Next Article in Journal
A Highly Sensitive Molecular Technique for RNA Virus Detection
Previous Article in Journal
Thirteen Ovary-Enriched Genes Are Individually Not Essential for Female Fertility in Mice
Previous Article in Special Issue
GPR55 Inactivation Diminishes Splenic Responses and Improves Neurological Outcomes in the Mouse Ischemia/Reperfusion Stroke Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut– and Lung–Brain Axis

by
Alexandria Ruggles
and
Corinne Benakis
*
Institute for Stroke and Dementia Research, University Hospital, LMU Munich, 81337 Munich, Germany
*
Author to whom correspondence should be addressed.
Cells 2024, 13(10), 803; https://doi.org/10.3390/cells13100803
Submission received: 2 April 2024 / Revised: 24 April 2024 / Accepted: 2 May 2024 / Published: 8 May 2024
(This article belongs to the Special Issue Stroke Immunology: Mechanisms and Therapeutic Prospects)

Abstract

:
Recent evidence indicates that exposure to environmental toxins, both short-term and long-term, can increase the risk of developing neurological disorders, including neurodegenerative diseases (i.e., Alzheimer’s disease and other dementias) and acute brain injury (i.e., stroke). For stroke, the latest systematic analysis revealed that exposure to ambient particulate matter is the second most frequent stroke risk after high blood pressure. However, preclinical and clinical stroke investigations on the deleterious consequences of environmental pollutants are scarce. This review examines recent evidence of how environmental toxins, absorbed along the digestive tract or inhaled through the lungs, affect the host cellular response. We particularly address the consequences of environmental toxins on the immune response and the microbiome at the gut and lung barrier sites. Additionally, this review highlights findings showing the potential contribution of environmental toxins to an increased risk of stroke. A better understanding of the biological mechanisms underlying exposure to environmental toxins has the potential to mitigate stroke risk and other neurological disorders.

1. Introduction

Over the past two centuries, society has undergone profound changes driven by the rise of urbanization and industrialization. Advances in society have introduced various hazards directly impacting human health [1]. Among these hazards are biological and chemical pollutants, extreme climate conditions, gas emissions, and heavy metal accumulation, all of which have been linked to an increased risk of various health conditions, not only respiratory complications, but also endocrine disruptions, cancer, cardiovascular, and neurological disorders [2,3,4,5]. Globally, stroke is a highly prevalent neurological disease and a main contributor to long-term disabilities, remaining the second leading cause of death [6]. Importantly, stroke is no longer a disease of just the elderly; over the past two decades, the prevalence and incidence rates of stroke in people under the age of seventy have increased [7]. This phenomenon has been attributed in part to increased risk factors, including poor diet, smoking habits, and chronic exposure to stressors. Additionally, emerging evidence suggests that environmental toxins, including household air pollution, ambient particulate matter, and exposure to heavy metals, contribute to the global stroke burden [8,9]. In fact, the most recent systematic analysis shows that among stroke risk factors, ambient particulate matter exposure is second only to high blood pressure in terms of prevalence [6].
Human exposure to environmental toxins can occur through three primary routes: dietary intake, inhalation, and dermal absorption [1]. Short and long-term exposure to toxins has shown bioaccumulation in different tissues, including the gut, lungs, and central nervous system [10]. In consequence, these toxins can interfere with cellular processes, inducing neurotoxicity and altering the host immune response [11]. Indeed, experimental studies have demonstrated that exposure to a range of environmental toxins can disrupt host homeostasis by impairing the cellular response of the immune system, particularly at barrier sites such as the gut and lungs [12,13,14]. This disruption is further reinforced by recent findings that environmental contaminants also significantly alter the gut microbiome’s composition and function [12,15], suggesting exposure to such toxins has an impact on biological interfaces. Because recent studies highlighted the critical role of the gut–brain axis, in neurological disorders [16] and given that the primary route of exposure to environmental toxins is through food consumption and inhalation, the lung and intestinal microbiota could be significant intermediaries in the influence of these chemical pollutants on neurological disorders.
Despite growing evidence linking environmental toxins to stroke risk factors, the field lacks a thorough mechanistic understanding of the precise pathways through which these toxins influence stroke pathobiology. Research on this topic is limited, leaving a gap in our knowledge of the long-term effects of toxin exposure and the synergistic impact of multiple pollutants on brain diseases. Additionally, socioeconomic status, geographical region, sex difference, and pre-existing conditions that may affect individual susceptibility to these toxins are not well characterized, further complicating risk assessment and mitigation strategies. Bridging these gaps is crucial for developing targeted interventions and refining policy regulations to reduce stroke incidence linked to environmental neurotoxins. Therefore, this review aims to examine the biological mechanisms associated with environmental toxins and deleterious consequences on the host. To do so, it explores the impact of these toxins on the lungs and gut, especially delving into the cellular pathways and microbiome-related changes, and further emphasizing their potential contribution to an increased risk of stroke onset and its outcomes. This narrative review aims to raise awareness of the importance of the gut- and lung-brain axis linked to exposure to environmental toxins and stroke risks, which we believe is relevant for other neurological and neuroinflammatory diseases.

2. Environmental Toxins and Stroke Risk

Environmental toxicants are either airborne pollutants or chemical toxins that have been shown to cause systemic inflammation and ultimately becoming neurotoxic [17]. These environmental toxins, grouped under the name of xenobiotics, are nearly impossible to evade in today’s industrialized world and include particulate matter, dioxins, persistent organic pollutants (POPs), and polycyclic aromatic hydrocarbons (PAHs) [18]. Chronic interactions with these substances can incrementally impact human health, leading to the emergence of disease through different exposure pathways. Importantly, exposure to environmental toxins has been shown to increase the occurrence, risk, and development of neurological disorders, including Alzheimer’s disease and dementia, and be a possible risk factor for Parkinson’s disease, and it impacts neuronal development [19,20]. The neuroinflammatory response is a key common component of the pathobiology of these neurological diseases [21,22]. Neuroinflammation is not only observed from repeated or chronic exposure periods but can be induced even after a single insult [23], demonstrating the need to monitor toxin exposure and prolongation to minimize potential threats to human health. Although observational and prospective studies have offered valuable insights into the correlation between exposure to environmental toxins and the onset of various neurological and neuroinflammatory disorders and neurodegenerative diseases [19,24], there is a lack of research elucidating the mechanisms and pathways underlying the role of environmental toxins in neurological conditions such as stroke.
In this section, we discuss pertinent environmental toxins and their metabolic pathways that lead to host toxicity, particularly in the context of stroke. The categories of toxins reviewed include (1) persistent organic pollutants (POPs), (2) per- and polyfluorinated substances (PFASs), (3) air pollutants, and (4) micro- and nanoplastics. Table 1 summarizes experimental studies that demonstrated the biological consequences of environmental toxins on the host cellular response and, when available, the implications for stroke.

2.1. Persistent Organic Pollutants

Persistent organic pollutants (POPs) represent a broad class of toxins abundant in the environment and formed as byproducts from industrial or chemical processes, such as waste incineration, vehicle exhaust, and cigarette smoke [50,51,52,53]. Due to their physical properties, these pollutants resist natural degradation processes and persist in the environment. Common POPs include organochlorine pesticides (OCPs), polychlorinated biphenyls (PCBs), polycyclic aromatic hydrocarbons (PAHs), dioxins, and dibenzofurans [50]. These persistent pollutants are ubiquitous in atmospheric, terrestrial, and aquatic environments [52]. Due to their lipophilic properties, POPs accumulate in lipid-rich tissues and organs, including the liver, blood, brain, adipose tissue, and kidney, and bioaccumulate in mammals, humans, fish, and invertebrates [54,55].

2.1.1. Polycyclic Aromatic Hydrocarbons

Organic pollutants characterized by two or more fused benzene rings are classified as PAHs. Biodegradation or detoxification of PAHs by the host involves several enzymatic pathways, including the binding of xenobiotics to the aryl hydrocarbon receptor (AhR) and subsequent expression of P450 (CYP) genes [56,57]. Most of the metabolic products of xenobiotics are excreted from the body, while a significant amount fails to be detoxified and is retained in the body. Indeed, CYP-mediated biotransformation of various POPs can result in toxic compounds that either elicit a mutagenic or genotoxic effect [58,59,60]. For instance, the highly toxic environmental chemical TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) has been shown to alter several cellular and tissue responses, leading to neurotoxicity, immunotoxicity, reproductive toxicity, and the formation of tumors [60]. Mechanistically, inhibition of AhR activity, using AhR-deficient mice, prevented the carcinogenic effect of the PAHs: TCDD and benzo(a)pyrene (BaP) [61,62], highlighting the functional role of AhR in mediating deleterious effects of environmental toxins.
Epidemiological studies have shown that PAHs, including BaP, have been associated with hypertension [63] and cardiovascular diseases [64], both known to increase ischemic stroke risks [65]. Regarding the association between stroke and exposure to PAHs, Rahman et al. found that higher levels of specific urinary PAHs were positively correlated with increased odds of stroke onset in men and women [66]. In experimental studies, BaP exposure has been associated with high blood pressure and vasoconstriction (Table 1). Indeed, following both acute and chronic BaP exposure in animal models, BaP increased blood pressure, exceeding 20% of baseline values [65,66]. Moreover, chronic treatment with BaP administered by oral gavage in mice susceptible to atherosclerosis (ApoE-deficient mice) increased the size of atherosclerotic plaques and a local inflammatory response characterized by the infiltration of T lymphocytes and macrophages [67]. Exposure to BaP has a direct effect on brain function since BaP and its metabolites can accumulate in lipid-rich tissues such as the brain [68] and have been shown to impair the blood–brain barrier function [69]. Tanaka et al. demonstrated that exposure to PAHs altered stroke outcomes in mice after intranasal exposure, which induced edema in the brain and impaired locomotor functions in rats [70]. These findings suggest that despite the liver’s primary role in detoxifying xenobiotics [71], there exists a tissue-specific vascular response, potentially highlighting the link between certain PAHs and stroke pathophysiology. The complex interplay between the increased risk of cardiovascular complications linked to PAH exposure and stroke occurrence and outcomes underscores the necessity for deeper insights into how environmental toxin exposure heightens the risk of stroke onset, mortality, and morbidity.

2.1.2. Polychlorinated Biphenyls

Despite the implementation of new regulations to ban the production of some PCBs in 1978 [72], the lasting repercussions of industrial manufacturing continue to persist and have lasting effects on human health. Even decades after the ban of PCBs, Raffetti et al. identified correlations between the fasting blood serum levels of several PCB compounds and the health status of middle-aged individuals residing in a heavily polluted area with a history of PCB production. Significantly, specific PCB congeners, such as 138, 153, and 180, which are considered to be mid- and highly chlorinated and are more resistant to being metabolized, were found to be associated with the onset of hypertension based on fasting blood serum levels [73]. Elevated serum levels of PCBs have also been positively associated with increased blood pressure [74] and an increased risk for ischemic stroke in both men and women [75]. Moreover, as shown in Table 1, passive dietary consumption of PCBs has also been demonstrated to increase the risk of stroke onset, specifically in women [31]. Mechanistically, PCBs, which are ligands of AhR, activate the nuclear transcription factor nuclear factor- κB (NF-κB) pathway, inducing oxidative stress, production of inflammatory cytokines, and upregulation of the expression of inflammatory genes [76,77]. This indicates the role of PCBs in inducing a pro-inflammatory response.

2.2. Per- and Polyfluorinated Substances

The production of PFASs began just less than a century ago [78]. Due to their structural and chemical properties, these chemicals are highly resistant to degradation and persist in the environment for extended periods of time. PFAS have been extensively utilized in household and industrial products, including non-stick cookware, medical devices, insulating packaging material, cosmetics, and textiles [79]. The broad utilization of PFAS across various sectors has resulted in widespread accumulation in both the environment and animals, raising concerns regarding their potential impact and health hazards for individuals [78,80]. Ingestion and inhalation are the two primary routes of human exposure to PFAS through dietary consumption, mainly from drinking water [81,82]. It has been reported that the average half-life in human blood serum for some of these substances is up to 8.5 years [83]. Several studies have shown exposure to PFAS is positively associated with an increased risk for hypertension [84]. Despite elevated levels of cholesterol and hypertension observed in the previous studies, which are known risk factors for cardiovascular disease and stroke [85], there have been conflicting results regarding whether exposure to PFAS increases the risk of stroke onset. Simpson et al. found a modest association with stroke incidence in individuals living near a PFAS chemical plant [86], while Feng et al. demonstrated that higher PFAS levels increased the risk for cardiovascular disease and stroke, particularly in males [87]. Another study showed that individuals with higher serum levels of PFAS exhibited dyslipidemia, yet no significant association between stroke and myocardial infarction [88].
Experimental research indicates that exposure to members of the PFAS family induces immunotoxicity by activating signaling pathways, including the NF-kB pathway. This activation triggers the generation of reactive oxygen species (ROS) and facilitates the upregulation of cytokine production [89]. Moreover, tissue inflammation after exposure to PFAS is mediated by several signaling pathways such as peroxisome proliferator-activated receptor α (PPARα) and Janus kinase 2/signal transducer and activator of transcription 3 [90,91]. Although these signaling pathways are activated following experimental stroke [90,91], the exact mechanistic pathways linking PFAS exposure to influencing the risk for stroke remain elusive. Additionally, a new hypothesis has emerged that exposure to PFAS may target the cardiovascular system by activating platelet aggregation and adhesion, promoting thrombosis [92]. A deeper understanding of the health hazards associated with PFAS exposure and the underlying mechanistic pathways could provide insights into how these compounds contribute to stroke risk.

2.3. Air Pollutants

The World Health Organization (WHO) reports that a significant portion of premature deaths linked to outdoor air pollution is due to cardiovascular diseases and ischemic stroke (37%), and respiratory infections (23%) (https://www.who.int/news-room/fact-sheets/detail/ambient-(outdoor)-air-quality-and-health; accessed on 22 January 2024). More than a hundred hazardous air pollutants originating from activities including oil and gas production, battery manufacturing, iron and steel forging, plastic manufacturing, and wood preserving are monitored by The Environmental Protection Agency (EPA) monitors (https://www.epa.gov/haps/area-sources-urban-air-toxics; accessed on 22 January 2024). Both short-term and long-term exposure to ambient air pollution are now recognized as a modifiable risk factor contributing to the onset and mortality of ischemic stroke [93]. While everyone is affected by air pollution, people living in urban regions are exposed to a higher concentration of air pollutants compared to those living in rural areas [94]. Consequently, urban residents face an elevated risk for the development of health complications as a result of prolonged exposure and concentration.
According to the National Institute of Health (NIH), air pollution is a mixture of hazardous compounds, which include PAHs, ozone, particulate matter, noxious gases, nitrogen oxides, and sulfur oxides (https://www.niehs.nih.gov/health/topics/agents/air-pollution; accessed on 22 January 2024). More specifically, particulate matter detected in air pollution is characterized by its aerodynamic diameter. Particulate matter with a diameter of ≤10 µm (PM10) is considered coarse, ≤2.5 µm (PM2.5) is considered fine, and ≤0.1 µm (PM0.1) is considered ultrafine [95,96]. Mechanistically, when particulate matter is inhaled, it can accumulate along the respiratory tract. PM10 typically accumulates in the nasal cavity and trachea, whereas PM2.5 can penetrate deeper into the lungs, primarily targeting the bronchioles. The finest particles, those smaller than 2.5 µm, target the alveoli and can even diffuse into the bloodstream [97]. Particulate matter can enter the circulation and can traffic to the brain, acting as a pro-inflammatory stimulus and triggering chronic inflammation [98]. This sustained brain inflammation has been characterized as a consequence of continuous particulate matter exposure inducing ROS production, microglial activation, neuroinflammation, and neuronal damage [23,99,100,101].
In animal models, passive exposure to air pollution induced significant alterations in the lung epithelium, characterized by increased mucus secretion and morphological changes in the pulmonary cell lining. These changes manifested as an augmented presence of microvilli and a reduction in the population of ciliated cells within the lung tissue [102]. In a controlled laboratory setting, individuals without pre-existing health conditions who were exposed to a mixture of common air pollutants, including sulfur dioxide, sulfate aerosols, and ozone, demonstrated an increased risk of developing pulmonary dysfunction, including a decrease in forced expiratory volume and vital capacity [103]. In an epidemiological study investigating the association between PM2.5 exposure and respiratory and cardiovascular complications, findings revealed that women were more likely to be hospitalized for such complications compared to men when exposed to PM2.5 [104].
Numerous epidemiological studies have shown that exposure to PM2.5 and PM10 is associated with a higher ischemic stroke incidence rate and mortality compared to studies investigating ultrafine ambient pollutants [93,105,106,107]. Szyszkowicz et al. observed a correlation between low-level exposure to ambient air pollution and an increased number of visits to the hospital for hypertension [108], which may be an indicator of higher stroke risk for those individuals exposed to ambient air pollution. These findings highlight that air contaminants may compromise an already vulnerable region of the lungs, potentially increasing the risk of lung infections, hospitalizations associated with lung complications, and post-stroke pneumonia, and stroke outcomes should be further explored.

2.4. Nano- and Microplastics

The use of plastic globally has continued to increase at a staggering rate, reaching nearly 500 million tons annually. Despite policy efforts and heightened awareness of methods to recycle and dispose of plastics, less than 10% is recycled, and a quarter of the plastic is inadequately disposed of (https://www.oecd.org/environment/plastic-pollution-is-growing-relentlessly-as-waste-management-and-recycling-fall-short.htm; accessed on 14 March 2024). Improperly disposed plastic can persist in the environment and be degraded and broken into fragmented pieces known as microplastics (particles less than 5 mm) and nanoplastics (less than 1 nm) [109,110]. Abundant in the environment, these particles can be both inhaled and ingested as they have been detected in soil, air, and drinking water [111,112]. Not only are these particles accumulating in the environment, but they are also found in several human samples, including stool, blood, and breastmilk [113,114,115]. Many of the most recent studies have investigated the effects of microplastics on animals living in aquatic environments [116,117]. However, there are relatively few studies focusing on the effects in humans. In mice, chronic exposure to micro- and nanoplastics led to the accumulation of particles in the gut [29] and perturbed gut homeostasis. In an experimental model, analysis of the gastrointestinal tract revealed that post-exposure, there was a reduction in overall microbial diversity and compromised gut architecture, characterized by a reduction in the mucosal wall lining and the absence of villi along the gastrointestinal tract [15]. In humans, a recent study showed that individuals with microplastics detected in the coronary arteries were at heightened risk for developing cardiovascular complications and development of stroke [118]. These findings suggest that exposure to micro- and nanoplastics accumulates in the host and influences diseases of the gut, heart, and brain.

3. Exposure to Environmental Toxins at Barrier Sites

Barrier sites, including the lungs, gut, and skin, act as physical barriers and immune barriers to maintain host homeostasis and protect from external threats [119,120,121]. If compromised by environmental toxins, these barrier sites have been shown to contribute to brain diseases, specifically regarding the lung–brain axis and gut–brain axis. Cellular mechanisms linking environmental toxins, barrier sites, and stroke pathobiology are discussed in the next sections.
The respiratory tract is lined with an epithelial layer consisting primarily of pseudostratified ciliated columnar cells, which function as a barrier against foreign particles. The cells lined with cilia and mucosal cells initiate a defense mechanism by generating mucus and trapping foreign particles and pathogens [122]. Exposure to environmental toxins such as particulate matter increases ROS production, compromising the epithelial barrier integrity and creating an environment where bacteria are more likely to invade [123], demonstrating the vulnerability of the barrier site. Short-term exposure to PM10 reduced the tight junction protein occludin in human and rat alveolar epithelial cells [124]. In contrast, exposure to PCB126, a dioxin-like compound, increased the expression of the tight junction proteins occludin and claudin [32]. Jang et al. found that ozone exposure also significantly impacts the structural integrity of the lung’s epithelial layer and immune response in the lungs. After exposure to ozone, the researchers observed initial epithelial cell death followed by an increased proliferation of epithelial cells coupled with an increase in neutrophils [125]. In wild-type mice, it was shown that acute exposure to ozone compromised the epithelial lining, increased epithelial damage in the lungs, and increased permeability in ST2-deficient mice [39]. Overall, these findings highlight the complex interaction between environmental toxins and the integrity of the epithelial barrier and immune regulation. While different environmental pollutants can have contrasting effects on epithelial barrier function and immune regulation, exposure may have implications for respiratory health and disease susceptibility.
In the gut, the digestive tract serves as an interface between the environment and the host, acting as a direct entry sight for pathogenic invaders, including bacteria, pathogens, and contaminated food components [126]. As a defense mechanism to prevent bacteria and pathogens from invading the tissue, specialized intestinal epithelial cells known as goblet cells produce mucus, forming a mucosal layer [127,128]. It has been shown that even within the first 24 h post-stroke, intestinal permeability is compromised, contributing to inflammation and bacterial translocation to secondary sites. The impaired permeability of the gut has been directly correlated with an increased severity of stroke outcomes [129]. Environmental toxins can also lead to altered permeability of the gut lining. A 3D model of human intestinal samples revealed that exposure to particulate matter at a concentration of 500 μg/cm2 for up to two weeks reduced the levels of zonula occludens protein 1 (ZO−1) and claudin-1 [36]. Mice acutely and chronically exposed to high concentrations of ambient PM2.5 increased the permeability of the gut barrier by reducing the expression of the tight junction protein, ZO-1, at acute and chronic time points [130]. Histological analysis of the gastric epithelium in mice deficient in the tight junction protein occludin did not compromise the structure of the epithelium but rather promoted chronic inflammation [131]. The subsequent inflammatory response in the gut activated by the upregulation of IL-17-producing T cells can increase gut permeability by disrupting the tight junction barrier [132]. One explanation for the impaired architecture of the tight junctions is that cells, as a stress mechanism in response to exposure to particulate matter, will generate ROS, which disrupt tight junction formation and increase epithelial cell permeability [35].

3.1. Lung–Brain Axis and Environmental Toxins

Recent findings suggest there is a bidirectional communication pathway involving pulmonary microbiota, their metabolites in the lungs, the central and autonomic nervous system, and the hypothalamic–pituitary–adrenal (HPA) axis, which are collectively known as the lung–brain axis [133]. While still an emerging concept in understanding the functional role of the lung–brain axis in health and disease, the lung–brain axis has been demonstrated to play an integral role in the pathology of neurological diseases. In a recent study, Hosang et al. manipulated the pulmonary microbiome using a cocktail of antibiotics, which in turn influenced the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a rat model of multiple sclerosis [134]. Further evidence supports the idea that environmental toxins can alter the lung–brain axis and may influence the likelihood of stroke onset and post-stroke outcomes. Tanaka et al. demonstrated that intranasal exposure to urban aerosols in mice can affect post-stroke recovery by augmenting neuroinflammation, activating brain resident microglial cells, and impairing motor function post-stroke [70].
Among the various complications experienced by patients following a stroke, urinary infections, and post-stroke pneumonia are the most prevalent [135]. Post-ischemic stroke not only causes site-specific damage but has been shown to impact secondary organs such as the lungs in experimental stroke models [136]. Stroke onset activates an immune cascade and consequently leads to immunosuppression in the periphery, increasing the risk of developing infections, including stroke-associated pneumonia and the patients’ 30-day mortality rate [137,138]. Exposure to air pollution has also been linked to an increased risk of bacterial lung infections, in particular bacterial pneumonia [139]. The association between stroke and lung infection highlights the need for more research unveiling the lung–brain axis (Figure 1).

3.1.1. Environmental Toxins and Immune Response in the Lungs

The lungs are one of the first organs to interact with inhaled particulate matter. Exposure to particulate matter has been shown to impair immune cell functions, modify the immune response, and disturb lung architecture. PM10 can penetrate deeper into the lungs, targeting the lung bronchioles and resulting in remodeling of lung tissue and the upregulation of immune cells [140]. Damage to lung architecture and altered immune response elevate the likelihood of lung diseases such as chronic obstructive pulmonary disease (COPD), chronic bronchitis, and respiratory tract infections due to resulting lung impairment and dysfunction post-exposure to environmental toxins [43,141,142].
Importantly, studies have shown that particulate matter can translocate to secondary organs and elicit a systemic effect. Upon inhalation, particulate matter first enters the nasal cavity and can then translocate into the brain through multiple routes, including the olfactory nerve, compromised blood–brain barrier, or damaged nasal epithelium [143]. Chronic insults from inhaled particulate matter have revealed that aging is associated with a decline in the phagocytic capacity of macrophages and accumulation of particulate matter within the lung-associated lymph nodes, ultimately altering the lung-associated lymph nodes’ follicular architecture [144]. Similarly, Samary et al. demonstrated in rats that ischemic stroke resulted in changes in the respiratory environment, impairing the phagocytic capacity of macrophages, upregulating inflammatory markers including TNF-α and IL-6 levels in the lungs, and inducing lung edema [145]. While it has been shown that exposure to particulate matter can alter the tissue and immune architecture, even in the absence of lung damage, exposure to particulate matter can influence coagulation factors and fibrinogen, bridging the connection between inhaled particulate matter and cardiovascular diseases [146]. Impaired lung function has been demonstrated to result in a higher incidence of ischemic stroke [147]. Together, this highlights the intricate connection between exposure to particulate matter and the crosstalk between the lungs and the cardiovascular system, which may influence the onset of disease.

3.1.2. Effect of Environmental Toxins on the Lung Microbiome

The pulmonary microbiome should be considered when determining the effects of environmental toxins. While it was once thought that the lungs were sterile [148], emerging evidence suggests that manipulating the pulmonary microbiota can activate immune cells in the brain and influence neurological disease outcomes [134]. Variation in bacterial taxa has been mapped along the respiratory tract from the oral cavity through the trachea and to the lungs [149]. Despite the limited number of studies investigating pulmonary microbiota, a recent study demonstrated that inhalation exposure to PM2.5 led to acute inflammation in the bronchioles, increased the number of goblet cells, and reduced the abundance of proteobacteria in the lungs [42]. Potential consequences of exposure to environmental toxins disturb mediators within the lung–brain axis. We suggest that future investigations should invest in understanding the role of the pulmonary microbiota influenced by environmental factors in stroke pathogenesis.

3.2. Gut–Brain Axis and Environmental Toxins

The gut–brain axis is a bidirectional communication pathway that signals information between the gut and the brain via the autonomic and enteric nervous system, the hypothalamic–pituitary axis, the vagal nerve, and systemic circulation [150,151]. Humans are exposed to millions of foreign substances referred to as xenobiotics directly and passively. Upon exposure, the body metabolizes and eliminates these substances, with toxic xenobiotics being transformed into less harmful derivatives before being excreted through stool, urine, perspiration, and bile [152]. Xenobiotics are also metabolized by the resident gut microbiota, which can influence bioaccumulation and toxicity [153]. However, accumulation of metabolites in the body from xenobiotics, when not excreted or eliminated, can pose significant health risks by disrupting cellular homeostasis and causing tissue damage [57]. Moreover, oral exposure to environmental toxins will not only accumulate in the digestive tract but can also enter the bloodstream through enterohepatic circulation. Consequently, this can lead to the bioaccumulation of toxic derivatives in secondary organs such as the brain [154].
Trillions of microorganisms reside within the intestinal tract, comprised of bacteria, fungi, protozoans, archaea, and viruses [155]. Metagenomic analysis of the human microbiota without underlying conditions revealed that the phyla change along the digestion tract; however, it is dominated primarily by commensal bacteria: Bacteroidetes, Firmicutes, Proteobacteria, and Actinobacteria phyla [156]. Several factors shape the composition and function of the microbiota, including diet, aging, and the environment [157,158,159]. In particular, the gut microbiota plays an integral role in training and educating the immune system and facilitating tolerance mechanisms [160]. Signaling from the brain can modulate the immune response in the gut, intestinal permeability, and bacterial–host interactions [161]. Recent studies have revealed exposure to environmental toxins shifts the relative abundance of bacteria and alters microbial profiles in animal models. Dietary exposure to heavy metals and PCBs has been shown to reduce the abundance of Proteobacteria while simultaneously increasing the presence of Bacteroidetes [25,26,30]. Chronic exposure to PAHs (BaP and TCDD) also shifts the bacterial ratios, increasing the relative abundance of Bacteroidetes while reducing the abundance of Firmicutes [12,162]. Interestingly, long-term inhalation exposure to PM2.5 altered the ratio of bacteria in mice, marked by a significant reduction in Akkermensia and Firmicutes, and significantly increased the Bacteroidetes [130], demonstrating the impact of inhaled air pollutants on the gut microbiota. Collectively, these experimental findings highlight the impact of environmental toxins on the gut microbiota, yet the impact of alterations of the composition in the context of neurological diseases such as stroke risk and outcomes remain unknown.

Exposure to Environmental Toxins and Stroke Risk

Stroke onset triggers an immune response in both the brain and the periphery. It has been established that the occlusion of a cerebral artery causes shear stress in the brain, altering the blood–brain barrier permeability and limiting the amount of glucose and oxygen to the brain tissue, leading to neuronal cell death [163]. This acute response triggers an immune cascade signaling to resident immune cells to activate the innate and adaptive immune response [164]. Inflammation after stroke extends beyond the site of injury and propagates a systemic effect, greatly impacting secondary organs in the periphery [165]. Immune cells have been shown to traffic from peripheral organs, such as the gut and brain, post-stroke to mediate inflammation [166,167]. Because neuroinflammation plays an essential role in the pathobiology of stroke, the effect of environmental toxins on the immune system warrants further investigation.
Several preclinical and clinical studies have demonstrated stroke induces dysbiosis of the gut microbiota, which can influence the severity of stroke outcomes [168,169,170,171]. Singh et al. found that in an experimental stroke model, the expansion of Bacteroidetes is associated with a proinflammatory response marked by the increased expression of IFN-γ+ and IL-17 cytokines produced by T helper cells [168]. Moreover, prior to stroke onset, mice treated with Ampicillin shifted the microbial profile, leading to the expansion of Proteobacteria and Lactobacilliales and a reduction in Bacteroidetes, which were deemed to be neuroprotective by reducing the lesion size after stroke [172]. These changes in the microbiota composition induced an expansion of regulatory T cells in the gut and down-regulation of IL-17 γδT cells, which prevented their trafficking into the brain and thereby prevented the formation of the primary ischemic damage [166]. Together, these studies reveal the intricate relationship between the host microbiota, the gut immune system, and stroke pathobiology. Because environmental toxin exposure induces alterations in the microbiome’s relative abundance, inducing a proinflammatory environment, this complex interplay may influence the immune response and, subsequently, impact stroke outcomes. However, further research is needed to understand the detailed mechanisms and implications of these interactions and how exposure to xenobiotics can influence stroke outcomes.
Inhalation of particulate matter can directly impact the lungs, but it can also indirectly induce inflammation in the gut [34]. Experimental models have shown that inhalation of particulate matter alters the bacterial ratio, as previously mentioned [130], but it also has an impact on the immune cell reservoir in the gut. The authors demonstrated that exposure to air pollution in dense urban regions for two weeks upregulated the expression of inflammatory cytokine production in the colon [34]. These findings highlight that environmental toxins exert a systemic effect at barrier sites, which can disrupt immune cell reservoirs in the periphery. Understanding how environmental toxins shape the immune compartments in peripheral organs and potentially influence the outcomes of stroke could further reveal the complexity of these interactions and improve our understanding of the impact of environmental factors on health outcomes.

4. Conclusions and Outlook

Reducing the incidence of strokes requires a comprehensive approach, including addressing exposure to environmental toxins. Prevention requires the implementation of new laws and regulations targeting exposure limits and the production of hazardous material to minimize exposure. Concurrently, detection strategies, on the other hand, provide a measure of an individual’s risk. Measuring metabolite concentrations in urine samples can be used to monitor toxin exposure [173]. Whereas prevention and detection strategies have long-term implications, immediate measures are required to counter-effect the deleterious consequences of environmental toxin exposure for stroke.
To date, there are limited studies understanding how environmental toxins act to mediate their potent effects contributing to stroke development. In this review, we examined numerous clinical, experimental, and epidemiological studies that have demonstrated that exposure to toxins via ingestion and inhalation disrupts cellular homeostasis, in particular the immune system and microbiome, and exerts a systemic effect on the gut, lungs, and brain. In addition, we highlighted how environmental toxins could increase the risk of stroke or exacerbate stroke outcomes.
By understanding the mechanisms of action related to environmental toxin exposure on the gut and lung–brain axis, we could limit the impact of environmental toxins-induced toxicity and associated brain diseases. To develop new therapeutic or preventive strategies, we need future research to address the systemic impact (gut and lungs) of environmental toxins and their contribution to stroke onset. For instance, dietary interventions rich in fiber and polyphenols have been demonstrated to play a role in reducing the bioavailability of xenobiotics by absorbing and minimizing their uptake [174,175]. Characterization of the gut and lung microbiomes associated with immune changes may be another approach to mitigate the effects of environmental toxin exposure. This would lead to new therapeutic strategies targeting microbiome–host interactions to counteract the deleterious effects of environmental pollutant exposure for brain diseases.
In conclusion, this review addresses how environmental toxins induce a systemic effect that may play a critical role in stroke pathogenesis. To our knowledge, there are limited studies that address the impact of environmental toxins in the context of stroke pathogenesis. While this review article provides insights into the role of environmental factors as risk factors for stroke, several gaps in the literature remain. Firstly, the precise mechanisms through which environmental toxins contribute to stroke pathogenesis remain unclear. Secondly, the exploration of sex differences in susceptibility to environmental toxins and their subsequent impact on stroke risk remains largely unexplored. Furthermore, there is a need for research to elucidate the systemic effects of environmental toxin exposure on stroke outcomes. Addressing these gaps will be crucial for advancing a more comprehensive understanding of environmental influences on stroke pathogenesis.

Author Contributions

Conceptualization, A.R. and C.B.; writing—original draft preparation, A.R.; writing—review and editing, A.R. and C.B; visualization, A.R.; supervision, C.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Manisalidis, I.; Stavropoulou, E.; Stavropoulos, A.; Bezirtzoglou, E. Environmental and Health Impacts of Air Pollution: A Review. Front. Public Health 2020, 8, 14. [Google Scholar] [CrossRef]
  2. Iwasaki, M.; Itoh, H.; Sawada, N.; Tsugane, S. Exposure to Environmental Chemicals and Cancer Risk: Epidemiological Evidence from Japanese Studies. Genes Environ. 2023, 45, 10. [Google Scholar] [CrossRef]
  3. Béjot, Y.; Reis, J.; Giroud, M.; Feigin, V. A Review of Epidemiological Research on Stroke and Dementia and Exposure to Air Pollution. Int. J. Stroke 2018, 13, 687–695. [Google Scholar] [CrossRef]
  4. Kurt, O.K.; Zhang, J.; Pinkerton, K.E. Pulmonary Health Effects of Air Pollution. Curr. Opin. Pulm. Med. 2016, 22, 138–143. [Google Scholar] [CrossRef]
  5. Yang, A.-M.; Lo, K.; Zheng, T.-Z.; Yang, J.-L.; Bai, Y.-N.; Feng, Y.-Q.; Cheng, N.; Liu, S.-M. Environmental Heavy Metals and Cardiovascular Diseases: Status and Future Direction. Chronic Dis. Transl. Med. 2020, 6, 251–259. [Google Scholar] [CrossRef]
  6. Steinmetz, J.D.; Seeher, K.M.; Schiess, N.; Nichols, E.; Cao, B.; Servili, C.; Cavallera, V.; Cousin, E.; Hagins, H.; Moberg, M.E.; et al. Global, Regional, and National Burden of Disorders Affecting the Nervous System, 1990–2021: A Systematic Analysis for the Global Burden of Disease Study 2021. Lancet Neurol. 2024, 4, 344–381. [Google Scholar] [CrossRef]
  7. Krishnamurthi, R.V.; Moran, A.E.; Feigin, V.L.; Barker-Collo, S.; Norrving, B.; Mensah, G.A.; Taylor, S.; Naghavi, M.; Forouzanfar, M.H.; Nguyen, G.; et al. Stroke Prevalence, Mortality and Disability-Adjusted Life Years in Adults Aged 20–64 Years in 1990-2013: Data from the Global Burden of Disease 2013 Study. Neuroepidemiology 2015, 45, 190–202. [Google Scholar] [CrossRef]
  8. Feigin, V.L.; Brainin, M.; Norrving, B.; Martins, S.; Sacco, R.L.; Hacke, W.; Fisher, M.; Pandian, J.; Lindsay, P. World Stroke Organization (WSO): Global Stroke Fact Sheet 2022. Int. J. Stroke 2021, 17, 18–29. [Google Scholar] [CrossRef]
  9. Feigin, V.L.; Roth, G.A.; Naghavi, M.; Parmar, P.; Krishnamurthi, R.; Chugh, S.; Mensah, G.A.; Norrving, B.; Shiue, I.; Ng, M.; et al. Global Burden of Stroke and Risk Factors in 188 Countries, during 1990–2013: A Systematic Analysis for the Global Burden of Disease Study 2013. Lancet Neurol. 2016, 15, 913–924. [Google Scholar] [CrossRef]
  10. Genuis, S.J.; Kelln, K.L. Toxicant Exposure and Bioaccumulation: A Common and Potentially Reversible Cause of Cognitive Dysfunction and Dementia. Behav. Neurol. 2015, 2015, 620143. [Google Scholar] [CrossRef]
  11. Cresto, N.; Forner-Piquer, I.; Baig, A.; Chatterjee, M.; Perroy, J.; Goracci, J.; Marchi, N. Pesticides at Brain Borders: Impact on the Blood-Brain Barrier, Neuroinflammation, and Neurological Risk Trajectories. Chemosphere 2023, 324, 138251. [Google Scholar] [CrossRef]
  12. Ribière, C.; Peyret, P.; Parisot, N.; Darcha, C.; Déchelotte, P.J.; Barnich, N.; Peyretaillade, E.; Boucher, D. Oral Exposure to Environmental Pollutant Benzo[a]Pyrene Impacts the Intestinal Epithelium and Induces Gut Microbial Shifts in Murine Model. Sci. Rep-UK 2016, 6, 31027. [Google Scholar] [CrossRef]
  13. Yang, Q.; Dai, H.; Cheng, Y.; Wang, B.; Xu, J.; Zhang, Y.; Chen, Y.; Xu, F.; Ma, Q.; Lin, F.; et al. Oral Feeding of Nanoplastics Affects Brain Function of Mice by Inducing Macrophage IL-1 Signal in the Intestine. Cell Rep. 2023, 42, 112346. [Google Scholar] [CrossRef]
  14. Michaudel, C.; Fauconnier, L.; Julé, Y.; Ryffel, B. Functional and Morphological Differences of the Lung upon Acute and Chronic Ozone Exposure in Mice. Sci. Rep. 2018, 8, 10611. [Google Scholar] [CrossRef]
  15. Qiao, J.; Chen, R.; Wang, M.; Bai, R.; Cui, X.; Liu, Y.; Wu, C.; Chen, C. Perturbation of Gut Microbiota Plays an Important Role in Micro/Nanoplastics-Induced Gut Barrier Dysfunction. Nanoscale 2021, 13, 8806–8816. [Google Scholar] [CrossRef]
  16. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  17. Fuller, R.; Landrigan, P.J.; Balakrishnan, K.; Bathan, G.; Bose-O’Reilly, S.; Brauer, M.; Caravanos, J.; Chiles, T.; Cohen, A.; Corra, L.; et al. Pollution and Health: A Progress Update. Lancet Planet. Health 2022, 6, e535–e547. [Google Scholar] [CrossRef]
  18. Thompson, L.A.; Darwish, W.S. Environmental Chemical Contaminants in Food: Review of a Global Problem. J. Toxicol. 2019, 2019, 2345283. [Google Scholar] [CrossRef]
  19. Nabi, M.; Tabassum, N. Role of Environmental Toxicants on Neurodegenerative Disorders. Front. Toxicol. 2022, 4, 837579. [Google Scholar] [CrossRef]
  20. Lanphear, B.P. The Impact of Toxins on the Developing Brain. Annu. Rev. Public Health 2015, 36, 1–20. [Google Scholar] [CrossRef]
  21. Iadecola, C.; Anrather, J. The Immunology of Stroke: From Mechanisms to Translation. Nat. Med. 2011, 17, 796–808. [Google Scholar] [CrossRef] [PubMed]
  22. Kwon, H.S.; Koh, S.-H. Neuroinflammation in Neurodegenerative Disorders: The Roles of Microglia and Astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  23. Block, M.L.; Calderón-Garcidueñas, L. Air Pollution: Mechanisms of Neuroinflammation and CNS Disease. Trends Neurosci. 2009, 32, 506–516. [Google Scholar] [CrossRef] [PubMed]
  24. Cannon, J.R.; Greenamyre, J.T. The Role of Environmental Exposures in Neurodegeneration and Neurodegenerative Diseases. Toxicol. Sci. 2011, 124, 225–250. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, S.; Deng, X.; Li, Z.; Zhou, W.; Wang, G.; Zhan, J.; Hu, B. Environmental Cadmium Exposure Alters the Internal Microbiota and Metabolome of Sprague–Dawley Rats. Front. Vet. Sci. 2023, 10, 1219729. [Google Scholar] [CrossRef] [PubMed]
  26. Li, X.; Brejnrod, A.D.; Ernst, M.; Rykær, M.; Herschend, J.; Olsen, N.M.C.; Dorrestein, P.C.; Rensing, C.; Sørensen, S.J. Heavy Metal Exposure Causes Changes in the Metabolic Health-Associated Gut Microbiome and Metabolites. Environ. Int. 2019, 126, 454–467. [Google Scholar] [CrossRef]
  27. Ersbøll, A.K.; Monrad, M.; Sørensen, M.; Baastrup, R.; Hansen, B.; Bach, F.W.; Tjønneland, A.; Overvad, K.; Raaschou-Nielsen, O. Low-level exposure to arsenic in drinking water and incidence rate of stroke: A cohort study in Denmark. Environ. Int. 2018, 120, 72–80. [Google Scholar] [CrossRef] [PubMed]
  28. Tamargo, A.; Molinero, N.; Reinosa, J.J.; Alcolea-Rodriguez, V.; Portela, R.; Bañares, M.A.; Fernández, J.F.; Moreno-Arribas, M.V. PET microplastics affect human gut microbiota communities during simulated gastrointestinal digestion, first evidence of plausible polymer biodegradation during human digestion. Sci. Rep. 2022, 12, 528. [Google Scholar] [CrossRef] [PubMed]
  29. Deng, Y.; Zhang, Y.; Lemos, B.; Ren, H. Tissue accumulation of microplastics in mice and biomarker responses suggest widespread health risks of exposure. Sci. Rep. 2017, 7, 46687. [Google Scholar] [CrossRef]
  30. Choi, J.J.; Eum, S.Y.; Rampersaud, E.; Daunert, S.; Abreu, M.T.; Toborek, M. Exercise Attenuates PCB-Induced Changes in the Mouse Gut Microbiome. Environ. Health Perspect. 2013, 121, 725–730. [Google Scholar] [CrossRef]
  31. Bergkvist, C.; Kippler, M.; Larsson, S.C.; Berglund, M.; Glynn, A.; Wolk, A.; Åkesson, A. Dietary Exposure to Polychlorinated Biphenyls Is Associated with Increased Risk of Stroke in Women. J. Intern. Med. 2014, 276, 248–259. [Google Scholar] [CrossRef] [PubMed]
  32. Petriello, M.C.; Hoffman, J.B.; Vsevolozhskaya, O.; Morris, A.J.; Hennig, B. Dioxin-like PCB 126 Increases Intestinal Inflammation and Disrupts Gut Microbiota and Metabolic Homeostasis. Environ. Pollut. 2018, 242, 1022–1032. [Google Scholar] [CrossRef] [PubMed]
  33. Phillips, M.C.; Dheer, R.; Santaolalla, R.; Davies, J.M.; Burgueño, J.; Lang, J.K.; Toborek, M.; Abreu, M.T. Intestinal exposure to PCB 153 induces inflammation via the ATM/NEMO pathway. Toxicol. Appl. Pharmacol. 2018, 339, 24–33. [Google Scholar] [CrossRef] [PubMed]
  34. Vignal, C.; Pichavant, M.; Alleman, L.Y.; Djouina, M.; Dingreville, F.; Perdrix, E.; Waxin, C.; Alami, A.O.; Gower-Rousseau, C.; Desreumaux, P.; et al. Effects of Urban Coarse Particles Inhalation on Oxidative and Inflammatory Parameters in the Mouse Lung and Colon. Part. Fibre Toxicol. 2017, 14, 46. [Google Scholar] [CrossRef] [PubMed]
  35. Mutlu, E.A.; Engen, P.A.; Soberanes, S.; Urich, D.; Forsyth, C.B.; Nigdelioglu, R.; Chiarella, S.E.; Radigan, K.A.; Gonzalez, A.; Jakate, S.; et al. Particulate Matter Air Pollution Causes Oxidant-Mediated Increase in Gut Permeability in Mice. Part. Fibre Toxicol. 2011, 8, 19. [Google Scholar] [CrossRef]
  36. Woodby, B.; Schiavone, M.L.; Pambianchi, E.; Mastaloudis, A.; Hester, S.N.; Wood, S.M.; Pecorelli, A.; Valacchi, G. Particulate Matter Decreases Intestinal Barrier-Associated Proteins Levels in 3D Human Intestinal Model. Int. J. Environ. Res. Public Health 2020, 17, 3234. [Google Scholar] [CrossRef] [PubMed]
  37. Jeong, M.-J.; Jeon, S.; Yu, H.-S.; Cho, W.-S.; Lee, S.; Kang, D.; Kim, Y.; Kim, Y.-J.; Kim, S.-Y. Exposure to Nickel Oxide Nanoparticles Induces Acute and Chronic Inflammatory Responses in Rat Lungs and Perturbs the Lung Microbiome. Int. J. Environ. Res. Public Health 2022, 19, 522. [Google Scholar] [CrossRef]
  38. Li, X.; Zhang, T.; Lv, W.; Wang, H.; Chen, H.; Xu, Q.; Cai, H.; Dai, J. Intratracheal administration of polystyrene microplastics induces pulmonary fibrosis by activating oxidative stress and Wnt/β-catenin signaling pathway in mice. Ecotoxicol. Environ. Saf. 2022, 232, 113238. [Google Scholar] [CrossRef]
  39. Michaudel, C.; Mackowiak, C.; Maillet, I.; Fauconnier, L.; Akdis, C.A.; Sokolowska, M.; Dreher, A.; Tan, H.-T.T.; Quesniaux, V.F.; Ryffel, B.; et al. Ozone Exposure Induces Respiratory Barrier Biphasic Injury and Inflammation Controlled by IL-33. J. Allergy Clin. Immunol. 2018, 142, 942–958. [Google Scholar] [CrossRef]
  40. Gentner, N.J.; Weber, L.P. Intranasal benzo[a]pyrene alters circadian blood pressure patterns and causes lung inflammation in rats. Arch. Toxicol. 2011, 85, 337–346. [Google Scholar] [CrossRef]
  41. Bai, H.; Wu, M.; Zhang, H.; Tang, G. Chronic polycyclic aromatic hydrocarbon exposure causes DNA damage and genomic instability in lung epithelial cells. Oncotarget 2017, 8, 79034–79045. [Google Scholar] [CrossRef] [PubMed]
  42. Li, J.; Hu, Y.; Liu, L.; Wang, Q.; Zeng, J.; Chen, C. PM2.5 Exposure Perturbs Lung Microbiome and Its Metabolic Profile in Mice. Sci. Total Environ. 2020, 721, 137432. [Google Scholar] [CrossRef] [PubMed]
  43. Kaspersen, K.A.; Antonsen, S.; Horsdal, H.T.; Kjerulff, B.; Brandt, J.; Geels, C.; Christensen, J.H.; Frohn, L.M.; Sabel, C.E.; Dinh, K.M.; et al. Exposure to Air Pollution and Risk of Respiratory Tract Infections in the Adult Danish Population—A Nationwide Study. Clin. Microbiol. Infect. 2024, 30, 122–129. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, C.; Xun, P.; Tsinovoi, C.; McClure, L.A.; Brockman, J.; MacDonald, L.; Cushman, M.; Cai, J.; Kamendulis, L.; Mackey, J.; et al. Urinary cadmium concentration and the risk of ischemic stroke. Neurology. 2018, 91, e382–e391. [Google Scholar] [CrossRef]
  45. Wen, Y.; Huang, S.; Zhang, Y.; Zhang, H.; Zhou, L.; Li, D.; Xie, C.; Lv, Z.; Guo, Y.; Ke, Y.; et al. Associations of multiple plasma metals with the risk of ischemic stroke: A case-control study. Environ. Int. 2019, 125, 125–134. [Google Scholar] [CrossRef]
  46. Kopf, P.G.; Walker, M.K. 2,3,7,8-tetrachlorodibenzo-p-dioxin increases reactive oxygen species production in human endothelial cells via induction of cytochrome P4501A1. Toxicol. Appl. Pharmacol. 2010, 245, 91–99. [Google Scholar] [CrossRef]
  47. Tzeng, H.P.; Yang, T.H.; Wu, C.T.; Chiu, H.C.; Liu, S.H.; Lan, K.C. Benzo[a]pyrene alters vascular function in rat aortas ex vivo and in vivo. Vasc. Pharmacol. 2019, 121, 106578. [Google Scholar] [CrossRef]
  48. Lee, D.-H.; Lind, P.M.; Jacobs, D.R., Jr.; Salihovic, S.; van Bavel, B.; Lind, L. Background exposure to persistent organic pollutants predicts stroke in the elderly. Environ. Int. 2012, 47, 115–1120. [Google Scholar] [CrossRef] [PubMed]
  49. Toborek, M.; Barger, S.W.; Mattson, M.P.; Espandiari, P.; Robertson, L.W.; Hennig, B. Exposure to polychlorinated biphenyls causes endothelial cell dysfunction. J. Biochem. Toxicol. 1995, 10, 219–226. [Google Scholar] [CrossRef]
  50. Gaur, N.; Dutta, D.; Singh, A.; Dubey, R.; Kamboj, D.V. Recent Advances in the Elimination of Persistent Organic Pollutants by Photocatalysis. Front. Environ. Sci. 2022, 10, 872514. [Google Scholar] [CrossRef]
  51. Samanta, S.K.; Singh, O.V.; Jain, R.K. Polycyclic Aromatic Hydrocarbons: Environmental Pollution and Bioremediation. Trends Biotechnol. 2002, 20, 243–248. [Google Scholar] [CrossRef] [PubMed]
  52. Adeniji, A.O.; Okoh, O.O.; Okoh, A.I. Levels of Polycyclic Aromatic Hydrocarbons in the Water and Sediment of Buffalo River Estuary, South Africa and Their Health Risk Assessment. Arch. Environ. Contam. Toxicol. 2019, 76, 657–669. [Google Scholar] [CrossRef] [PubMed]
  53. Rodgman, A.; Smith, C.J.; Perfetti, T.A. The Composition of Cigarette Smoke: A Retrospective, with Emphasis on Polycyclic Components. Hum. Exp. Toxicol. 2000, 19, 573–595. [Google Scholar] [CrossRef] [PubMed]
  54. Sharma, B.M.; Bharat, G.K.; Tayal, S.; Nizzetto, L.; Čupr, P.; Larssen, T. Environment and Human Exposure to Persistent Organic Pollutants (POPs) in India: A Systematic Review of Recent and Historical Data. Environ. Int. 2014, 66, 48–64. [Google Scholar] [CrossRef] [PubMed]
  55. Daley, J.M.; Paterson, G.; Drouillard, K.G. Bioamplification as a Bioaccumulation Mechanism for Persistent Organic Pollutants (POPs) in Wildlife. Rev. Environ. Contam. Toxicol. 2013, 227, 107–155. [Google Scholar] [CrossRef] [PubMed]
  56. Larigot, L.; Juricek, L.; Dairou, J.; Coumoul, X. AhR Signaling Pathways and Regulatory Functions. Biochim. Open 2018, 7, 1–9. [Google Scholar] [CrossRef] [PubMed]
  57. Esteves, F.; Rueff, J.; Kranendonk, M. The Central Role of Cytochrome P450 in Xenobiotic Metabolism—A Brief Review on a Fascinating Enzyme Family. J. Xenobiotics 2021, 11, 94–114. [Google Scholar] [CrossRef] [PubMed]
  58. Bukowska, B.; Mokra, K.; Michałowicz, J. Benzo[a]Pyrene—Environmental Occurrence, Human Exposure, and Mechanisms of Toxicity. Int. J. Mol. Sci. 2022, 23, 6348. [Google Scholar] [CrossRef] [PubMed]
  59. Knecht, A.L.; Goodale, B.C.; Truong, L.; Simonich, M.T.; Swanson, A.J.; Matzke, M.M.; Anderson, K.A.; Waters, K.M.; Tanguay, R.L. Comparative Developmental Toxicity of Environmentally Relevant Oxygenated PAHs. Toxicol. Appl. Pharmacol. 2013, 271, 266–275. [Google Scholar] [CrossRef]
  60. Mandal, P.K. Dioxin: A Review of Its Environmental Effects and Its Aryl Hydrocarbon Receptor Biology. J. Comp. Physiol. B 2005, 175, 221–230. [Google Scholar] [CrossRef]
  61. Shimizu, Y.; Nakatsuru, Y.; Ichinose, M.; Takahashi, Y.; Kume, H.; Mimura, J.; Fujii-Kuriyama, Y.; Ishikawa, T. Benzo[a]Pyrene Carcinogenicity Is Lost in Mice Lacking the Aryl Hydrocarbon Receptor. Proc Natl. Acad. Sci. USA 2000, 97, 779–782. [Google Scholar] [CrossRef]
  62. Gonzalez, F.J.; Fernandez-Salguero, P. The Aryl Hydrocarbon Receptor: Studies Using the AHR-Null Mice. Drug Metab. Dispos. Biol. Fate Chem. 1998, 26, 1194–1198. [Google Scholar]
  63. Ranjbar, M.; Rotondi, M.A.; Ardern, C.I.; Kuk, J.L. Urinary Biomarkers of Polycyclic Aromatic Hydrocarbons Are Associated with Cardiometabolic Health Risk. PLoS ONE 2015, 10, e0137536. [Google Scholar] [CrossRef]
  64. Xu, X.; Cook, R.L.; Ilacqua, V.A.; Kan, H.; Talbott, E.O.; Kearney, G. Studying Associations between Urinary Metabolites of Polycyclic Aromatic Hydrocarbons (PAHs) and Cardiovascular Diseases in the United States. Sci. Total Environ. 2010, 408, 4943–4948. [Google Scholar] [CrossRef]
  65. Mallah, M.A.; Mallah, M.A.; Liu, Y.; Xi, H.; Wang, W.; Feng, F.; Zhang, Q. Relationship Between Polycyclic Aromatic Hydrocarbons and Cardiovascular Diseases: A Systematic Review. Front. Public Health 2021, 9, 763706. [Google Scholar] [CrossRef]
  66. Rahman, H.H.; Sheikh, S.P.; Munson-McGee, S.H. Arsenic, Polycyclic Aromatic Hydrocarbons, and Metal Exposure and Risk Assessment of Stroke. Environ. Sci. Pollut. Res. 2023, 30, 86973–86986. [Google Scholar] [CrossRef]
  67. Curfs, D.M.J.; Lutgens, E.; Gijbels, M.J.J.; Kockx, M.M.; Daemen, M.J.A.P.; van Schooten, F.J. Chronic Exposure to the Carcinogenic Compound Benzo[a]Pyrene Induces Larger and Phenotypically Different Atherosclerotic Plaques in ApoE-Knockout Mice. Am. J. Pathol. 2003, 164, 101–108. [Google Scholar] [CrossRef]
  68. Grova, N.; Schroeder, H.; Farinelle, S.; Prodhomme, E.; Valley, A.; Muller, C.P. Sub-Acute Administration of Benzo[a]Pyrene (B[a]P) Reduces Anxiety-Related Behaviour in Adult Mice and Modulates Regional Expression of N-Methyl-d-Aspartate (NMDA) Receptors Genes in Relevant Brain Regions. Chemosphere 2008, 73, S295–S302. [Google Scholar] [CrossRef]
  69. Ho, D.H.; Burggren, W.W. Blood-Brain Barrier Function, Cell Viability, and Gene Expression of Tight Junction-Associated Proteins in the Mouse Are Disrupted by Crude Oil, Benzo[a]Pyrene, and the Dispersant COREXIT. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2019, 223, 96–105. [Google Scholar] [CrossRef]
  70. Tanaka, M.; Okuda, T.; Itoh, K.; Ishihara, N.; Oguro, A.; Fujii-Kuriyama, Y.; Nabetani, Y.; Yamamoto, M.; Vogel, C.F.A.; Ishihara, Y. Polycyclic Aromatic Hydrocarbons in Urban Particle Matter Exacerbate Movement Disorder after Ischemic Stroke via Potentiation of Neuroinflammation. Part. Fibre Toxicol. 2023, 20, 6. [Google Scholar] [CrossRef]
  71. Gu, X.; Manautou, J.E. Molecular Mechanisms Underlying Chemical Liver Injury. Expert Rev. Mol. Med. 2012, 14, e4. [Google Scholar] [CrossRef]
  72. Erickson, M.D. Analytical Chemistry of PCBs; Routledge: London, UK, 1997; pp. 125–151. [Google Scholar] [CrossRef]
  73. Raffetti, E.; Donato, F.; Palma, G.D.; Leonardi, L.; Sileo, C.; Magoni, M. Polychlorinated Biphenyls (PCBs) and Risk of Hypertension: A Population-Based Cohort Study in a North Italian Highly Polluted Area. Sci. Total Environ. 2020, 714, 136660. [Google Scholar] [CrossRef]
  74. Goncharov, A.; Pavuk, M.; Foushee, H.R.; Carpenter, D.O.; Consortium, A.E.H.R. Blood Pressure in Relation to Concentrations of PCB Congeners and Chlorinated Pesticides. Environ. Health Perspect. 2011, 119, 319–325. [Google Scholar] [CrossRef]
  75. Lim, J.; Lee, S.; Lee, S.; Jee, S.H. Serum Persistent Organic Pollutants Levels and Stroke Risk. Environ. Pollut. 2018, 233, 855–861. [Google Scholar] [CrossRef]
  76. Schlezinger, J.J.; Struntz, W.D.J.; Goldstone, J.V.; Stegeman, J.J. Uncoupling of Cytochrome P450 1A and Stimulation of Reactive Oxygen Species Production by Co-Planar Polychlorinated Biphenyl Congeners. Aquat. Toxicol. 2006, 77, 422–432. [Google Scholar] [CrossRef]
  77. Hennig, B.; Meerarani, P.; Slim, R.; Toborek, M.; Daugherty, A.; Silverstone, A.E.; Robertson, L.W. Proinflammatory Properties of Coplanar PCBs: In Vitro and in Vivo Evidence. Toxicol. Appl. Pharmacol. 2002, 181, 174–183. [Google Scholar] [CrossRef]
  78. Evich, M.G.; Davis, M.J.B.; McCord, J.P.; Acrey, B.; Awkerman, J.A.; Knappe, D.R.U.; Lindstrom, A.B.; Speth, T.F.; Tebes-Stevens, C.; Strynar, M.J.; et al. Per- and Polyfluoroalkyl Substances in the Environment. Science 2022, 375, eabg9065. [Google Scholar] [CrossRef]
  79. Glüge, J.; Scheringer, M.; Cousins, I.T.; DeWitt, J.C.; Goldenman, G.; Herzke, D.; Lohmann, R.; Ng, C.A.; Trier, X.; Wang, Z. An Overview of the Uses of Per- and Polyfluoroalkyl Substances (PFAS). Environ. Sci. Process. Impacts 2020, 22, 2345–2373. [Google Scholar] [CrossRef]
  80. Cousins, I.T.; DeWitt, J.C.; Glüge, J.; Goldenman, G.; Herzke, D.; Lohmann, R.; Ng, C.A.; Scheringer, M.; Wang, Z. The High Persistence of PFAS Is Sufficient for Their Management as a Chemical Class. Environ. Sci. Process. Impacts 2020, 22, 2307–2312. [Google Scholar] [CrossRef]
  81. Domingo, J.L.; Nadal, M. Human Exposure to Per- and Polyfluoroalkyl Substances (PFAS) through Drinking Water: A Review of the Recent Scientific Literature. Environ. Res. 2019, 177, 108648. [Google Scholar] [CrossRef]
  82. Roth, K.; Imran, Z.; Liu, W.; Petriello, M.C. Diet as an Exposure Source and Mediator of Per- and Polyfluoroalkyl Substance (PFAS) Toxicity. Front. Toxicol. 2020, 2, 601149. [Google Scholar] [CrossRef]
  83. Olsen, G.W.; Burris, J.M.; Ehresman, D.J.; Froehlich, J.W.; Seacat, A.M.; Butenhoff, J.L.; Zobel, L.R. Half-Life of Serum Elimination of Perfluorooctanesulfonate, Perfluorohexanesulfonate, and Perfluorooctanoate in Retired Fluorochemical Production Workers. Environ. Health Perspect. 2007, 115, 1298–1305. [Google Scholar] [CrossRef]
  84. Xiao, F.; An, Z.; Lv, J.; Sun, X.; Sun, H.; Liu, Y.; Liu, X.; Guo, H. Association between Per- and Polyfluoroalkyl Substances and Risk of Hypertension: A Systematic Review and Meta-Analysis. Front. Public Health 2023, 11, 1173101. [Google Scholar] [CrossRef]
  85. Boehme, A.K.; Esenwa, C.; Elkind, M.S.V. Stroke Risk Factors, Genetics, and Prevention. Circ. Res. 2017, 120, 472–495. [Google Scholar] [CrossRef]
  86. Simpson, C.; Winquist, A.; Lally, C.; Steenland, K. Relation between Perfluorooctanoic Acid Exposure and Strokes in a Large Cohort Living near a Chemical Plant. Environ. Res. 2013, 127, 22–28. [Google Scholar] [CrossRef]
  87. Feng, X.; Long, G.; Zeng, G.; Zhang, Q.; Song, B.; Wu, K.-H. Association of Increased Risk of Cardiovascular Diseases with Higher Levels of Perfluoroalkylated Substances in the Serum of Adults. Environ. Sci. Pollut. Res. 2022, 29, 89081–89092. [Google Scholar] [CrossRef]
  88. Schillemans, T.; Donat-Vargas, C.; Lindh, C.H.; de Faire, U.; Wolk, A.; Leander, K.; Åkesson, A. Per- and Polyfluoroalkyl Substances and Risk of Myocardial Infarction and Stroke: A Nested Case–Control Study in Sweden. Environ. Health Perspect. 2022, 130, 037007. [Google Scholar] [CrossRef]
  89. Zhang, H.; Shen, L.; Fang, W.; Zhang, X.; Zhong, Y. Perfluorooctanoic Acid-Induced Immunotoxicity via NF-Kappa B Pathway in Zebrafish (Danio Rerio) Kidney. Fish Shellfish Immunol. 2021, 113, 9–19. [Google Scholar] [CrossRef]
  90. Vemuganti, R. Therapeutic Potential of PPAR γ Activation in Stroke. PPAR Res. 2008, 2008, 461981. [Google Scholar] [CrossRef]
  91. Satriotomo, I.; Bowen, K.K.; Vemuganti, R. JAK2 and STAT3 Activation Contributes to Neuronal Damage Following Transient Focal Cerebral Ischemia. J. Neurochem. 2006, 98, 1353–1368. [Google Scholar] [CrossRef]
  92. Meneguzzi, A.; Fava, C.; Castelli, M.; Minuz, P. Exposure to Perfluoroalkyl Chemicals and Cardiovascular Disease: Experimental and Epidemiological Evidence. Front. Endocrinol. 2021, 12, 706352. [Google Scholar] [CrossRef]
  93. Verhoeven, J.I.; Allach, Y.; Vaartjes, I.C.H.; Klijn, C.J.M.; de Leeuw, F.-E. Ambient Air Pollution and the Risk of Ischaemic and Haemorrhagic Stroke. Lancet Planet. Health 2021, 5, e542–e552. [Google Scholar] [CrossRef]
  94. Strosnider, H.; Kennedy, C.; Monti, M.; Yip, F. Rural and Urban Differences in Air Quality, 2008–2012, and Community Drinking Water Quality, 2010–2015—United States. MMWR Surveill. Summ. 2017, 66, 1–10. [Google Scholar] [CrossRef]
  95. Mannucci, P.M.; Franchini, M. Health Effects of Ambient Air Pollution in Developing Countries. Int. J. Environ. Res. Public Health 2017, 14, 1048. [Google Scholar] [CrossRef]
  96. Schwarze, P.E.; Øvrevik, J.; Låg, M.; Refsnes, M.; Nafstad, P.; Hetland, R.B.; Dybing, E. Particulate Matter Properties and Health Effects: Consistency of Epidemiological and Toxicological Studies. Hum. Exp. Toxicol. 2006, 25, 559–579. [Google Scholar] [CrossRef]
  97. Poh, T.Y.; Ali, N.A.B.M.; Aogáin, M.M.; Kathawala, M.H.; Setyawati, M.I.; Ng, K.W.; Chotirmall, S.H. Inhaled Nanomaterials and the Respiratory Microbiome: Clinical, Immunological and Toxicological Perspectives. Part. Fibre Toxicol. 2018, 15, 46. [Google Scholar] [CrossRef]
  98. Mühlfeld, 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]
  99. Hameed, S.; Zhao, J.; Zare, R.N. Ambient PM Particles Reach Mouse Brain, Generate Ultrastructural Hallmarks of Neuroinflammation, and Stimulate Amyloid Deposition, Tangles, and Plaque Formation. Talanta. Open 2020, 2, 100013. [Google Scholar] [CrossRef]
  100. Hartz, A.M.S.; Bauer, B.; Block, M.L.; Hong, J.-S.; Miller, D.S. Diesel Exhaust Particles Induce Oxidative Stress, Proinflammatory Signaling, and P-glycoprotein Up-regulation at the Blood-brain Barrier. FASEB J. 2008, 22, 2723–2733. [Google Scholar] [CrossRef]
  101. Block, M.L.; Wu, X.; Pei, Z.; Li, G.; Wang, T.; Qin, L.; Wilson, B.; Yang, J.; Hong, J.S.; Veronesi, B. Nanometer Size Diesel Exhaust Particles Are Selectively Toxic to Dopaminergic Neurons: The Role of Microglia, Phagocytosis, and NADPH Oxidase. FASEB J. 2004, 18, 1618–1620. [Google Scholar] [CrossRef]
  102. Gulisano, M.; Pacini, P.; Marceddu, S.; Orlandini, G.E. Scanning Electron Microscopic Evaluation of the Alterations Induced by Polluted Air in the Rabbit Bronchial Epithelium. Ann. Anat. Anat. Anz. 1995, 177, 125–131. [Google Scholar] [CrossRef] [PubMed]
  103. Kleinman, M.T.; Iley, R.M.; Chang, Y.-T.C.; Clark, K.W.; Jones, M.P.; Linn, W.S.; Hackney, J.D. Exposures of Human Volunteers to a Controlled Atmospheric Mixture of Ozone, Sulfur Dioxide and Sulfuric Acid. AIHA J. 1981, 42, 61–69. [Google Scholar] [CrossRef] [PubMed]
  104. Bell, M.L.; Son, J.-Y.; Peng, R.D.; Wang, Y.; Dominici, F. Ambient PM2.5 and Risk of Hospital Admissions: Do Risks Differ for Men and Women? Epidemiology 2015, 26, 575–579. [Google Scholar] [CrossRef]
  105. Andersen, Z.J.; Olsen, T.S.; Andersen, K.K.; Loft, S.; Ketzel, M.; Raaschou-Nielsen, O. Association between Short-Term Exposure to Ultrafine Particles and Hospital Admissions for Stroke in Copenhagen, Denmark. Eur. Heart, J. 2010, 31, 2034–2040. [Google Scholar] [CrossRef]
  106. Li, B.; Ma, Y.; Zhou, Y.; Chai, E. Research Progress of Different Components of PM2.5 and Ischemic Stroke. Sci. Rep. 2023, 13, 15965. [Google Scholar] [CrossRef]
  107. Zhang, R.; Liu, G.; Jiang, Y.; Li, G.; Pan, Y.; Wang, Y.; Wei, Z.; Wang, J.; Wang, Y. Acute Effects of Particulate Air Pollution on Ischemic Stroke and Hemorrhagic Stroke Mortality. Front. Neurol. 2018, 9, 827. [Google Scholar] [CrossRef]
  108. Szyszkowicz, M.; Rowe, B.H.; Brook, R.D. Even Low Levels of Ambient Air Pollutants Are Associated With Increased Emergency Department Visits for Hypertension. Can. J. Cardiol. 2012, 28, 360–366. [Google Scholar] [CrossRef] [PubMed]
  109. Lai, H.; Liu, X.; Qu, M. Nanoplastics and Human Health: Hazard Identification and Biointerface. Nanomaterials 2022, 12, 1298. [Google Scholar] [CrossRef]
  110. Ghosh, S.; Sinha, J.K.; Ghosh, S.; Vashisth, K.; Han, S.; Bhaskar, R. Microplastics as an Emerging Threat to the Global Environment and Human Health. Sustainability 2023, 15, 10821. [Google Scholar] [CrossRef]
  111. Mason, S.A.; Welch, V.G.; Neratko, J. Synthetic Polymer Contamination in Bottled Water. Front. Chem. 2018, 6, 407. [Google Scholar] [CrossRef]
  112. Gasperi, J.; Wright, S.L.; Dris, R.; Collard, F.; Mandin, C.; Guerrouache, M.; Langlois, V.; Kelly, F.J.; Tassin, B. Microplastics in Air: Are We Breathing It In? Curr. Opin. Environ. Sci. Health 2018, 1, 1–5. [Google Scholar] [CrossRef]
  113. Leslie, H.A.; van Velzen, M.J.M.; Brandsma, S.H.; Vethaak, A.D.; Garcia-Vallejo, J.J.; Lamoree, M.H. Discovery and Quantification of Plastic Particle Pollution in Human Blood. Environ. Int. 2022, 163, 107199. [Google Scholar] [CrossRef] [PubMed]
  114. Ragusa, A.; Notarstefano, V.; Svelato, A.; Belloni, A.; Gioacchini, G.; Blondeel, C.; Zucchelli, E.; Luca, C.D.; D’Avino, S.; Gulotta, A.; et al. Raman Microspectroscopy Detection and Characterisation of Microplastics in Human Breastmilk. Polymers 2022, 14, 2700. [Google Scholar] [CrossRef] [PubMed]
  115. Schwabl, P.; Köppel, S.; Königshofer, P.; Bucsics, T.; Trauner, M.; Reiberger, T.; Liebmann, B. Detection of Various Microplastics in Human Stool: A Prospective Case Series. Ann. Intern. Med. 2019, 171, 453–457. [Google Scholar] [CrossRef] [PubMed]
  116. Meaza, I.; Toyoda, J.H.; Wise, J.P. Microplastics in Sea Turtles, Marine Mammals and Humans: A One Environmental Health Perspective. Front. Environ. Sci. 2020, 8, 575614. [Google Scholar] [CrossRef] [PubMed]
  117. Nelms, S.E.; Barnett, J.; Brownlow, A.; Davison, N.J.; Deaville, R.; Galloway, T.S.; Lindeque, P.K.; Santillo, D.; Godley, B.J. Microplastics in Marine Mammals Stranded around the British Coast: Ubiquitous but Transitory? Sci. Rep. 2019, 9, 1075. [Google Scholar] [CrossRef] [PubMed]
  118. Marfella, R.; Prattichizzo, F.; Sardu, C.; Fulgenzi, G.; Graciotti, L.; Spadoni, T.; D’Onofrio, N.; Scisciola, L.; Grotta, R.L.; Frigé, C.; et al. Microplastics and Nanoplastics in Atheromas and Cardiovascular Events. N. Engl. J. Med. 2024, 390, 900–910. [Google Scholar] [CrossRef] [PubMed]
  119. Harris-Tryon, T.A.; Grice, E.A. Microbiota and Maintenance of Skin Barrier Function. Science 2022, 376, 940–945. [Google Scholar] [CrossRef] [PubMed]
  120. Brune, K.; Frank, J.; Schwingshackl, A.; Finigan, J.; Sidhaye, V.K. Pulmonary Epithelial Barrier Function: Some New Players and Mechanisms. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2015, 308, L731–L745. [Google Scholar] [CrossRef]
  121. Turner, J.R. Intestinal Mucosal Barrier Function in Health and Disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef]
  122. Ganesan, S.; Comstock, A.T.; Sajjan, U.S. Barrier Function of Airway Tract Epithelium. Tissue Barriers 2013, 1, e24997. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, J.; Chen, X.; Dou, M.; He, H.; Ju, M.; Ji, S.; Zhou, J.; Chen, C.; Zhang, D.; Miao, C.; et al. Particulate Matter Disrupts Airway Epithelial Barrier via Oxidative Stress to Promote Pseudomonas Aeruginosa Infection. J. Thorac. Dis. 2019, 11, 2617–2627. [Google Scholar] [CrossRef] [PubMed]
  124. Caraballo, J.C.; Yshii, C.; Westphal, W.; Moninger, T.; Comellas, A.P. Ambient Particulate Matter Affects Occludin Distribution and Increases Alveolar Transepithelial Electrical Conductance. Respirology 2011, 16, 340–349. [Google Scholar] [CrossRef] [PubMed]
  125. Jang, A.-S.; Choi, I.-S.; Koh, Y.-I.; Park, C.-S.; Lee, J.-S. The Relationship between Alveolar Epithelial Proliferation and Airway Obstruction after Ozone Exposure. Allergy 2002, 57, 737–740. [Google Scholar] [CrossRef] [PubMed]
  126. Awad, W.A.; Hess, C.; Hess, M. Enteric Pathogens and Their Toxin-Induced Disruption of the Intestinal Barrier through Alteration of Tight Junctions in Chickens. Toxins 2017, 9, 60. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, M.; Wu, C. The Relationship between Intestinal Goblet Cells and the Immune Response. Biosci. Rep. 2020, 40, BSR20201471. [Google Scholar] [CrossRef] [PubMed]
  128. Iacob, S.; Iacob, D.G.; Luminos, L.M. Intestinal Microbiota as a Host Defense Mechanism to Infectious Threats. Front. Microbiol. 2019, 9, 3328. [Google Scholar] [CrossRef] [PubMed]
  129. Crapser, J.; Ritzel, R.; Verma, R.; Venna, V.R.; Liu, F.; Chauhan, A.; Koellhoffer, E.; Patel, A.; Ricker, A.; Maas, K.; et al. Ischemic Stroke Induces Gut Permeability and Enhances Bacterial Translocation Leading to Sepsis in Aged Mice. Aging Albany Ny 2016, 8, 1049–1060. [Google Scholar] [CrossRef]
  130. Xie, S.; Zhang, C.; Zhao, J.; Li, D.; Chen, J. Exposure to Concentrated Ambient PM2.5 (CAPM) Induces Intestinal Disturbance via Inflammation and Alternation of Gut Microbiome. Environ. Int. 2022, 161, 107138. [Google Scholar] [CrossRef]
  131. Saitou, M.; Furuse, M.; Sasaki, H.; Schulzke, J.-D.; Fromm, M.; Takano, H.; Noda, T.; Tsukita, S. Complex Phenotype of Mice Lacking Occludin, a Component of Tight Junction Strands. Mol. Biol. Cell 2000, 11, 4131–4142. [Google Scholar] [CrossRef]
  132. Nouri, M.; Bredberg, A.; Weström, B.; Lavasani, S. Intestinal Barrier Dysfunction Develops at the Onset of Experimental Autoimmune Encephalomyelitis, and Can Be Induced by Adoptive Transfer of Auto-Reactive T Cells. PLoS ONE 2014, 9, e106335. [Google Scholar] [CrossRef]
  133. Li, C.; Chen, W.; Lin, F.; Li, W.; Wang, P.; Liao, G.; Zhang, L. Functional Two-Way Crosstalk Between Brain and Lung: The Brain–Lung Axis. Cell. Mol. Neurobiol. 2023, 43, 991–1003. [Google Scholar] [CrossRef] [PubMed]
  134. Hosang, L.; Canals, R.C.; van der Flier, F.J.; Hollensteiner, J.; Daniel, R.; Flügel, A.; Odoardi, F. The Lung Microbiome Regulates Brain Autoimmunity. Nature 2022, 603, 138–144. [Google Scholar] [CrossRef] [PubMed]
  135. Aslanyan, S.; Weir, C.J.; Diener, H.-C.; Kaste, M.; Lees, K.R.; GAIN International Steering Committee and Investigators. Pneumonia and Urinary Tract Infection after Acute Ischaemic Stroke: A Tertiary Analysis of the GAIN International Trial. Eur. J. Neurol. 2004, 11, 49–53. [Google Scholar] [CrossRef] [PubMed]
  136. Faura, J.; Ramiro, L.; Simats, A.; Ma, F.; Penalba, A.; Gasull, T.; Rosell, A.; Montaner, J.; Bustamante, A. Evaluation and Characterization of Post-Stroke Lung Damage in a Murine Model of Cerebral Ischemia. Int. J. Mol. Sci. 2022, 23, 8093. [Google Scholar] [CrossRef] [PubMed]
  137. Faura, J.; Bustamante, A.; Miró-Mur, F.; Montaner, J. Stroke-Induced Immunosuppression: Implications for the Prevention and Prediction of Post-Stroke Infections. J. Neuroinflamm. 2021, 18, 127. [Google Scholar] [CrossRef] [PubMed]
  138. Katzan, I.L.; Cebul, R.D.; Husak, S.H.; Dawson, N.V.; Baker, D.W. The Effect of Pneumonia on Mortality among Patients Hospitalized for Acute Stroke. Neurology 2003, 60, 620–625. [Google Scholar] [CrossRef] [PubMed]
  139. Sigaud, S.; Goldsmith, C.-A.W.; Zhou, H.; Yang, Z.; Fedulov, A.; Imrich, A.; Kobzik, L. Air Pollution Particles Diminish Bacterial Clearance in the Primed Lungs of Mice. Toxicol. Appl. Pharmacol. 2007, 223, 1–9. [Google Scholar] [CrossRef] [PubMed]
  140. Pinkerton, K.E.; Green, F.H.; Saiki, C.; Vallyathan, V.; Plopper, C.G.; Gopal, V.; Hung, D.; Bahne, E.B.; Lin, S.S.; Ménache, 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]
  141. Kurmi, O.P.; Semple, S.; Simkhada, P.; Smith, W.C.S.; Ayres, J.G. COPD and Chronic Bronchitis Risk of Indoor Air Pollution from Solid Fuel: A Systematic Review and Meta-Analysis. Thorax 2010, 65, 221. [Google Scholar] [CrossRef]
  142. Elonheimo, H.M.; Mattila, T.; Andersen, H.R.; Bocca, B.; Ruggieri, F.; Haverinen, E.; Tolonen, H. Environmental Substances Associated with Chronic Obstructive Pulmonary Disease—A Scoping Review. Int. J. Environ. Res. Public Health 2022, 19, 3945. [Google Scholar] [CrossRef] [PubMed]
  143. O’Piela, D.R.; Durisek, G.R.; Escobar, Y.-N.H.; Mackos, A.R.; Wold, L.E. Particulate Matter and Alzheimer’s Disease: An Intimate Connection. Trends Mol. Med. 2022, 28, 770–780. [Google Scholar] [CrossRef] [PubMed]
  144. Ural, B.B.; Caron, D.P.; Dogra, P.; Wells, S.B.; Szabo, P.A.; Granot, T.; Senda, T.; Poon, M.M.L.; Lam, N.; Thapa, P.; et al. Inhaled Particulate Accumulation with Age Impairs Immune Function and Architecture in Human Lung Lymph Nodes. Nat. Med. 2022, 28, 2622–2632. [Google Scholar] [CrossRef] [PubMed]
  145. Samary, C.S.; Ramos, A.B.; Maia, L.A.; Rocha, N.N.; Santos, C.L.; Magalhães, R.F.; Clevelario, A.L.; Pimentel-Coelho, P.M.; Mendez-Otero, R.; Cruz, F.F.; et al. Focal Ischemic Stroke Leads to Lung Injury and Reduces Alveolar Macrophage Phagocytic Capability in Rats. Crit. Care 2018, 22, 249. [Google Scholar] [CrossRef] [PubMed]
  146. Brook, R.D.; Franklin, B.; Cascio, W.; Hong, Y.; Howard, G.; Lipsett, M.; Luepker, R.; Mittleman, M.; Samet, J.; Smith, S.C.; et al. Air Pollution and Cardiovascular Disease. Circulation 2004, 109, 2655–2671. [Google Scholar] [CrossRef] [PubMed]
  147. Hozawa, A.; Billings, J.L.; Shahar, E.; Ohira, T.; Rosamond, W.D.; Folsom, A.R. Lung Function and Ischemic Stroke Incidence The Atherosclerosis Risk in Communities Study. Chest 2006, 130, 1642–1649. [Google Scholar] [CrossRef] [PubMed]
  148. Dickson, R.P.; Erb-Downward, J.R.; Martinez, F.J.; Huffnagle, G.B. The Microbiome and the Respiratory Tract. Annu. Rev. Physiol. 2015, 78, 481–504. [Google Scholar] [CrossRef] [PubMed]
  149. Dickson, R.P.; Erb-Downward, J.R.; Freeman, C.M.; McCloskey, L.; Falkowski, N.R.; Huffnagle, G.B.; Curtis, J.L. Bacterial Topography of the Healthy Human Lower Respiratory Tract. mBio 2017, 8, e02287-16. [Google Scholar] [CrossRef]
  150. Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr. Med. 2018, 17, 28–32. [Google Scholar]
  151. Carloni, S.; Rescigno, M. The Gut-Brain Vascular Axis in Neuroinflammation. Semin. Immunol. 2023, 69, 101802. [Google Scholar] [CrossRef]
  152. Johnson, C.H.; Patterson, A.D.; Idle, J.R.; Gonzalez, F.J. Xenobiotic Metabolomics: Major Impact on the Metabolome. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 37–56. [Google Scholar] [CrossRef]
  153. Koppel, N.; Rekdal, V.M.; Balskus, E.P. Chemical Transformation of Xenobiotics by the Human Gut Microbiota. Science 2018, 356, eaag2770. [Google Scholar] [CrossRef]
  154. Zhang, Y.; Hu, Q.; Fu, J.; Li, X.; Mao, H.; Wang, T. Influence of Exposure Pathways on Tissue Distribution and Health Impact of Polycyclic Aromatic Hydrocarbon Derivatives. Environ. Health 2023, 1, 150–167. [Google Scholar] [CrossRef]
  155. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The Human Microbiome Project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef] [PubMed]
  156. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; FitzGerald, M.G.; Fulton, R.S.; et al. Structure, Function and Diversity of the Healthy Human Microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [PubMed]
  157. Badal, V.D.; Vaccariello, E.D.; Murray, E.R.; Yu, K.E.; Knight, R.; Jeste, D.V.; Nguyen, T.T. The Gut Microbiome, Aging, and Longevity: A Systematic Review. Nutrients 2020, 12, 3759. [Google Scholar] [CrossRef]
  158. Gacesa, R.; Kurilshikov, A.; Vila, A.V.; Sinha, T.; Klaassen, M.A.Y.; Bolte, L.A.; Andreu-Sánchez, S.; Chen, L.; Collij, V.; Hu, S.; et al. Environmental Factors Shaping the Gut Microbiome in a Dutch Population. Nature 2022, 604, 732–739. [Google Scholar] [CrossRef]
  159. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human Genetics Shape the Gut Microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef] [PubMed]
  160. Belkaid, Y.; Hand, T.W. Role of the Microbiota in Immunity and Inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef]
  161. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and Clinical Implications of the Brain–Gut–Enteric Microbiota Axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef]
  162. Lefever, D.E.; Xu, J.; Chen, Y.; Huang, G.; Tamas, N.; Guo, T.L. TCDD Modulation of Gut Microbiome Correlated with Liver and Immune Toxicity in Streptozotocin (STZ)-Induced Hyperglycemic Mice. Toxicol. Appl. Pharmacol. 2016, 304, 48–58. [Google Scholar] [CrossRef] [PubMed]
  163. Dirnagl, U.; Iadecola, C.; Moskowitz, M.A.; Dirnagl, U.; Iadecola, C.; Moskowitz, M.A. Pathobiology of Ischaemic Stroke: An Integrated View. Trends Neurosci 1999, 22, 391–397. [Google Scholar] [CrossRef] [PubMed]
  164. Iadecola, C.; Buckwalter, M.S.; Anrather, J. Immune Responses to Stroke: Mechanisms, Modulation, and Therapeutic Potential. J. Clin. Investig. 2020, 130, 2777–2788. [Google Scholar] [CrossRef] [PubMed]
  165. Simats, A.; Liesz, A. Systemic Inflammation after Stroke: Implications for Post-stroke Comorbidities. Embo Mol. Med. 2022, 14, e16219. [Google Scholar] [CrossRef] [PubMed]
  166. Benakis, C.; Brea, D.; Caballero, S.; Faraco, G.; Moore, J.; Murphy, M.; Sita, G.; Racchumi, G.; Ling, L.; Pamer, E.G.; et al. Commensal Microbiota Affects Ischemic Stroke Outcome by Regulating Intestinal Γδ T Cells. Nat. Med. 2016, 22, 516–523. [Google Scholar] [CrossRef] [PubMed]
  167. Brea, D.; Poon, C.; Benakis, C.; Lubitz, G.; Murphy, M.; Iadecola, C.; Anrather, J. Stroke Affects Intestinal Immune Cell Trafficking to the Central Nervous System. Brain Behav. Immun. 2021, 96, 295–302. [Google Scholar] [CrossRef] [PubMed]
  168. Singh, V.; Roth, S.; Llovera, G.; Sadler, R.; Garzetti, D.; Stecher, B.; Dichgans, M.; Liesz, A. Microbiota Dysbiosis Controls the Neuroinflammatory Response after Stroke. J. Neurosci. 2016, 36, 7428–7440. [Google Scholar] [CrossRef] [PubMed]
  169. Houlden, A.; Goldrick, M.; Brough, D.; Vizi, E.S.; Lénárt, N.; Martinecz, B.; Roberts, I.S.; Denes, A. Brain Injury Induces Specific Changes in the Caecal Microbiota of Mice via Altered Autonomic Activity and Mucoprotein Production. Brain Behav. Immun. 2016, 57, 10–20. [Google Scholar] [CrossRef] [PubMed]
  170. Xu, K.; Gao, X.; Xia, G.; Chen, M.; Zeng, N.; Wang, S.; You, C.; Tian, X.; Di, H.; Tang, W.; et al. Rapid Gut Dysbiosis Induced by Stroke Exacerbates Brain Infarction in Turn. Gut 2021, 70, 1486–1494. [Google Scholar] [CrossRef]
  171. Haak, B.W.; Westendorp, W.F.; van Engelen, T.S.R.; Brands, X.; Brouwer, M.C.; Vermeij, J.-D.; Hugenholtz, F.; Verhoeven, A.; Derks, R.J.; Giera, M.; et al. Disruptions of Anaerobic Gut Bacteria Are Associated with Stroke and Post-Stroke Infection: A Prospective Case–Control Study. Transl. Stroke Res. 2021, 12, 581–592. [Google Scholar] [CrossRef]
  172. Benakis, C.; Poon, C.; Lane, D.; Brea, D.; Sita, G.; Moore, J.; Murphy, M.; Racchumi, G.; Iadecola, C.; Anrather, J. Distinct Commensal Bacterial Signature in the Gut Is Associated With Acute and Long-Term Protection From Ischemic Stroke. Stroke 2020, 51, 1844–1854. [Google Scholar] [CrossRef] [PubMed]
  173. Strickland, P.; Kang, D.; Sithisarankul, P. Polycyclic Aromatic Hydrocarbon Metabolites in Urine as Biomarkers of Exposure and Effect. Environ. Health Perspect. 1996, 104, 927–932. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, J.-C.; Lai, C.-S.; Tsai, M.-L.; Ho, C.-T.; Wang, Y.-J.; Pan, M.-H. Chemopreventive Effect of Natural Dietary Compounds on Xenobiotic-Induced Toxicity. J. Food Drug Anal. 2017, 25, 176–186. [Google Scholar] [CrossRef]
  175. Stavric, B.; Klassen, R. Dietary Effects on the Uptake of Benzo[a]Pyrene. Food Chem. Toxicol. 1994, 32, 727–734. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of environmental toxins inhaled or ingested potentially leading to increased susceptibility to stroke and severity. Environmental toxins originate from several sources, including industrial processes and as byproducts from incomplete combustion. These toxins can enter the environment and contaminate sources such as food, water, and soil and can also be abundant in the air, increasing human exposure via inhalation and ingestion as two primary routes. Once exposed, these toxins can target the primary organs, gut and lungs, and induce perturbation of the microbiota composition (dysbiosis), a pro-inflammatory response leading to disruption of tissue architecture, immune cell activation, and increased permeability at epithelial barrier sites, which may increase the exposure risk to harmful bacteria and pathogens. Inhaled or ingested xenobiotics are metabolized and bind to their respective receptors, upregulating ligand-activated transcription factors and inflammatory signaling pathways (AhR, PPAR, and NF-κB). Alterations at the gut and lung barriers can indirectly affect brain function. Metabolites generated from the metabolism of xenobiotics can traffic to the brain and may directly increase the risk for stroke by compromising the blood–brain barrier, activating brain resident immune cells and immune cells trafficking from the periphery, and causing cell death. AhR, aryl hydrocarbon receptor; BBB, blood–brain barrier; NF-κB, nuclear factor-κB; PAHs, polycyclic aromatic hydrocarbons; PCBs, polychlorinated biphenyls; PET, polyethylene terephthalate; PFASs, per- and polyfluoroalkyl substances; POPs, persistent organic pollutants; PPAR-α and -δ, peroxisome proliferator-activated receptor-alpha and -delta; ROS, reactive oxygen species; TNF-α, tumor necrosis factor-alpha; Tregs, regulatory T cells, created with Biorender.com.
Figure 1. Mechanisms of environmental toxins inhaled or ingested potentially leading to increased susceptibility to stroke and severity. Environmental toxins originate from several sources, including industrial processes and as byproducts from incomplete combustion. These toxins can enter the environment and contaminate sources such as food, water, and soil and can also be abundant in the air, increasing human exposure via inhalation and ingestion as two primary routes. Once exposed, these toxins can target the primary organs, gut and lungs, and induce perturbation of the microbiota composition (dysbiosis), a pro-inflammatory response leading to disruption of tissue architecture, immune cell activation, and increased permeability at epithelial barrier sites, which may increase the exposure risk to harmful bacteria and pathogens. Inhaled or ingested xenobiotics are metabolized and bind to their respective receptors, upregulating ligand-activated transcription factors and inflammatory signaling pathways (AhR, PPAR, and NF-κB). Alterations at the gut and lung barriers can indirectly affect brain function. Metabolites generated from the metabolism of xenobiotics can traffic to the brain and may directly increase the risk for stroke by compromising the blood–brain barrier, activating brain resident immune cells and immune cells trafficking from the periphery, and causing cell death. AhR, aryl hydrocarbon receptor; BBB, blood–brain barrier; NF-κB, nuclear factor-κB; PAHs, polycyclic aromatic hydrocarbons; PCBs, polychlorinated biphenyls; PET, polyethylene terephthalate; PFASs, per- and polyfluoroalkyl substances; POPs, persistent organic pollutants; PPAR-α and -δ, peroxisome proliferator-activated receptor-alpha and -delta; ROS, reactive oxygen species; TNF-α, tumor necrosis factor-alpha; Tregs, regulatory T cells, created with Biorender.com.
Cells 13 00803 g001
Table 1. Studies linking the effect of environmental toxins on the gut, lungs, and circulatory system and their consequences for stroke risk.
Table 1. Studies linking the effect of environmental toxins on the gut, lungs, and circulatory system and their consequences for stroke risk.
Environmental ToxinsCompound/
Substance
ModelDosageExposure Method and DurationEffectReferences
Gut
Heavy MetalCadmiumSprague–Dawley rats5 mg/kgGastric infusion daily for 30 days↓ Expression of the tight junction protein ZO-1
↑ Expression of TNF-α, and IL-6 in the gut
[25]
Heavy MetalsCadmium/ArsenicC57BL/6 mice50 ppmDietary exposure through drinking water for 2 weeks↓ Alpha diversity of the microbiota[26]
Heavy MetalArsenicParticipants in cohort study<50 μg/L20-year exposure
window
Higher incidence of stroke[27]
Microplastics and nanoplasticsMicro/nanoplasticsC57/B6 mice0.2 and 2 mg/kg28 daysHigh dose led to morphological changes along the GI including the followings:
-Absence of villi
-Damaged crypts
↓ Mucosa wall lining
↓ Tight junction expression.
↓ Microbial diversity and ratio of bacteria
[15]
MicroplasticsPolyethylene terephthalate (PET)In vitro simulation0.166 g/intake72 hShift in the relative abundance of bacteria in the upper region of the colon. Specifically, ↑ Desulfobacterota and ↓ Proteobacteria over time.
↓ Viable bacterial count
[28]
MicroplasticsPS-MPsMale mice0.1mg/day of 5 or 20 μm PS-MPs28 days via oral gavageAccumulation of PS-MPs in the gut, liver, and kidney.
↑ Inflammation and lipid accumulation in the liver
[29]
PAHBaPC57BL/6 mice10 mL/kg of BW28 days oral exposure↑ Inflammation in ileal segments
-Altered relative abundance of fecal and mucosa associated microbiota
↓ Lactobacillus
[12]
PCBsPCB153, PCB138, and PCB180C57BL/6 mice150 µmol/kg2 days↓ Proteobacteria[30]
PCBsProspective population based Follow upMiddle-aged and elderly womenPassive dietary exposure12 years of follow-upPositive association observed between stroke risk and PCB exposure.[31]
PCBPCB 126Ldlr−/− mice1 μmol/kg of PCB 12614-week atherogenic diet and exposed to PCB 126 (1 μmol/kg) at weeks 2 and 4↑ Expression of TNF-α, IL-6, and interleukin IL-18 in the jejunum
↑ Tight junction proteins: Occludin and claudin in the colon
↓ Expression of PPAR-δ in the colon
↓ Abundance of Clostridiales, Bifidobacterium, Lactobacillus, Ruminococcus, and Oscillospira.
↑ Abundance of Akkermansia.
↓ Alpha diversity in cecum contents
[32]
PCBPCB153C57BL/6 mice300 μmol/kg1× per day for 2-days↑ Expression of TNF-α and IL-6 in the intestinal epithelial cells of the small intestines
-Activates NF-κB pathway
-Induces DNA damage
[33]
Particulate matterUrban particulate matterC57BL/6 mice40 μg course particulate matter/mLMice were placed 4 h/day or 5 days/week for 2 weeks in inhalation chamber↑ Expression of TNF-α, IFN-γ, CXCL10, and IL-10
↓ Expression of IL-5
[34]
Particulate matterUrban particulate matterC57BL/6 mice200 μgGastric gavage↑ ROS production
↑ Colonic epithelial cell death
[35]
Particulate matterAtmosphericparticulate matterIntestinal tissue50–500 µg/cm21 week and 2 weeks↓ ZO−1 and claudin−1 expression level[36]
Lungs
NanoparticlesNickel Oxide NanoparticlesRats50 and 150 cm2 for 10 minIntratracheally instilled 1 day and 4 weeks-Narrowed alveolar ducts and alveoli
↑ Levels of neutrophils and cytokines
-Induced pulmonary microbiome dysbiosis in the acute phase
[37]
Nano and microplasticsPolyethylene particlesBALB/c mice10 mg/kg 1× daily for 1 weekOral administration↑ Percentage of Th17, Tregs, and Th2 cells
↓ Colon length
↑ Percentages of IL-4+, Foxp3+(Tregs), and IL-17 in CD4+T cells
↑ Secretion of IL-4 and IL-17 cytokines
[13]
MicroplasticsPS-MPsC57BL/6 mice6.25 mg/kg PS-MPsIntratracheally instilled 3× per week for 3 weeks↑ Expression of collagen
↑ Fibrosis with increased exposure
↑ Oxidative stress
[38]
OzoneOzone exposureC57BL/6 mice1 ppm ozone for 1 hInhalation chamber 1×↑ Barrier disruption and epithelial cell permeability[39]
OzoneOzone exposureC57BL/6 mice1 ppm for the acute model and 1.5 ppm for the chronic model.Acute phase: 1 h
Chronic phase: 2 h, 2× per week for 6 weeks
Acute exposure induced the following:
Disrupted tight junctions
Desquamation of epithelial layer
Chronic exposure to ozone particles remodeled the airway in mice using the following:
↑ Airspace density and diameter
↓ Number of airspaces
↓ Epithelium thickness
[14]
PAHBaPSprague Dawley
rats
0.01 mg/kgIntratracheally instilled for 7 days↑ Neutrophil recruitment
↑ Lung inflammation
[40]
PAHAtmospheric PAHsHuman lung epithelial cell linesLow dose30 daysChronic exposure:
Induced DNA damage
Altered cellular homeostasis and
↑ ROS production
[41]
Particulate matterPM2.5C57BL/6N mice1.8, 5.4, and 16.2 mg/kgIntratracheally instilled for 1 weekPM2.5 exposure led to the following:
Infiltration of inflammatory cells, ↑ serum cytokine levels in the serum
↑ Lung microbiome diversity
↓ Relative abundance of proteobacteria post-exposure
↑ Number of goblet cells
[42]
Particulate matterVarious18–64 yearsPassive exposureAverage daily concentrationExposure to air pollutants
increased the risk for respiratory tract infections.
[43]
Circulatory system
Heavy MetalCadmiumCohort studyPassive exposureMeasured urinary cadmium concentrationMay increase the incidence of ischemic stroke[44]
Heavy MetalMixed metalsIschemic stroke patientsPassive exposureFasting blood concentration within 48 h post-diagnosisHigher plasma concentrations of aluminum, arsenic, and cadmium
may increase the risk of ischemic stroke.
[45]
DioxinTCDDPrimary human aortic endothelial cell0.1% TCDD24 h↑ Hypertension and endothelial dysfunction.[46]
PAHBaPSprague Dawley
rats
0.01 mg/kgIntratracheally instilled for 7 days↑ Systolic and diastolic pressure and heart rate[40]
PAHBaPWistar rats20 mg/kg for 4 and 8 weeksIntra-peritoneal. injection↑ Blood pressure after 8 weeks in vivo
↑ Vasoconstriction ex vivo
[47]
POPsPassive exposureAged 70 yrs. in Sweden. 5-year follow-upPassive exposureBaseline plasma samplesPCB congeners, organochlorine pesticides, and octachlorodibenzo-p-dioxin showed increased risk of developing stroke in elderly population.[48]
PCBsVarious PCBsEndothelial cells from porcine pulmonary arteriesPCB 77, PCB 153, and PCB11424 hExposure led to the development of atherosclerosis and endothelial barrier dysfunction.
↑ Albumin flux and oxidative stress
[49]
BaP, benzo-a-pyrene; CXCL10, interferon gamma-induced protein 10; DNA, deoxyribonucleic acid; Foxp3, forkhead box P3; IFN-γ, interferon-gamma; IL-4, interleukin-4; IL-5, interleukin-5; IL-6, interleukin-6; IL-10, interleukin-10; IL-17, interleukin-17; Ldlr−/−, LDL receptor deficient mouse; NF-κB, nuclear factor-κB; PAH, polycyclic aromatic hydrocarbon; PCB, polychlorinated biphenyls; PET, polyethylene terephthalate; PM2.5, particulate matter ≤ 2.5 μm; PS-MP, polystyrene microplastics; POP, persistent organic pollutant; ROS, reactive oxygen species; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; Th2, T helper 2 cells; Th17, T helper 17 cells; TNF-α, tumor necrosis factor-alpha; Tregs, regulatory T cells; ZO-1, zona occludens 1. Upward and downward arrows indicate an increase or a decrease of the observed effect, respectively.
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

Ruggles, A.; Benakis, C. Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut– and Lung–Brain Axis. Cells 2024, 13, 803. https://doi.org/10.3390/cells13100803

AMA Style

Ruggles A, Benakis C. Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut– and Lung–Brain Axis. Cells. 2024; 13(10):803. https://doi.org/10.3390/cells13100803

Chicago/Turabian Style

Ruggles, Alexandria, and Corinne Benakis. 2024. "Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut– and Lung–Brain Axis" Cells 13, no. 10: 803. https://doi.org/10.3390/cells13100803

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