubMed]
  181. Hogan, M.K.; Kovalycsik, T.; Sun, Q.; Rajagopalan, S.; Nelson, R.J. Combined effects of exposure to dim light at night and fine particulate matter on C3H/HeNHsd mice. Behav. Brain Res. 2015, 294, 81–88. [Google Scholar] [CrossRef]
  182. Cheng, H.; Saffari, A.; Sioutas, C.; Forman, H.J.; Morgan, T.E.; Finch, C.E. Nanoscale Particulate Matter from Urban Traffic Rapidly Induces Oxidative Stress and Inflammation in Olfactory Epithelium with Concomitant Effects on Brain. Environ. Health Perspect. 2016, 124, 1537–1546. [Google Scholar] [CrossRef]
  183. Tyler, C.R.; Zychowski, K.E.; Sanchez, B.N.; Rivero, V.; Lucas, S.; Herbert, G.; Liu, J.; Irshad, H.; McDonald, J.D.; Bleske, B.E.; et al. Surface area-dependence of gas-particle interactions influences pulmonary and neuroinflammatory outcomes. Part Fibre Toxicol. 2016, 13, 64. [Google Scholar] [CrossRef]
  184. Herr, D.; Jew, K.; Wong, C.; Kennell, A.; Gelein, R.; Chalupa, D.; Raab, A.; Oberdorster, G.; Olschowka, J.; O’Banion, M.K.; et al. Effects of concentrated ambient ultrafine particulate matter on hallmarks of Alzheimer’s disease in the 3xTgAD mouse model. Neurotoxicology 2021, 84, 172–183. [Google Scholar] [CrossRef]
  185. Calderon-Garciduenas, L.; Herrera-Soto, A.; Jury, N.; Maher, B.A.; Gonzalez-Maciel, A.; Reynoso-Robles, R.; Ruiz-Rudolph, P.; van Zundert, B.; Varela-Nallar, L. Reduced repressive epigenetic marks, increased DNA damage and Alzheimer’s disease hallmarks in the brain of humans and mice exposed to particulate urban air pollution. Environ. Res. 2020, 183, 109226. [Google Scholar] [CrossRef]
  186. Bhatt, D.P.; Puig, K.L.; Gorr, M.W.; Wold, L.E.; Combs, C.K. A pilot study to assess effects of long-term inhalation of airborne particulate matter on early Alzheimer-like changes in the mouse brain. PLoS ONE 2015, 10, e0127102. [Google Scholar] [CrossRef]
  187. Cacciottolo, M.; Wang, X.; Driscoll, I.; Woodward, N.; Saffari, A.; Reyes, J.; Serre, M.L.; Vizuete, W.; Sioutas, C.; Morgan, T.E.; et al. Particulate air pollutants, APOE alleles and their contributions to cognitive impairment in older women and to amyloidogenesis in experimental models. Transl. Psychiatry 2017, 7, e1022. [Google Scholar] [CrossRef]
  188. Patten, K.T.; Valenzuela, A.E.; Wallis, C.; Berg, E.L.; Silverman, J.L.; Bein, K.J.; Wexler, A.S.; Lein, P.J. The Effects of Chronic Exposure to Ambient Traffic-Related Air Pollution on Alzheimer’s Disease Phenotypes in Wildtype and Genetically Predisposed Male and Female Rats. Environ. Health Perspect. 2021, 129, 57005. [Google Scholar] [CrossRef]
  189. Sahu, B.; Mackos, A.R.; Floden, A.M.; Wold, L.E.; Combs, C.K. Particulate Matter Exposure Exacerbates Amyloid-beta Plaque Deposition and Gliosis in APP/PS1 Mice. J. Alzheimer’s Dis. JAD 2021, 80, 761–774. [Google Scholar] [CrossRef]
  190. Lee, S.H.; Chen, Y.H.; Chien, C.C.; Yan, Y.H.; Chen, H.C.; Chuang, H.C.; Hsieh, H.I.; Cho, K.H.; Kuo, L.W.; Chou, C.C.; et al. Three month inhalation exposure to low-level PM2.5 induced brain toxicity in an Alzheimer’s disease mouse model. PLoS ONE 2021, 16, e0254587. [Google Scholar] [CrossRef] [PubMed]
  191. Ku, T.; Li, B.; Gao, R.; Zhang, Y.; Yan, W.; Ji, X.; Li, G.; Sang, N. NF-kappaB-regulated microRNA-574-5p underlies synaptic and cognitive impairment in response to atmospheric PM(2.5) aspiration. Part Fibre Toxicol. 2017, 14, 34. [Google Scholar] [CrossRef] [PubMed]
  192. Fonken, L.K.; Xu, X.; Weil, Z.M.; Chen, G.; Sun, Q.; Rajagopalan, S.; Nelson, R.J. Air pollution impairs cognition, provokes depressive-like behaviors and alters hippocampal cytokine expression and morphology. Mol. Psychiatry 2011, 16, 987–995, 973. [Google Scholar] [CrossRef]
  193. Rivas-Arancibia, S.; Zimbron, L.F.; Rodriguez-Martinez, E.; Maldonado, P.D.; Borgonio Perez, G.; Sepulveda-Parada, M. Oxidative stress-dependent changes in immune responses and cell death in the substantia nigra after ozone exposure in rat. Front. Aging Neurosci. 2015, 7, 65. [Google Scholar] [CrossRef] [PubMed]
  194. Santiago-Lopez, D.; Bautista-Martinez, J.A.; Reyes-Hernandez, C.I.; Aguilar-Martinez, M.; Rivas-Arancibia, S. Oxidative stress, progressive damage in the substantia nigra and plasma dopamine oxidation, in rats chronically exposed to ozone. Toxicol. Lett. 2010, 197, 193–200. [Google Scholar] [CrossRef]
  195. Mumaw, C.L.; Levesque, S.; McGraw, C.; Robertson, S.; Lucas, S.; Stafflinger, J.E.; Campen, M.J.; Hall, P.; Norenberg, J.P.; Anderson, T.; et al. Microglial priming through the lung-brain axis: The role of air pollution-induced circulating factors. Faseb. J. 2016, 30, 1880–1891. [Google Scholar] [CrossRef]
  196. Hernandez-Zimbron, L.F.; Rivas-Arancibia, S. Oxidative stress caused by ozone exposure induces beta-amyloid 1-42 overproduction and mitochondrial accumulation by activating the amyloidogenic pathway. Neuroscience 2015, 304, 340–348. [Google Scholar] [CrossRef] [PubMed]
  197. Calderon-Garciduenas, L.; Azzarelli, B.; Acuna, H.; Garcia, R.; Gambling, T.M.; Osnaya, N.; Monroy, S.; MR, D.E.L.T.; Carson, J.L.; Villarreal-Calderon, A.; et al. Air pollution and brain damage. Toxicol. Pathol. 2002, 30, 373–389. [Google Scholar] [CrossRef]
  198. Calderon-Garciduenas, L.; Maronpot, R.R.; Torres-Jardon, R.; Henriquez-Roldan, C.; Schoonhoven, R.; Acuna-Ayala, H.; Villarreal-Calderon, A.; Nakamura, J.; Fernando, R.; Reed, W.; et al. DNA damage in nasal and brain tissues of canines exposed to air pollutants is associated with evidence of chronic brain inflammation and neurodegeneration. Toxicol. Pathol. 2003, 31, 524–538. [Google Scholar] [CrossRef] [PubMed]
  199. Calderon-Garciduenas, L.; Mora-Tiscareno, A.; Gomez-Garza, G.; Carrasco-Portugal Mdel, C.; Perez-Guille, B.; Flores-Murrieta, F.J.; Perez-Guille, G.; Osnaya, N.; Juarez-Olguin, H.; Monroy, M.E.; et al. Effects of a cyclooxygenase-2 preferential inhibitor in young healthy dogs exposed to air pollution: A pilot study. Toxicol. Pathol. 2009, 37, 644–660. [Google Scholar] [CrossRef]
  200. Levesque, S.; Taetzsch, T.; Lull, M.E.; Kodavanti, U.; Stadler, K.; Wagner, A.; Johnson, J.A.; Duke, L.; Kodavanti, P.; Surace, M.J.; et al. Diesel exhaust activates and primes microglia: Air pollution, neuroinflammation, and regulation of dopaminergic neurotoxicity. Environ. Health Perspect. 2011, 119, 1149–1155. [Google Scholar] [CrossRef] [PubMed]
  201. Gerlofs-Nijland, M.E.; van Berlo, D.; Cassee, F.R.; Schins, R.P.; Wang, K.; Campbell, A. Effect of prolonged exposure to diesel engine exhaust on proinflammatory markers in different regions of the rat brain. Part Fibre Toxicol. 2010, 7, 12. [Google Scholar] [CrossRef]
  202. Oppenheim, H.A.; Lucero, J.; Guyot, A.C.; Herbert, L.M.; McDonald, J.D.; Mabondzo, A.; Lund, A.K. Exposure to vehicle emissions results in altered blood brain barrier permeability and expression of matrix metalloproteinases and tight junction proteins in mice. Part Fibre Toxicol. 2013, 10, 62. [Google Scholar] [CrossRef]
  203. Mumaw, C.L.; Surace, M.; Levesque, S.; Kodavanti, U.P.; Kodavanti, P.R.S.; Royland, J.E.; Block, M.L. Atypical microglial response to biodiesel exhaust in healthy and hypertensive rats. Neurotoxicology 2017, 59, 155–163. [Google Scholar] [CrossRef]
  204. Kilian, J.G.; Mejias-Ortega, M.; Hsu, H.W.; Herman, D.A.; Vidal, J.; Arechavala, R.J.; Renusch, S.; Dalal, H.; Hasen, I.; Ting, A.; et al. Exposure to quasi-ultrafine particulate matter accelerates memory impairment and Alzheimer’s disease-like neuropathology in the AppNL-G-F knock-in mouse model. Toxicol. Sci. 2023, 193, 175–191. [Google Scholar] [CrossRef]
  205. 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]
  206. Yuan, X.; Yang, Y.; Liu, C.; Tian, Y.; Xia, D.; Liu, Z.; Pan, L.; Xiong, M.; Xiong, J.; Meng, L.; et al. Fine Particulate Matter Triggers alpha-Synuclein Fibrillization and Parkinson-like Neurodegeneration. Mov. Disord. 2022, 37, 1817–1830. [Google Scholar] [CrossRef]
  207. Jayaraj, R.L.; Rodriguez, E.A.; Wang, Y.; Block, M.L. Outdoor Ambient Air Pollution and Neurodegenerative Diseases: The Neuroinflammation Hypothesis. Curr. Environ. Health Rep. 2017, 4, 166–179. [Google Scholar] [CrossRef]
  208. Calderon-Garciduenas, L.; Solt, A.C.; Henriquez-Roldan, C.; Torres-Jardon, R.; Nuse, B.; Herritt, L.; Villarreal-Calderon, R.; Osnaya, N.; Stone, I.; Garcia, R.; et al. Long-term air pollution exposure is associated with neuroinflammation, an altered innate immune response, disruption of the blood-brain barrier, ultrafine particulate deposition, and accumulation of amyloid beta-42 and alpha-synuclein in children and young adults. Toxicol. Pathol. 2008, 36, 289–310. [Google Scholar] [CrossRef]
  209. Muhlfeld, C.; Rothen-Rutishauser, B.; Blank, F.; Vanhecke, D.; Ochs, M.; Gehr, P. Interactions of nanoparticles with pulmonary structures and cellular responses. Am. J. Physiol. Lung Cell Mol. Physiol. 2008, 294, L817–L829. [Google Scholar] [CrossRef] [PubMed]
  210. Oberdorster, 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] [PubMed]
  211. Kuntic, M.; Kuntic, I.; Krishnankutty, R.; Gericke, A.; Oelze, M.; Junglas, T.; Bayo Jimenez, M.T.; Stamm, P.; Nandudu, M.; Hahad, O.; et al. Co-exposure to urban particulate matter and aircraft noise adversely impacts the cerebro-pulmonary-cardiovascular axis in mice. Redox Biol. 2023, 59, 102580. [Google Scholar] [CrossRef] [PubMed]
  212. Block, M.L.; Calderon-Garciduenas, L. Air pollution: Mechanisms of neuroinflammation and CNS disease. Trends Neurosci. 2009, 32, 506–516. [Google Scholar] [CrossRef] [PubMed]
  213. Calderon-Garciduenas, L.; Cross, J.V.; Franco-Lira, M.; Aragon-Flores, M.; Kavanaugh, M.; Torres-Jardon, R.; Chao, C.K.; Thompson, C.; Chang, J.; Zhu, H.; et al. Brain immune interactions and air pollution: Macrophage inhibitory factor (MIF), prion cellular protein (PrP(C)), Interleukin-6 (IL-6), interleukin 1 receptor antagonist (IL-1Ra), and interleukin-2 (IL-2) in cerebrospinal fluid and MIF in serum differentiate urban children exposed to severe vs. low air pollution. Front. Neurosci. 2013, 7, 183. [Google Scholar] [CrossRef] [PubMed]
  214. Tamagawa, E.; van Eeden, S.F. Impaired lung function and risk for stroke: Role of the systemic inflammation response? Chest 2006, 130, 1631–1633. [Google Scholar] [CrossRef] [PubMed]
  215. Kodavanti, U.P. Stretching the stress boundary: Linking air pollution health effects to a neurohormonal stress response. Biochim. Biophys. Acta 2016, 1860, 2880–2890. [Google Scholar] [CrossRef] [PubMed]
  216. Babisch, W. The Noise/Stress Concept, Risk Assessment and Research Needs. Noise Health 2002, 4, 1–11. [Google Scholar]
  217. Campos-Rodriguez, R.; Godinez-Victoria, M.; Abarca-Rojano, E.; Pacheco-Yepez, J.; Reyna-Garfias, H.; Barbosa-Cabrera, R.E.; Drago-Serrano, M.E. Stress modulates intestinal secretory immunoglobulin A. Front. Integr. Neurosci. 2013, 7, 86. [Google Scholar] [CrossRef]
  218. Munzel, T.; Sorensen, M.; Daiber, A. Transportation noise pollution and cardiovascular disease. Nat. Rev. Cardiol. 2021, 18, 619–636. [Google Scholar] [CrossRef]
  219. Daiber, A.; Kroller-Schon, S.; Frenis, K.; Oelze, M.; Kalinovic, S.; Vujacic-Mirski, K.; Kuntic, M.; Bayo Jimenez, M.T.; Helmstadter, J.; Steven, S.; et al. Environmental noise induces the release of stress hormones and inflammatory signaling molecules leading to oxidative stress and vascular dysfunction-Signatures of the internal exposome. Biofactors 2019, 45, 495–506. [Google Scholar] [CrossRef] [PubMed]
  220. Manukyan, A.L. Noise as a cause of neurodegenerative disorders: Molecular and cellular mechanisms. Neurol. Sci. 2022, 43, 2983–2993. [Google Scholar] [CrossRef] [PubMed]
  221. Jafari, Z.; Kolb, B.E.; Mohajerani, M.H. Noise exposure accelerates the risk of cognitive impairment and Alzheimer’s disease: Adulthood, gestational, and prenatal mechanistic evidence from animal studies. Neurosci. Biobehav. Rev. 2020, 117, 110–128. [Google Scholar] [CrossRef] [PubMed]
  222. Kroller-Schon, S.; Daiber, A.; Steven, S.; Oelze, M.; Frenis, K.; Kalinovic, S.; Heimann, A.; Schmidt, F.P.; Pinto, A.; Kvandova, M.; et al. Crucial role for Nox2 and sleep deprivation in aircraft noise-induced vascular and cerebral oxidative stress, inflammation, and gene regulation. Eur. Heart J. 2018, 39, 3528–3539. [Google Scholar] [CrossRef] [PubMed]
  223. Munzel, T.; Daiber, A.; Steven, S.; Tran, L.P.; Ullmann, E.; Kossmann, S.; Schmidt, F.P.; Oelze, M.; Xia, N.; Li, H.; et al. Effects of noise on vascular function, oxidative stress, and inflammation: Mechanistic insight from studies in mice. Eur. Heart J. 2017, 38, 2838–2849. [Google Scholar] [CrossRef] [PubMed]
  224. Gai, Z.; Li, K.; Sun, H.; She, X.; Cui, B.; Wang, R. Effects of chronic noise on mRNA and protein expression of CRF family molecules and its relationship with p-tau in the rat prefrontal cortex. J. Neurol. Sci. 2016, 368, 307–313. [Google Scholar] [CrossRef]
  225. Cui, B.; Zhu, L.; She, X.; Wu, M.; Ma, Q.; Wang, T.; Zhang, N.; Xu, C.; Chen, X.; An, G.; et al. Chronic noise exposure causes persistence of tau hyperphosphorylation and formation of NFT tau in the rat hippocampus and prefrontal cortex. Exp. Neurol. 2012, 238, 122–129. [Google Scholar] [CrossRef]
  226. Zheng, P.; She, X.; Wang, C.; Zhu, Y.; Fu, B.; Ma, K.; Yang, H.; Gao, X.; Li, X.; Wu, F.; et al. Around-the-Clock Noise Induces AD-like Neuropathology by Disrupting Autophagy Flux Homeostasis. Cells 2022, 11, 2742. [Google Scholar] [CrossRef]
  227. Li, Q.; Liu, Y.; Sun, M. Autophagy and Alzheimer’s Disease. Cell Mol. Neurobiol. 2017, 37, 377–388. [Google Scholar] [CrossRef]
  228. Su, D.; Li, W.; Chi, H.; Yang, H.; She, X.; Wang, K.; Gao, X.; Ma, K.; Zhang, M.; Cui, B. Transcriptome analysis of the hippocampus in environmental noise-exposed SAMP8 mice reveals regulatory pathways associated with Alzheimer’s disease neuropathology. Environ. Health Prev. Med. 2020, 25, 3. [Google Scholar] [CrossRef]
  229. Cui, B.; Su, D.; Li, W.; She, X.; Zhang, M.; Wang, R.; Zhai, Q. Effects of chronic noise exposure on the microbiome-gut-brain axis in senescence-accelerated prone mice: Implications for Alzheimer’s disease. J. Neuroinflammation 2018, 15, 190. [Google Scholar] [CrossRef] [PubMed]
  230. Su, D.; Li, W.; She, X.; Chen, X.; Zhai, Q.; Cui, B.; Wang, R. Chronic noise exposure exacerbates AD-like neuropathology in SAMP8 mice in relation to Wnt signaling in the PFC and hippocampus. Sci. Rep. 2018, 8, 14622. [Google Scholar] [CrossRef] [PubMed]
  231. Chi, H.; Cao, W.; Zhang, M.; Su, D.; Yang, H.; Li, Z.; Li, C.; She, X.; Wang, K.; Gao, X.; et al. Environmental noise stress disturbs commensal microbiota homeostasis and induces oxi-inflammmation and AD-like neuropathology through epithelial barrier disruption in the EOAD mouse model. J. Neuroinflammation 2021, 18, 9. [Google Scholar] [CrossRef] [PubMed]
  232. Jafari, Z.; Okuma, M.; Karem, H.; Mehla, J.; Kolb, B.E.; Mohajerani, M.H. Prenatal noise stress aggravates cognitive decline and the onset and progression of beta amyloid pathology in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2019, 77, 66–86. [Google Scholar] [CrossRef]
  233. Saljo, A.; Bao, F.; Shi, J.; Hamberger, A.; Hansson, H.A.; Haglid, K.G. Expression of c-Fos and c-Myc and deposition of beta-APP in neurons in the adult rat brain as a result of exposure to short-lasting impulse noise. J. Neurotrauma 2002, 19, 379–385. [Google Scholar] [CrossRef]
  234. Paciello, F.; Rinaudo, M.; Longo, V.; Cocco, S.; Conforto, G.; Pisani, A.; Podda, M.V.; Fetoni, A.R.; Paludetti, G.; Grassi, C. Auditory sensory deprivation induced by noise exposure exacerbates cognitive decline in a mouse model of Alzheimer’s disease. eLife 2021, 10, e70908. [Google Scholar] [CrossRef]
  235. Paciello, F.; Pisani, A.; Rinaudo, M.; Cocco, S.; Paludetti, G.; Fetoni, A.R.; Grassi, C. Noise-induced auditory damage affects hippocampus causing memory deficits in a model of early age-related hearing loss. Neurobiol. Dis. 2023, 178, 106024. [Google Scholar] [CrossRef]
  236. Qian, M.; Wang, Q.; Wang, Z.; Ma, Q.; Wang, X.; Han, K.; Wu, H.; Huang, Z. Dose-Dependent Pattern of Cochlear Synaptic Degeneration in C57BL/6J Mice Induced by Repeated Noise Exposure. Neural. Plast. 2021, 2021, 9919977. [Google Scholar] [CrossRef] [PubMed]
  237. Hahad, O.; Bayo Jimenez, M.T.; Kuntic, M.; Frenis, K.; Steven, S.; Daiber, A.; Munzel, T. Cerebral consequences of environmental noise exposure. Environ. Int. 2022, 165, 107306. [Google Scholar] [CrossRef] [PubMed]
  238. Hahad, O.; Lelieveld, J.; Birklein, F.; Lieb, K.; Daiber, A.; Munzel, T. Ambient Air Pollution Increases the Risk of Cerebrovascular and Neuropsychiatric Disorders through Induction of Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2020, 21, 4306. [Google Scholar] [CrossRef] [PubMed]
  239. 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] [PubMed]
  240. 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. Hydrogen Environ. Health 2019, 222, 402–409. [Google Scholar] [CrossRef] [PubMed]
  241. 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]
  242. 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]
  243. Fu, P.; Guo, X.; Cheung, F.M.H.; Yung, K.K.L. The association between PM2.5 exposure and neurological disorders: A systematic review and meta-analysis. Sci. Total Environ. 2019, 655, 1240–1248. [Google Scholar] [CrossRef]
  244. Grande, G.; Ljungman, P.L.S.; Eneroth, K.; Bellander, T.; Rizzuto, D. Association Between Cardiovascular Disease and Long-term Exposure to Air Pollution With the Risk of Dementia. JAMA Neurol. 2020, 77, 801–809. [Google Scholar] [CrossRef]
  245. Ilango, S.D.; Chen, H.; Hystad, P.; van Donkelaar, A.; Kwong, J.C.; Tu, K.; Martin, R.V.; Benmarhnia, T. The role of cardiovascular disease in the relationship between air pollution and incident dementia: A population-based cohort study. Int. J. Epidemiol. 2020, 49, 36–44. [Google Scholar] [CrossRef]
  246. Cerza, F.; Renzi, M.; Gariazzo, C.; Davoli, M.; Michelozzi, P.; Forastiere, F.; Cesaroni, G. Long-term exposure to air pollution and hospitalization for dementia in the Rome longitudinal study. Environ. Health A Glob. Access Sci. Source 2019, 18, 72. [Google Scholar] [CrossRef]
  247. Li, C.Y.; Li, C.H.; Martini, S.; Hou, W.H. Association between air pollution and risk of vascular dementia: A multipollutant analysis in Taiwan. Environ. Int. 2019, 133, 105233. [Google Scholar] [CrossRef]
  248. 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]
  249. Oudin, A.; Andersson, J.; Sundstrom, A.; Nordin Adolfsson, A.; Oudin Astrom, D.; Adolfsson, R.; Forsberg, B.; Nordin, M. Traffic-Related Air Pollution as a Risk Factor for Dementia: No Clear Modifying Effects of APOEvarepsilon4 in the Betula Cohort. J. Alzheimer’s Dis. JAD 2019, 71, 733–740. [Google Scholar] [CrossRef] [PubMed]
  250. Chen, H.; Kwong, J.C.; Copes, R.; Hystad, P.; van Donkelaar, A.; Tu, K.; Brook, J.R.; Goldberg, M.S.; Martin, R.V.; Murray, B.J.; et al. Exposure to ambient air pollution and the incidence of dementia: A population-based cohort study. Environ. Int. 2017, 108, 271–277. [Google Scholar] [CrossRef] [PubMed]
  251. Chen, H.; Kwong, J.C.; Copes, R.; Tu, K.; Villeneuve, P.J.; van Donkelaar, A.; Hystad, P.; Martin, R.V.; Murray, B.J.; Jessiman, B.; et al. Living near major roads and the incidence of dementia, Parkinson’s disease, and multiple sclerosis: A population-based cohort study. Lancet 2017, 389, 718–726. [Google Scholar] [CrossRef] [PubMed]
  252. Andersson, J.; Oudin, A.; Sundstrom, A.; Forsberg, B.; Adolfsson, R.; Nordin, M. Road traffic noise, air pollution, and risk of dementia—results from the Betula project. Environ. Res. 2018, 166, 334–339. [Google Scholar] [CrossRef]
  253. Carey, I.M.; Anderson, H.R.; Atkinson, R.W.; Beevers, S.D.; Cook, D.G.; Strachan, D.P.; Dajnak, D.; Gulliver, J.; Kelly, F.J. Are noise and air pollution related to the incidence of dementia? A cohort study in London, England. BMJ Open 2018, 8, e022404. [Google Scholar] [CrossRef] [PubMed]
  254. Malek, A.M.; Arena, V.C.; Song, R.; Whitsel, E.A.; Rager, J.R.; Stewart, J.; Yanosky, J.D.; Liao, D.; Talbott, E.O. Long-term air pollution and risk of amyotrophic lateral sclerosis mortality in the Women’s Health Initiative cohort. Environ. Res. 2023, 216, 114510. [Google Scholar] [CrossRef]
  255. Malek, A.M.; Barchowsky, A.; Bowser, R.; Heiman-Patterson, T.; Lacomis, D.; Rana, S.; Ada, Y.; Talbott, E.O. Exposure to hazardous air pollutants and the risk of amyotrophic lateral sclerosis. Environ. Pollut. 2015, 197, 181–186. [Google Scholar] [CrossRef]
  256. 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]
  257. Parks, R.M.; Nunez, Y.; Balalian, A.A.; Gibson, E.A.; Hansen, J.; Raaschou-Nielsen, O.; Ketzel, M.; Khan, J.; Brandt, J.; Vermeulen, R.; et al. Long-term Traffic-related Air Pollutant Exposure and Amyotrophic Lateral Sclerosis Diagnosis in Denmark: A Bayesian Hierarchical Analysis. Epidemiology 2022, 33, 757–766. [Google Scholar] [CrossRef]
  258. 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] [PubMed]
  259. Antonioni, A.; Govoni, V.; Brancaleoni, L.; Dona, A.; Granieri, E.; Bergamini, M.; Gerdol, R.; Pugliatti, M. Amyotrophic Lateral Sclerosis and Air Pollutants in the Province of Ferrara, Northern Italy: An Ecological Study. Int. J. Environ. Res. Public Health 2023, 20, 5591. [Google Scholar] [CrossRef] [PubMed]
  260. Yu, Z.; Peters, S.; van Boxmeer, L.; Downward, G.S.; Hoek, G.; Kioumourtzoglou, M.A.; Weisskopf, M.G.; Hansen, J.; van den Berg, L.H.; Vermeulen, R.C.H. Long-Term Exposure to Ultrafine Particles and Particulate Matter Constituents and the Risk of Amyotrophic Lateral Sclerosis. Environ. Health Perspect. 2021, 129, 97702. [Google Scholar] [CrossRef]
  261. 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] [PubMed]
  262. Angelici, L.; Piola, M.; Cavalleri, T.; Randi, G.; Cortini, F.; Bergamaschi, R.; Baccarelli, A.A.; Bertazzi, P.A.; Pesatori, A.C.; Bollati, V. Effects of particulate matter exposure on multiple sclerosis hospital admission in Lombardy region, Italy. Environ. Res. 2016, 145, 68–73. [Google Scholar] [CrossRef]
  263. Vojinovic, S.; Savic, D.; Lukic, S.; Savic, L.; Vojinovic, J. Disease relapses in multiple sclerosis can be influenced by air pollution and climate seasonal conditions. Vojnosanit Pregl. 2015, 72, 44–49. [Google Scholar] [CrossRef]
  264. Weuve, J.; D’Souza, J.; Beck, T.; Evans, D.A.; Kaufman, J.D.; Rajan, K.B.; de Leon, C.F.M.; Adar, S.D. Long-term community noise exposure in relation to dementia, cognition, and cognitive decline in older adults. Alzheimers Dement. 2021, 17, 525–533. [Google Scholar] [CrossRef]
  265. Tzivian, L.; Dlugaj, M.; Winkler, A.; Weinmayr, G.; Hennig, F.; Fuks, K.B.; Vossoughi, M.; Schikowski, T.; Weimar, C.; Erbel, R.; et al. Long-Term Air Pollution and Traffic Noise Exposures and Mild Cognitive Impairment in Older Adults: A Cross-Sectional Analysis of the Heinz Nixdorf Recall Study. Environ. Health Perspect. 2016, 124, 1361–1368. [Google Scholar] [CrossRef]
  266. Tzivian, L.; Dlugaj, M.; Winkler, A.; Hennig, F.; Fuks, K.; Sugiri, D.; Schikowski, T.; Jakobs, H.; Erbel, R.; Jockel, K.H.; et al. Long-term air pollution and traffic noise exposures and cognitive function:A cross-sectional analysis of the Heinz Nixdorf Recall study. J. Toxicol. Environ. Health A 2016, 79, 1057–1069. [Google Scholar] [CrossRef]
  267. Tzivian, L.; Jokisch, M.; Winkler, A.; Weimar, C.; Hennig, F.; Sugiri, D.; Soppa, V.J.; Dragano, N.; Erbel, R.; Jockel, K.H.; et al. Associations of long-term exposure to air pollution and road traffic noise with cognitive function-An analysis of effect measure modification. Environ. Int. 2017, 103, 30–38. [Google Scholar] [CrossRef]
  268. Fuks, K.B.; Wigmann, C.; Altug, H.; Schikowski, T. Road Traffic Noise at the Residence, Annoyance, and Cognitive Function in Elderly Women. Int. J. Environ. Res. Public Health 2019, 16, 1790. [Google Scholar] [CrossRef]
  269. Mac Domhnaill, C.; Douglas, O.; Lyons, S.; Murphy, E.; Nolan, A. Road traffic noise and cognitive function in older adults: A cross-sectional investigation of The Irish Longitudinal Study on Ageing. BMC Public Health 2021, 21, 1814. [Google Scholar] [CrossRef]
  270. Yu, Y.; Mayeda, E.R.; Paul, K.C.; Lee, E.; Jerrett, M.; Su, J.; Wu, J.; Shih, I.F.; Haan, M.; Ritz, B. Traffic-related Noise Exposure and Late-life Dementia and Cognitive Impairment in Mexican-Americans. Epidemiology 2020, 31, 771–778. [Google Scholar] [CrossRef] [PubMed]
  271. Yu, Y.; Haan, M.; Paul, K.C.; Mayeda, E.R.; Jerrett, M.; Wu, J.; Lee, E.; Su, J.; Shih, I.F.; Inoue, K.; et al. Metabolic dysfunction modifies the influence of traffic-related air pollution and noise exposure on late-life dementia and cognitive impairment: A cohort study of older Mexican-Americans. Environ. Epidemiol. 2020, 4, e122. [Google Scholar] [CrossRef]
  272. Linares, C.; Culqui, D.; Carmona, R.; Ortiz, C.; Diaz, J. Short-term association between environmental factors and hospital admissions due to dementia in Madrid. Environ. Res. 2017, 152, 214–220. [Google Scholar] [CrossRef]
  273. Yuchi, W.; Sbihi, H.; Davies, H.; Tamburic, L.; Brauer, M. Road proximity, air pollution, noise, green space and neurologic disease incidence: A population-based cohort study. Environ. Health A Glob. Access Sci. Source 2020, 19, 8. [Google Scholar] [CrossRef] [PubMed]
  274. Cantuaria, M.L.; Waldorff, F.B.; Wermuth, L.; Pedersen, E.R.; Poulsen, A.H.; Thacher, J.D.; Raaschou-Nielsen, O.; Ketzel, M.; Khan, J.; Valencia, V.H.; et al. Residential exposure to transportation noise in Denmark and incidence of dementia: National cohort study. BMJ 2021, 374, n1954. [Google Scholar] [CrossRef]
  275. Cole-Hunter, T.; So, R.; Amini, H.; Backalarz, C.; Brandt, J.; Brauner, E.V.; Hertel, O.; Jensen, S.S.; Jorgensen, J.T.; Ketzel, M.; et al. Long-term exposure to road traffic noise and all-cause and cause-specific mortality: A Danish Nurse Cohort study. Sci. Total Environ. 2022, 820, 153057. [Google Scholar] [CrossRef] [PubMed]
  276. Parra, K.L.; Alexander, G.E.; Raichlen, D.A.; Klimentidis, Y.C.; Furlong, M.A. Exposure to air pollution and risk of incident dementia in the UK Biobank. Environ. Res. 2022, 209, 112895. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Interplay between neuroinflammation and oxidative stress in neurodegeneration. Direct injury to the neuronal tissue or systemic inflammatory response trigger the activation of damage-associated molecular patterns (DAMPs—such as amyloid-beta, fibrinogen, high mobility group box 1 (HMGB1) protein, mitochondrial transcription factor A (TFAM) and cytochrome c). DAMPs signal activation of microglia (resident macrophages) through pattern recognition receptors (PRRs), such as complement receptor 3 (CD11b), Toll-like receptor 4 (TLR4), and NLR family pyrin domain containing 3 (NLRP3) inflammasome. These signals are transduced in microglia by nuclear factor κB (NF-κB), activator protein 1 (AP-1), and mitogen-activated protein kinases (MAPKs)/extracellular signal-regulated kinase (ERK). Activated microglia produce superoxide (O2) through NADPH oxidase (NOX-2), promoting oxidative stress. Oxidative stress can be based on lower antioxidant defense (reduced/oxidised glutathione—GHS/GSSG; superoxide dismutase—SOD; catalase—CAT), and an increase in reactive oxygen and nitrogen species (peroxynitrite—ONOO and O2) formation. Both neuroinflammation and oxidative stress have a detrimental effect on neuronal tissue, causing neuronal death and promoting neurodegeneration. Adapted from [66] under Creative Common CC BY license. Created with BioRender.com.
Figure 1. Interplay between neuroinflammation and oxidative stress in neurodegeneration. Direct injury to the neuronal tissue or systemic inflammatory response trigger the activation of damage-associated molecular patterns (DAMPs—such as amyloid-beta, fibrinogen, high mobility group box 1 (HMGB1) protein, mitochondrial transcription factor A (TFAM) and cytochrome c). DAMPs signal activation of microglia (resident macrophages) through pattern recognition receptors (PRRs), such as complement receptor 3 (CD11b), Toll-like receptor 4 (TLR4), and NLR family pyrin domain containing 3 (NLRP3) inflammasome. These signals are transduced in microglia by nuclear factor κB (NF-κB), activator protein 1 (AP-1), and mitogen-activated protein kinases (MAPKs)/extracellular signal-regulated kinase (ERK). Activated microglia produce superoxide (O2) through NADPH oxidase (NOX-2), promoting oxidative stress. Oxidative stress can be based on lower antioxidant defense (reduced/oxidised glutathione—GHS/GSSG; superoxide dismutase—SOD; catalase—CAT), and an increase in reactive oxygen and nitrogen species (peroxynitrite—ONOO and O2) formation. Both neuroinflammation and oxidative stress have a detrimental effect on neuronal tissue, causing neuronal death and promoting neurodegeneration. Adapted from [66] under Creative Common CC BY license. Created with BioRender.com.
Antioxidants 13 00266 g001
Figure 2. Overview on redox-dependent exacerbation of inflammatory processes and pathways. Primary sources of superoxide are the phagocytic NADPH oxidase in activated immune cells and damaged mitochondria. Superoxide either dismutates to hydrogen peroxide or reacts with nitric oxide from iNOS to form peroxynitrite (ONOO). These ROS can cause redox modifications in master regulators of inflammation such as HMGB1, NF-κB, and NLRP3 or of central inflammatory processes such as NETosis. Also upstream regulators of these processes are redox-regulated. The red-groups indicate redox modifications. S—S, disulfide bridge; SO3H, sulfonic acid; SOCH3, oxidized methionine; SG, S-glutathionylation; 3NT, 3-nitrotyrosine; SNO, S-nitrosothiol; SH, reduced thiol. Other abbreviations are as follows: ER, endoplasmic reticulum; UPR, unfolded protein response; mPTP, mitochondrial permeability transition pore; mtDNA, mitochondrial DNA; Trx, thioredoxin; AP-1, activator protein 1 transcription factor; IkB, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor; TLR, Toll-like receptor; RAGE, receptor for advanced glycation end products; OCl, hypochlorite; MPO, myeloperoxidase; Keap1, Kelch-like ECH-associated protein 1; Nrf2, nuclear factor erythroid 2-related factor 2; Casp-1, caspase 1; Nox, NADPH oxidase. Scheme was generated from data reported in [65,158]. Created with BioRender.com.
Figure 2. Overview on redox-dependent exacerbation of inflammatory processes and pathways. Primary sources of superoxide are the phagocytic NADPH oxidase in activated immune cells and damaged mitochondria. Superoxide either dismutates to hydrogen peroxide or reacts with nitric oxide from iNOS to form peroxynitrite (ONOO). These ROS can cause redox modifications in master regulators of inflammation such as HMGB1, NF-κB, and NLRP3 or of central inflammatory processes such as NETosis. Also upstream regulators of these processes are redox-regulated. The red-groups indicate redox modifications. S—S, disulfide bridge; SO3H, sulfonic acid; SOCH3, oxidized methionine; SG, S-glutathionylation; 3NT, 3-nitrotyrosine; SNO, S-nitrosothiol; SH, reduced thiol. Other abbreviations are as follows: ER, endoplasmic reticulum; UPR, unfolded protein response; mPTP, mitochondrial permeability transition pore; mtDNA, mitochondrial DNA; Trx, thioredoxin; AP-1, activator protein 1 transcription factor; IkB, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor; TLR, Toll-like receptor; RAGE, receptor for advanced glycation end products; OCl, hypochlorite; MPO, myeloperoxidase; Keap1, Kelch-like ECH-associated protein 1; Nrf2, nuclear factor erythroid 2-related factor 2; Casp-1, caspase 1; Nox, NADPH oxidase. Scheme was generated from data reported in [65,158]. Created with BioRender.com.
Antioxidants 13 00266 g002
Figure 3. Mechanisms of air pollution-induced neuroinflammation and oxidative stress. Air pollution can cause neuroinflammation and oxidative stress through two pathways: direct and indirect. Air pollution components directly interact with the neuronal tissue in the direct pathway. This can happen by direct contact of air pollution components, such as particulate matter (PM), with the nerves in the olfactory bulb, helping them translocate deeper into the neuronal tissue. Another way is for ultrafine particles (UFP) with a diameter in the nanometer range to enter the systemic circulation after inhalation, causing disturbance of the blood–brain barrier (BBB) and reaching the cerebral tissue. The indirect pathway starts by initiating local inflammation in the lung, the first point of impact for inhaled air pollution components, which then gains systemic character. This systemic inflammation, in the form of circulating cytokines and pro-inflammatory mediators (such as tumor necrosis factor-alpha—TNFα; interleukins 1α/β and 6—IL-1α/β/6; and cyclooxygenase 2—COX-2), spreads to the brain where it promotes activation of microglia (resident macrophages). Both of these pathways result in neuroinflammation and cerebral oxidative stress, which promotes the development and progression of neurodegenerative diseases. Created with BioRender.com.
Figure 3. Mechanisms of air pollution-induced neuroinflammation and oxidative stress. Air pollution can cause neuroinflammation and oxidative stress through two pathways: direct and indirect. Air pollution components directly interact with the neuronal tissue in the direct pathway. This can happen by direct contact of air pollution components, such as particulate matter (PM), with the nerves in the olfactory bulb, helping them translocate deeper into the neuronal tissue. Another way is for ultrafine particles (UFP) with a diameter in the nanometer range to enter the systemic circulation after inhalation, causing disturbance of the blood–brain barrier (BBB) and reaching the cerebral tissue. The indirect pathway starts by initiating local inflammation in the lung, the first point of impact for inhaled air pollution components, which then gains systemic character. This systemic inflammation, in the form of circulating cytokines and pro-inflammatory mediators (such as tumor necrosis factor-alpha—TNFα; interleukins 1α/β and 6—IL-1α/β/6; and cyclooxygenase 2—COX-2), spreads to the brain where it promotes activation of microglia (resident macrophages). Both of these pathways result in neuroinflammation and cerebral oxidative stress, which promotes the development and progression of neurodegenerative diseases. Created with BioRender.com.
Antioxidants 13 00266 g003
Figure 4. Mechanisms of noise-induced neuroinflammation and oxidative stress. Noise can cause neuroinflammation and oxidative stress through two pathways: direct and indirect. In the direct pathway, noise causes inner ear damage, hearing loss, and sleep deprivation, while in the indirect pathway, it creates a state of annoyance and disturbs sleep and communication. Both of these pathways activate the stress response through the activation of the hypothalamic–pituitary–adrenal (HPA) axis and the activation of the sympathetic nervous system (SNS). The activation of the HPA axis results in the release of CRH (corticotropin-releasing hormone) and ACTH (adrenocorticotropic hormone), further promoting the release of glucocorticoids from the adrenal cortex. The activation of the SNS results in secretion of catecholamines from the adrenal medulla. Both the HPA axis and the SNS activation result in systemic inflammation signaling through macrophage activation and cytokine release (such as tumor necrosis factor-alpha—TNFα; and interleukin 6/1β—IL-6/1β), further activating neuroinflammation and oxidative stress. Abbreviations: NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), dAAC (dorsal anterior cingulate cortex), mPFC (medial prefrontal cortex). Adapted from [218]. Created with BioRender.com.
Figure 4. Mechanisms of noise-induced neuroinflammation and oxidative stress. Noise can cause neuroinflammation and oxidative stress through two pathways: direct and indirect. In the direct pathway, noise causes inner ear damage, hearing loss, and sleep deprivation, while in the indirect pathway, it creates a state of annoyance and disturbs sleep and communication. Both of these pathways activate the stress response through the activation of the hypothalamic–pituitary–adrenal (HPA) axis and the activation of the sympathetic nervous system (SNS). The activation of the HPA axis results in the release of CRH (corticotropin-releasing hormone) and ACTH (adrenocorticotropic hormone), further promoting the release of glucocorticoids from the adrenal cortex. The activation of the SNS results in secretion of catecholamines from the adrenal medulla. Both the HPA axis and the SNS activation result in systemic inflammation signaling through macrophage activation and cytokine release (such as tumor necrosis factor-alpha—TNFα; and interleukin 6/1β—IL-6/1β), further activating neuroinflammation and oxidative stress. Abbreviations: NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), dAAC (dorsal anterior cingulate cortex), mPFC (medial prefrontal cortex). Adapted from [218]. Created with BioRender.com.
Antioxidants 13 00266 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kuntić, M.; Hahad, O.; Münzel, T.; Daiber, A. Crosstalk between Oxidative Stress and Inflammation Caused by Noise and Air Pollution—Implications for Neurodegenerative Diseases. Antioxidants 2024, 13, 266. https://doi.org/10.3390/antiox13030266

AMA Style

Kuntić M, Hahad O, Münzel T, Daiber A. Crosstalk between Oxidative Stress and Inflammation Caused by Noise and Air Pollution—Implications for Neurodegenerative Diseases. Antioxidants. 2024; 13(3):266. https://doi.org/10.3390/antiox13030266

Chicago/Turabian Style

Kuntić, Marin, Omar Hahad, Thomas Münzel, and Andreas Daiber. 2024. "Crosstalk between Oxidative Stress and Inflammation Caused by Noise and Air Pollution—Implications for Neurodegenerative Diseases" Antioxidants 13, no. 3: 266. https://doi.org/10.3390/antiox13030266

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