Next Article in Journal
Ecotoxicological Assessment of Sediment Samples Impacted by Wastewater Treatment Plant Effluents Transporting Contaminants of Emerging Concern
Previous Article in Journal
Cardioprotective Effects of SAR Through Attenuating Cardiac-Specific Markers, Inflammatory Markers, Oxidative Stress, and Anxiety in Rats Challenged with 5-Fluorouracil
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Xenobiotic Toxicants and Particulate Matter: Effects, Mechanisms, Impacts on Human Health, and Mitigation Strategies

by
Tamara Lang
1,2,
Anna-Maria Lipp
3 and
Christian Wechselberger
1,2,*
1
Department of Pathophysiology, Institute of Physiology and Pathophysiology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
2
Clinical Research Institute for Cardiovascular and Metabolic Diseases, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
3
Core Facility Cytometry, Center for Medical Research, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
*
Author to whom correspondence should be addressed.
J. Xenobiot. 2025, 15(4), 131; https://doi.org/10.3390/jox15040131
Submission received: 4 July 2025 / Revised: 7 August 2025 / Accepted: 12 August 2025 / Published: 14 August 2025

Abstract

Particulate matter (PM), a complex mixture of solid particles and liquid droplets, originates from both natural sources, such as sand, pollen, and marine salts, and anthropogenic activities, including vehicle emissions and industrial processes. While PM itself is not inherently toxic in all its forms, it often acts as a carrier of xenobiotic toxicants, such as heavy metals and organic pollutants, which adhere to its surface. This combination can result in synergistic toxic effects, significantly enhancing the potential harm to biological systems. Due to its small size and composition, PM can penetrate deep into the respiratory tract, acting as a physical “shuttle” that facilitates the distribution and bioavailability of toxic substances to distant organs. The omnipresence of PM in the environment leads to unavoidable and constant exposure, contributing to increased morbidity and mortality rates, particularly among vulnerable populations like the elderly, children, and individuals with pre-existing health conditions. This exposure also imposes a substantial financial burden on healthcare systems, as treating PM-related illnesses requires significant medical resources and leads to higher healthcare costs. Addressing these challenges necessitates effective mitigation strategies, including reducing PM exposure, improving air quality, and exploring novel approaches such as AI-based exposure prediction and nutritional interventions to protect public health and minimize the adverse effects of PM pollution.

1. Introduction

Particulate matter (PM) refers to a mixture of solid particles and liquid droplets found in the air, which vary in size and composition. PM is typically classified based on its aerodynamic diameter into categories such as PM10 (particles with diameters less than 10 micrometers), PM2.5 (particles with diameters less than 2.5 μm), and ultrafine particles (UFPs; particles with diameters less than 0.1 μm) [1]. PM in the atmosphere originates from both natural sources, such as dust storms, wildfires, and volcanic eruptions, and anthropogenic sources, including vehicle emissions, industrial processes, and construction activities [2,3,4,5].
Xenobiotic toxicants can cause harmful biological responses in the human body, which range from immunotoxicity and mitochondrial dysfunction to genotoxicity, organ damage, or cancer depending on the type, dose and duration of exposure [6]. The presence of toxic xenobiotics on the surface of PM can exacerbate the harmful effects on human health. This occurs because PM increases the bioavailability of xenobiotics by providing a larger surface area and acting as a carrier, enabling the delivery of these toxic substances deep into the respiratory system, where they can enter the bloodstream and reach various organs, causing systemic damage. Additionally, xenobiotic toxicants can amplify the inflammatory and oxidative effects of PM, resulting in more severe health outcomes. Understanding the effects of PM loaded with xenobiotic toxicants is therefore crucial for medical research, as exposure can convincingly be linked to various adverse health outcomes, including respiratory, neurological and cardiovascular diseases, and recent studies have already highlighted the importance of investigating these effects to develop effective public health strategies [7,8,9]. The chemical composition of environmental and industrial PM is complex and varies significantly. Characterizing the chemical composition and sources is therefore essential for assessing its potential impacts on human health [10]. Toxic xenobiotic substances, on the other hand, also include a broad spectrum of representatives (Table 1). For example, organic compounds can include polycyclic aromatic hydrocarbons (PAHs) and volatile organic compounds (VOCs), while heavy metals such as Pb, Hg, and Cd are often found in PM from industrial emissions [11,12,13]. Inorganic salts, including sulfates, nitrates, and ammonium, are commonly present in PM from both natural and human activities [14,15]. This review explores the toxic mechanisms of particulate matter (PM) containing xenobiotic toxicants to better understand their role in adverse health outcomes. Given that ambient PM is associated with 3–4 million deaths annually (approximately 7.6% of global mortality) and 103.1 million disability-adjusted life years (4.2% globally) [16], effective public health strategies and mitigation initiatives are also provided.

2. Environmental PM

Environmental PM exerts its effects through a complex chain of events. The first critical factor is the pathway of exposure, as the entry route strongly influences whether xenobiotic toxicants act locally or are transported systemically. Xenobiotic toxicants attached to PM can exert immediate toxic effects at the primary site of contact (e.g., lung tissue), whereas PM particles themselves can translocate across biological barriers and act as shuttles, distributing adsorbed toxicants to distant organs. These physicochemical interactions determine the particles’ bioavailability and capacity to cross biological barriers, ultimately dictating their tissue distribution. Once deposited in the target organs, PM and its associated toxicants can trigger immune responses, chronic inflammation, granuloma formation, and long-term disease outcomes (Figure 1).
The following subsections describe this process stepwise, starting with the exposure pathways to highlight their role in shaping the fate and toxicity of PM-associated substances, and then linking the particle–xenobiotic interactions, systemic distribution, and pathophysiological effects. Unlike previous reviews that considered PM and toxicants separately, this review specifically examines PM as an active carrier of xenobiotic substances, highlighting how these interactions shape the systemic distribution and amplify the toxicity.

2.1. Pathways of Human Exposure to PM

PM can enter the human body through various routes of exposure, each associated with specific health concerns [31]. Inhalation is the primary route, with minor contributions from ingestion, ocular exposure, and dermal absorption [37,38,39].
Upon inhalation, PM is deposited along the respiratory tract in a size-dependent manner: coarse particles (PM10, ≤10 μm) settle in the upper airways (e.g., nasal cavity, trachea), fine particles (PM2.5, ≤2.5 μm) reach the bronchi and alveoli, and UFPs (PM0.1, ≤0.1 μm) can translocate into the bloodstream [40]. Once in circulation, UFPs may affect extrapulmonary organs such as the liver, spleen, heart, and kidneys, or even bypass the blood–brain barrier via the olfactory nerve [37,41,42,43,44]. As a protective mechanism, PM and its soluble components are primarily cleared by mucociliary mechanisms and renal excretion [45,46].
PM is able to interact with various cell types in the lungs, including epithelial cells and macrophages, triggering immune responses and inflammation [47,48]. Smaller PM fractions, particularly UFPs, pose increased health risks due to their higher particle number and greater surface area at the same mass concentration [49]. This facilitates stronger interactions with biological membranes and enhanced uptake. Whereas uptake of coarse and fine PM is generally mediated by macrophages through receptor- and actin-dependent mechanisms, UFPs are internalized by non-specific pathways that also allow entry into other cell types, such as erythrocytes [50].
Beyond the lungs, ingestion represents another route of exposure, where contaminated food or water can introduce PM into the gastrointestinal tract [51,52,53,54]. Ingested PM with or without the associated xenobiotic toxicants can affect cells of the gastrointestinal tract, leading to inflammation and more severe issues like cancer development [55,56,57]. Dermal exposure, though less common, can occur via direct skin contact with airborne particles or contaminated surfaces, potentially leading to skin irritation, allergic reactions, and barrier disruption [57,58,59]. Ocular exposure is also increasingly recognized as a relevant route, where PM can contact the ocular surface, leading to oxidative stress, inflammation, and symptoms consistent with dry eye disease, as shown in both animal models and human studies [39,60,61].
Satellite-based monitoring of PM has emerged as a critical tool for assessing air quality on the regional and global scales, offering extensive spatial coverage that complements traditional ground-based monitoring networks. Current methods leverage satellite sensors, such as Moderate Resolution Imaging Spectroradiometer (MODIS), Multi-angle Imaging SpectroRadiometer (MISR), and Visible Infrared Imaging Radiometer Suite (VIIRS), to estimate the aerosol optical depth (AOD), which serves as a proxy for the PM concentration [62]. However, the resolution of satellite data, both spatial and temporal, remains a key consideration. High-resolution sensors, such as those on Sentinel-5P and upcoming missions like Tropospheric Emissions: Monitoring of Pollution (TEMPO), are improving the ability to capture fine-scale variations in PM levels [63]. Validation of satellite-derived PM estimates is essential to ensure the accuracy and reliability, typically achieved through comparison with ground-based measurements from networks like the Aerosol Robotic Network (AERONET) and regulatory air quality monitoring stations [64,65,66,67]. Data fusion techniques, such as machine learning models, geostatistical methods, and chemical transport models, are increasingly employed to integrate satellite data with ground-level observations, enhancing the spatial and temporal resolution of PM estimates [68]. These approaches enable the generation of high-quality, near-real-time PM datasets that are critical for epidemiological studies, exposure assessments, and policy-making. Despite these advancements, challenges remain, including accounting for the vertical aerosol distribution, cloud cover interference, and the need for region-specific calibration to address local atmospheric conditions. Continued innovation in satellite technology, combined with robust validation and integration frameworks, holds significant promise for advancing PM monitoring and its applications in public health and environmental management.

2.2. Interaction Between PM and Toxic Substances

The size of particles from environmental sources is inversely related to their surface area-to-volume ratio. As the particle size decreases, the surface area relative to the volume increases significantly, thus providing more active sites for interactions and making them highly effective at binding (e.g., hydrophobic drugs) [69,70]. This property is particularly relevant in environmental and biological contexts, where smaller particles can also act as carriers of xenobiotic and toxic compounds, influencing their transport and bioavailability. The combined impact of PM-induced nonspecific stress responses and the disruption of physiological processes at the cellular level can ultimately result in systemic inflammation and oxidative stress, which are further intensified and amplified by the biological activity of the xenobiotic toxicants [71]. This can facilitate the development of cardiovascular diseases, including heart attacks and hypertension, as well as the induction of neurological effects like cognitive decline and neurodegenerative diseases [7,72,73,74]. Furthermore, PM exposure can disrupt endocrine function and affect reproductive health, leading to serious issues such as birth defects and low birth weight [75,76,77].
The health impacts of PM exposure are not uniformly distributed across populations, with certain groups experiencing heightened vulnerability due to a combination of biological, social, and environmental factors. Vulnerable populations, such as children, the elderly, individuals with pre-existing health conditions, and those from lower socioeconomic backgrounds, are disproportionately affected by PM exposure. Exploring the interplay between PM exposure and sociodemographic factors requires a life-course perspective, which considers how cumulative exposures and critical windows of susceptibility (e.g., during early development or aging) influence health outcomes [78,79]. For instance, prenatal and early-life exposure to PM has been linked to adverse developmental outcomes, while chronic exposure in later life exacerbates cardiovascular and respiratory diseases [80,81,82]. Incorporating the theme of environmental justice is essential, as marginalized communities often face higher PM exposure due to proximity to pollution sources, such as industrial facilities, highways, and densely populated urban areas, while also having limited access to healthcare and resources to mitigate exposure. Health disparities are further amplified by systemic inequities, including housing quality, occupational exposures, and access to clean air. To address these issues, research should integrate sociodemographic data with PM exposure assessments using tools like geographic information systems (GISs), land-use regression models, and exposome frameworks [83,84,85,86]. Additionally, studies should examine the intersection of PM exposure with other social determinants of health, such as race, income, and education, to identify at-risk populations and inform targeted interventions. By adopting an interdisciplinary approach that combines environmental science, epidemiology, and social justice, researchers can better understand and address the inequities in PM-related health outcomes, ultimately guiding policies that promote environmental equity and public health.
The toxicokinetics and toxicodynamics of PM are strongly influenced by its chemical composition, particularly when it carries adsorbed xenobiotic and toxic substances such as metals, PAHs, and endotoxins [87]. These substances can modulate cellular responses, increase bioavailability, and exacerbate toxicity (Table 2). Some compounds may even dissociate from the particle core, entering systemic circulation and exhibiting independent toxicological profiles.
As shown in Table 3, xenobiotic toxicants can interact in various ways, with PM acting as a carrier of and delivery system for these associated toxicants, enhancing their distribution to target tissues and intensifying their biological effects. Health outcomes are further modulated by individual susceptibility, co-exposure to other pollutants, and overall exposure levels [89]. Recent advances in environmental toxicology have led to the development of integrated toxicokinetic/toxicodynamic (TK/TD) models, which simulate the PM deposition, translocation, and cellular effects over time. These models are critical for improving risk prediction and refining public health assessment strategies [16,88,99].

2.3. Localized and Systemic Effects of PM

Genuine xenobiotic and toxic substances often exert their toxicity directly or at least close to the site of contact since this is the area where the highest concentration of the substances is present [114]. Systemic effects, on the other hand, often need higher loads of contamination and/or longer times of exposure to the respective substances [115]. On the other hand, subsequent effects exerted through PM in combination with xenobiotic toxicants are often more profound, since translocation from the initial site of deposition to other parts of the body is relatively easy depending on the size of the particles and can occur, for example, with the help of phagocytic immune cells [116]. Upon entering the bloodstream, PM can disseminate to vital organs such as the heart, brain, and kidneys. In response, macrophages and other immune cells attempt to engulf and neutralize the particles, often resulting in the formation of granulomas—organized collections of immune cells and epithelial cells that encapsulate foreign materials to isolate them from the surrounding tissue, often associated with chronic inflammation [117,118,119,120]. The release of specific inflammatory cytokines and chemokines can later on cause tissue damage and remodeling [121]. The tissues and organs most commonly affected by granuloma formation due to the accumulation of such particulate matter include, but are not limited to, lungs, the lymph nodes and skin [122]. In addition, tissues and organs rich in capillary structures, such as the liver, spleen and kidneys, can develop granulomas in response to embedded foreign particles [122]. In general, the severity and location of granuloma formation depends on the type of particulate matter, the route of exposure, and the individual’s immune response [123]. Complications associated with granuloma formation range from impaired organ function to chronic inflammation and fibrosis [124]. Also, an increased risk of cancer development has already been associated with PM-induced granulomas, underscoring the relevance of this area of research [125,126].

2.4. Emerging Pollutants

Emerging pollutants such as nanoplastics, fullerenes, and other organic and inorganic nanoparticles pose significant risks to public health due to their unique physicochemical properties, including their small size, high surface area, and potential for bioaccumulation [127,128]. These particles can penetrate biological barriers, such as the respiratory and gastrointestinal tracts, and even cross the blood–brain barrier, leading to oxidative stress, inflammation, and cellular damage [129]. Nanoplastics, for instance, can adsorb toxic chemicals and heavy metals, acting as carriers that amplify their harmful effects [130,131]. Similarly, engineered nanoparticles like fullerenes may generate ROS, contributing to chronic diseases such as cancer, cardiovascular disorders, and neurodegenerative conditions [132,133]. The long-term impacts of these pollutants remain poorly understood, but their persistence in the environment and potential for widespread exposure underscore the urgent need for research and regulatory measures to mitigate their effects on human health and ecosystems.

3. Health Impacts of PM Exposure

3.1. Role of Oxidative Stress and Inflammation

One of the primary mechanisms by which PM exerts its harmful effects is through oxidative stress and inflammation [134]. This oxidative stress can trigger inflammatory responses, leading to the release of cytokines and other inflammatory mediators, and numerous studies have already demonstrated the role of oxidative stress and inflammation in PM-induced health effects [135,136,137]. The induction of oxidative stress occurs through several mechanisms once PM becomes deposited in the respiratory tract and interacts with, for example, epithelial cells and macrophages [138,139]. This effect is exacerbated if PM contains additional components, such as heavy metals and organic compounds, which can directly generate reactive oxygen species (ROS) or disrupt cellular antioxidant defenses [140]. Due to the presence of transition metals like Fe and Cu, ROS formation can be catalyzed through Fenton reactions [141]. Additionally, organic compounds in PM, such as PAHs, can undergo metabolic activation to produce ROS [142]. The excessive production of ROS ultimately overwhelms the cellular antioxidant capacity, leading to oxidative damage to lipids, proteins, and DNA. This oxidative stress therefore represents a key mechanism underlying the adverse health effects of PM exposure, including respiratory and cardiovascular diseases [143].
PM exposure can lead to the induction of inflammation in human cells and tissues through multiple pathways [144,145]. When PM is inhaled, it can activate epithelial cells and alveolar macrophages in the respiratory tract, triggering the release of pro-inflammatory cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β), which recruit and activate additional immune cells, including neutrophils and lymphocytes, amplifying the inflammatory response [146,147,148]. PM components, such as endotoxins and organic compounds, can furthermore interact with toll-like receptors (TLRs) on immune cells, further promoting inflammation [149,150]. Such a chronic activation of inflammatory pathways can ultimately lead to tissue damage and contribute to the development of diseases such as asthma, chronic obstructive pulmonary disease (COPD), and cardiovascular conditions [151,152]. Recent studies indicate that PM loaded with xenobiotic toxicants induces prolonged oxidative stress compared to exposure to particles alone, highlighting a synergistic effect that has been receiving growing scientific attention in the past few years [153,154,155].

3.2. Immune System Effects and Allergic Reactions to PM

PM also exerts significant modulatory effects on different cell types of the immune system, which vary with the particle size, concentration, chemical composition, and exposure duration (Table 4). These effects, both immunostimulatory and immunosuppressive, contribute to a spectrum of diseases, including respiratory, cardiovascular, autoimmune, and systemic inflammatory conditions [37,156,157]. Key PM constituents, such as metals, PAHs, nitro-PAHs, and endotoxins, critically influence immune interactions in dictating how PM interacts with immune cells [87,89]. For instance, PAHs and nitro-PAHs are known to activate the AhR pathway, which can alter immune cell function and promote inflammatory responses [158]. Similarly, heavy metals like lead, cadmium, and arsenic can disrupt cellular signaling pathways, impairing immune regulation and contributing to systemic inflammation [159]. The combination of PM’s physical properties and its xenobiotic toxicant load amplifies its potential to trigger or worsen immune-related diseases, highlighting the need for comprehensive strategies to address both PM emissions and the toxic substances they carry.

3.2.1. Activation of Innate Immunity and Chronic Inflammation

PM primarily activates the innate immune system, with alveolar macrophages, neutrophils, and dendritic cells (DCs) detecting PM components via pattern recognition receptors, such as TLRs [176,177]. This triggers the release of pro-inflammatory cytokines (e.g., IL-6, TNF-α, IL-1β) and chemokines, initiating acute inflammation. Chronic exposure sustains immune activation, leading to tissue damage and diseases like asthma, COPD, and lung fibrosis [178,179]. The PM composition also modulates immune responses. For example, endotoxins activate TLRs, while metals like Fe and Ni catalyze ROS formation, amplifying inflammation and oxidative stress [180]. PAHs and nitro-PAHs interact with aryl hydrocarbon receptors, altering DC function and promoting autoimmunity and T helper 2 cell (Th2)-skewed responses [181].

3.2.2. Disruption of Adaptive Immune Responses

PM also affects adaptive immunity by altering T helper cell subsets (Th1, Th2, Th17) and regulatory T cells (Tregs). PAHs promote Th2-skewed responses, exacerbating allergic inflammation and asthma, while metals like Cd drive Th17 responses linked to autoimmune diseases [164,182,183]. PM may impair Treg function, reducing immune tolerance and promoting chronic inflammation. Diesel exhaust particles enhance IgE production, contributing to allergic sensitization, while PM disrupts DC antigen presentation, weakening adaptive immunity [184,185,186].

3.2.3. Immunosuppression and Impaired Host Defense Mechanisms

Despite its pro-inflammatory effects, PM can suppress immune functions, impairing macrophage phagocytosis and NK cell cytotoxicity, which reduces pathogen clearance and increases the infection risk [187,188,189]. These effects are pronounced with PM containing organic toxicants or metals, leading to increased susceptibility to respiratory infections (e.g., influenza, pneumonia, COVID-19), reduced vaccine efficacy as well as heightened vulnerability to emerging pathogens [190,191].

3.2.4. Autoimmune Disorders and Neuroinflammation

Chronic PM exposure is linked to autoimmune diseases like rheumatoid arthritis and systemic lupus erythematosus, driven by metal and organic components that act as adjuvants, enhancing immune responses to self-antigens and promoting oxidative stress [192]. UFP can translocate into systemic circulation and the central nervous system (CNS), where it activates microglia, inducing neuroinflammation associated with cognitive decline and neurodegenerative diseases such as Alzheimer’s and Parkinson’s [42,193].
While evidence of PM-induced immunotoxicity is constantly growing, the exact mechanisms underlying the cellular uptake, translocation, and distribution of PM components still remain poorly understood. Investigating these pathways in relevant cell types will therefore provide critical insights into PM internalization and trafficking, guiding therapeutic and preventive strategies to mitigate its health impacts.

3.3. Impact of PM on the Respiratory System

When PM is inhaled, it deposits in the respiratory tract, including the alveoli and bronchioles. The size and composition of the particles determine their deposition site within the lungs, frequently triggering an immune response [194,195]. Respiratory diseases such as asthma and COPD are significantly impacted by chronic inflammation upon PM exposure [9,196,197]. PM can therefore exacerbate asthma symptoms and increase the frequency of asthma attacks, as well as contribute to the progression of COPD by inducing inflammation and oxidative stress in the respiratory tract [197]. PM2.5 and UFP can penetrate deep into the lungs, where these particles disrupt macrophage function, including the production of TNF-α, which is a critical cytokine involved in granuloma formation [198]. The associated granuloma formation, pulmonary fibrosis, and mucous hyperplasia compromise the immune response, potentially leading to more severe manifestation of pulmonary diseases [199,200,201]. As a consequence, these results also suggest that the more PM2.5 is inhaled in the lungs, the greater the potential risk to lung health becomes [202,203].
In addition to the effects described, xenobiotic toxicants bound to PM can elicit a range of additional health impacts by amplifying the toxicological burden on the respiratory system, for example, interference with cellular signaling pathways through heavy metal compounds, impairing the repair mechanisms of lung tissue and promoting chronic inflammation [204]. These metals can also induce DNA damage, increasing the risk of lung cancer and other malignancies [205]. Similarly, PAHs and nitro-PAHs, which are common components of PM, can generate ROS and oxidative stress, leading to lipid peroxidation, protein damage, and mitochondrial dysfunction in lung cells [206]. This could worsen conditions like asthma and COPD by enhancing airway hyperresponsiveness or impairing the clearance of pathogens, thereby increasing susceptibility to respiratory infections.

3.4. Impact of PM on the Cardiovascular System

PM can penetrate deep into the lungs and enter the bloodstream, triggering systemic inflammation, oxidative stress, as well as endothelial dysfunction, and thereby poses significant dangers also to the cells and tissues of the cardiovascular system. Since PM exposure is associated with chronic inflammation, the release of inflammatory cytokines and chemokines can cause tissue damage and remodeling. After these inflammatory mediators have entered the bloodstream, they can lead to systemic inflammation and oxidative stress, also affecting the cardiovascular system. PM can therefore lead to heart attacks, hypertension, and other cardiovascular conditions by promoting systemic inflammation, oxidative stress, and endothelial dysfunction [207,208,209]. Such processes can also ultimately lead to the development or exacerbation of conditions like hypertension, atherosclerosis, and arrhythmias, and long-term exposure to PM has already been linked to an increased risk of heart attack, stroke, and cardiovascular mortality [210].
Chronic inflammation also promotes the formation of plaques in the arteries, initiating the formation of atherosclerosis [211]. The characteristic plaques consist of lipids, immune cells, and fibrous tissue, also increasing the overall risk of heart attack and stroke [212]. Furthermore, inflammatory cytokines can enhance the coagulation cascade, leading to an increased risk of thrombotic blood clot formation [213]. The adverse cardiovascular effects of PM exposure are particularly pronounced in vulnerable populations such as the elderly, individuals with pre-existing cardiovascular conditions, and those with compromised immune systems. These groups may have heightened susceptibility to the harmful effects of PM-induced inflammation and oxidative stress [214].
In addition to the cardiovascular effects described, toxic xenobiotic substances bound to PM can further exacerbate these health risks by introducing additional mechanisms of toxicity. For example, heavy metals such as cadmium, lead, and arsenic can interfere with calcium signaling and ion channel function, potentially contributing to arrhythmias and impaired cardiac contractility [215,216]. Xenobiotic toxicants may also amplify the pro-thrombotic effects of PM exposure. For instance, certain metals and organic compounds can enhance platelet activation and aggregation, increasing the risk of thrombus formation and subsequent cardiovascular events such as heart attacks and strokes [217,218]. Additionally, these substances may disrupt lipid metabolism, leading to the accumulation of oxidized low-density lipoprotein (LDL) in arterial walls, which accelerates the development of atherosclerotic plaques [219].

3.5. Reproductive and Neurodevelopmental Effects of PM Exposure

Systemic inflammation and oxidative stress elicited by PM exposure can also affect placental function, impacting the tissue crucial for nutrient and oxygen exchange between the mother and the developing fetus [220,221]. Inflammation can impair placental blood flow and nutrient transport, ultimately leading to fetal growth restriction since the developing organs and systems of the fetus are highly sensitive to environmental insults. Furthermore, exposure to inflammatory mediators can disrupt normal developmental processes, leading to structural and functional abnormalities [222]. The reproductive and developmental effects of PM exposure that have been described include birth defects and low birth weight [223]. PM can affect fetal development by disrupting placental function and inducing oxidative stress and inflammation, linking maternal exposure to PM during pregnancy to adverse birth outcomes [224,225].
PM exposure during early periods of brain development is especially critical since it might increase the risk of the development of neurodevelopmental problems such as autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD) [226,227,228]. Prenatal and early-life exposure to air pollutants, particularly PM2.5 and UFP, nitrogen dioxide, and PAHs, has been associated with an increased risk of ASD [229]. These pollutants can cross the placental barrier or enter the developing brain, leading to neuroinflammation, oxidative stress, and disruptions to neural development [230]. Additionally, air pollution may interfere with critical signaling pathways and epigenetic mechanisms that regulate brain function during early development [230]. While the exact biological mechanisms remain under investigation, the growing body of evidence underscores the importance of reducing air pollution exposure during pregnancy and early childhood to mitigate potential neurodevelopmental risks.
Later in life, the neurological effects induced by PM exposure include cognitive decline and the development of neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease [231,232]. The chronic inflammation and oxidative stress accelerate the pathological processes underlying these conditions. Systemic inflammation and oxidative stress can compromise the integrity of the blood–brain barrier, which normally protects the brain from harmful substances. This disruption allows inflammatory mediators and potentially PM itself to enter the CNS and induce neuroinflammation, oxidative stress, and neuronal damage [233].
Once inside the CNS, PM and inflammatory mediators can activate the microglia, the resident immune cells of the brain. Activated microglia release pro-inflammatory cytokines and ROS, leading to a process known as neuroinflammation [234]. Such chronic neuroinflammation and oxidative stress can in the long term disrupt normal neuronal function, leading to synaptic dysfunction, impaired neurotransmission, and neuronal loss [235]. This can ultimately contribute to conditions such as mood disorders, depression or mild cognitive impairment, and it is also linked to an increased risk of the development of neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease [236].
Other studies found elevated levels of TNF-α, the 42-residue amyloid β peptide, and tau hyperphosphorylation on serine 199 in both the temporal and frontal lobes of rats undergoing sub-chronic exposure to diesel exhaust, indicating the upregulation of Alzheimer’s disease markers [237,238]. Additionally, the synuclein levels were elevated in children and young adults, suggesting that prolonged exposure to air pollution might be associated with such an early Parkinson’s disease-like pathology [239]. Another study also noted an increase in TNF-α levels over time, while the response of other pro-inflammatory factors was not consistent, implying that longer exposure to air pollution might trigger a compensatory response to neuroinflammation in the midbrain [240].
Persistent organic pollutants (POPs) and PAHs bound to PM can also disrupt placental function by interfering with trophoblast differentiation and invasion, processes essential for proper placental development and function. These substances may impair the production of key placental hormones, such as human chorionic gonadotropin (hCG) and placental growth factor (PlGF), further compromising fetal growth and increasing the risk of preterm birth or low birth weight [241]. Later in life, xenobiotic toxicant substances may amplify the risk of neurodegenerative diseases by promoting protein misfolding and aggregation [242]. For example, heavy metals like cadmium and lead can enhance the aggregation of amyloid-β and tau proteins, key pathological markers of Alzheimer’s disease, while also impairing the clearance of these proteins by the brain’s glymphatic system [243]. Similarly, exposure to PAHs and other organic pollutants may exacerbate α-synuclein aggregation, a hallmark of Parkinson’s disease, by inducing oxidative stress and mitochondrial dysfunction in dopaminergic neurons [244,245].

3.6. Genotoxic and Carcinogenic Effects of PM

Genotoxicity and carcinogenicity are also significant concerns following exposure to pollutants from the environment through various modes of action (Table 5). Genotoxicity refers to the ability to cause damage to the genetic material within cells, leading to mutations and chromosomal aberrations. Since PM exposure has been reported to indirectly cause damage to DNA, chronic exposure to PM can potentially contribute to the development of various forms of cancer [246]. In addition, the direct interaction of PM with biological molecules, such as proteins and DNA, can also disrupt normal cellular functions and promote carcinogenesis [247].
The various harmful components that can be contained in PM can interact with DNA directly or indirectly. PAHs can be metabolized into reactive intermediates that form DNA adducts, leading to mutations during DNA replication [248]. Heavy metals like As and Cd can interfere with DNA repair mechanisms, increasing the likelihood of mutations [13]. Additionally, PM-induced oxidative stress generates ROS that can cause oxidative damage to DNA, resulting in strand breaks and base modifications [249]. This DNA damage can activate signaling pathways that lead to cell cycle arrest, apoptosis, or uncontrolled cell proliferation if the damage is not properly repaired [4,250,251]. Chronic exposure to PM and persistent DNA damage can therefore result in the accumulation of mutations, which may contribute to the initiation and progression of various malignancies. The inflammatory response triggered by PM exposure has been convincingly linked to cancer development, as chronic inflammation can create a microenvironment that supports tumor growth and progression [252,253].
Another potential mechanism involves the disruption of the cellular redox balance by xenobiotic toxicants. For example, heavy metals and organic pollutants can deplete cellular antioxidants such as glutathione, exacerbating oxidative stress and increasing the production of ROS [254]. This heightened oxidative environment not only damages DNA but also promotes lipid peroxidation and protein oxidation, which can further disrupt cellular signaling pathways and contribute to carcinogenesis [255].
Table 5. Mechanisms of action of carcinogenic substances bound to particulate matter.
Table 5. Mechanisms of action of carcinogenic substances bound to particulate matter.
Mode of ActionDescriptionExamples of Relevant Substances
Oxidative StressGeneration of ROS leads to DNA damage, protein oxidation, and lipid peroxidation, promoting mutations and genomic instability [134].Arsenic (As) [256].
DNA Damage and MutagenesisDirect damage to DNA, such as strand breaks, cross-linking, or formation of DNA adducts, resulting in mutations that initiate carcinogenesis [257].Hexavalent Chromium (Cr(VI)), Benzo[a]pyrene (a PAH) [258,259].
Chronic InflammationPersistent activation of immune cells and release of pro-inflammatory cytokines generate ROS and reactive nitrogen species, indirectly damaging DNA [260].Crystalline Silica [261].
Epigenetic AlterationsChanges in DNA methylation, histone modification, or microRNA expression alter gene regulation, silencing tumor suppressor genes or activating oncogenes [262].Arsenic (As), Cadmium (Cd) [263,264].
Disruption of Cellular Signaling PathwaysInterference with pathways regulating cell proliferation, apoptosis, and DNA repair leads to uncontrolled cell growth and resistance to cell death [265].Cadmium (Cd), Lead (Pb) [216,266].
ROS, reactive oxygen species.

3.7. Influence of PM on the Human Microbiome

PM and xenobiotic toxicant chemicals have been shown to significantly impact the human microbiome of the respiratory tract as well as the gastrointestinal tract system through mechanisms such as inflammation, oxidative stress, and gut barrier dysfunction (Table 6) [267,268,269]. However, xenobiotic toxicants primarily act through chemical toxicity and interference with microbial metabolism, while particulate matter exerts its effects through physical interactions and the delivery of adsorbed toxins. These disruptions can lead to dysbiosis, increased intestinal permeability compromising the intestinal barrier and promoting the translocation of harmful substances into the bloodstream, and the development of diseases such as inflammatory bowel disease, metabolic disorders, and systemic inflammation [270]. When inhaled, PM and xenobiotic toxicant chemicals can alter the composition and diversity of the respiratory microbiome, leading to dysbiosis—a state of microbial imbalance [271]. This dysbiosis can weaken the immune defenses of the respiratory tract, increasing susceptibility to infections, inflammation, and chronic conditions such as asthma and COPD [272]. These changes in the gut microbiota have been linked to a range of pathophysiological consequences, including metabolic disorders, cardiovascular diseases, and neuroinflammatory conditions, and even were associated with an increased risk of early-onset Crohn disease [273,274].
Another potential mechanism involves the impact of xenobiotic toxicant substances on the gut–brain axis, the bidirectional communication network between the gut microbiome and the central nervous system. Dysbiosis induced by toxins can lead to the production of neuroactive metabolites, such as lipopolysaccharides (LPSs) and microbial-derived tryptophan metabolites, which can cross the blood–brain barrier and contribute to neuroinflammation, mood disorders, and neurodegenerative diseases [275]. For example, elevated levels of LPSs in the bloodstream, resulting from increased intestinal permeability, have been linked to systemic inflammation and neuroinflammatory conditions such as Alzheimer’s disease and Parkinson’s disease [276].
Recent studies utilizing whole-genome sequencing approaches, as well as a comprehensive examination of the literature, also suggest that exposure to air pollutants may increase the risk of obesity and type 2 diabetes through alterations to the gut microbiome [277,278,279].
Table 6. Impact of xenobiotic toxicant substances and particulate matter on microbial composition, inflammation, and host–microbe interactions.
Table 6. Impact of xenobiotic toxicant substances and particulate matter on microbial composition, inflammation, and host–microbe interactions.
Xenobiotic Toxicant SubstancesParticulate Matter
Disruption of Microbial CompositionChemicals or heavy metals can selectively inhibit beneficial microbes and promote pathogens (e.g., Pseudomonas aeruginosa) [280,281].Can act as a physical carrier for pathogenic microbes introducing new microbes and shifting microbial balance toward pro-inflammatory species [282].
Induction of InflammationDirectly activates immune cells, leading to cytokine release (e.g., IL-6, IL-8, and TNF-α), ROS generation and dysbiosis [283].Activates pattern recognition receptors (e.g., TLRs), inducing ROS and inflammatory cytokines; exacerbates oxidative stress via physical interaction with epithelial cells [284].
Impairment of Epithelial FunctionReduces ciliary function in the lung and mucus production, allowing microbial overgrowth; promoting epithelial integrity in the gastrointestinal system (“leaky gut” syndrome) [285,286].Physically obstructs cilia and increases mucus viscosity, impairing clearance in the lung; physically disrupts the mucus layer and weakens tight junctions in the gastrointestinal tract [287,288].
Alterations of Host–Microbe InteractionsReduces immune defenses, promoting pathogen colonization [289].Acts as an adjuvant, hyperactivating immune responses and disrupting microbial homeostasis [173,290].
IL, interleukin; TNF, tumor necrosis factor; ROS, reactive oxygen species; TLR, toll-like receptor.

3.8. Systemic Risk Modeling

Advances in computational toxicology and integrated risk assessment tools have revolutionized the ability to evaluate the health risks associated with PM and its adsorbed toxicants [291]. Computational toxicology leverages in silico models to predict the biological effects of chemical exposures, reducing reliance on traditional in vivo and in vitro methods [292]. Tools such as the Stochastic Human Exposure and Dose Simulation (SHEDS) model enable detailed exposure assessments by simulating human contact with environmental contaminants across various pathways and time scales [293]. Similarly, physiologically based pharmacokinetic (PBPK) models provide a mechanistic framework for predicting the absorption, distribution, metabolism, and excretion of chemicals within the body, offering insights into dose–response relationships and interindividual variability [294,295]. Exposome-based approaches further enhance risk assessment by integrating data on the totality of environmental exposures over a lifetime, including PM and its adsorbed components, with biological responses such as gene expression and metabolomics [296]. These tools are particularly valuable for assessing the risks posed by PM-bound toxicants, such as PAHs, heavy metals, and persistent organic pollutants, which can exacerbate the health impacts of PM exposure. By combining computational models with high-throughput data from omics technologies, researchers can identify critical exposure pathways, vulnerable populations, and potential intervention points. Moreover, these approaches facilitate the development of predictive models that account for the complex interactions between PM, co-pollutants, and host factors, ultimately supporting more accurate and comprehensive risk assessments [297]. As these tools continue to evolve, their integration into regulatory frameworks and public health strategies will be essential for mitigating the health risks associated with PM exposure.
A deeper understanding of source-specific PM toxicity is critical for designing targeted interventions to reduce health risks and inform actionable policy measures. Different PM sources, such as vehicular emissions, industrial activities, biomass burning, and secondary aerosols, contribute distinct chemical compositions and toxicological profiles [298]. For instance, PM from traffic emissions is often enriched with black carbon and PAHs, which are associated with oxidative stress and cardiopulmonary diseases, while biomass burning releases fine particles with high organic carbon content, linked to respiratory inflammation [299,300]. Source apportionment studies using techniques like receptor modeling, isotopic analysis, and chemical transport models can help identify the most harmful sources and their contributions to ambient PM levels [301]. These insights can guide policy measures, such as stricter emission standards for vehicles, promotion of cleaner industrial technologies, and incentives for renewable energy adoption. Urban areas, in particular, require targeted strategies to mitigate emissions from key sources, including the expansion of public transportation, implementation of low-emission zones, and regulation of construction dust. By focusing on source-specific toxicity and integrating these findings into health burden assessments, policymakers can prioritize interventions that yield the greatest public health benefits.

4. Strategies for Mitigating PM Exposure

Mitigation strategies for addressing PM exposure are essential to safeguard public health and improve air quality. Addressing these challenges requires a multifaceted approach that combines technological, regulatory, and behavioral interventions. Effective mitigation strategies should ideally focus on reducing PM emissions at their source but should also include enhancing air quality monitoring systems and promoting public awareness about protective measures. By implementing these strategies, governments, industries, and individuals can work together to minimize exposure to harmful particulate matter and create healthier living environments.

4.1. Regulatory Policies and Standards for PM Emission Control

Established standards represent critical tools in the fight against air pollution and its harmful effects on public health and the environment. An outstanding example in this respect is the Environmental Protection Agency (EPA), which has established regulations on the limits of PM concentrations in the air to protect public health. Regulatory frameworks, such as the Clean Air Act in the United States or the European Union’s Air Quality Standards, provide guidelines for monitoring, reporting, and enforcing compliance [302]. The EPA’s National Ambient Air Quality Standards (NAAQS) specify the permissible levels of PM10 and PM2.5, aiming to reduce exposure and the associated health risks [303].
In addition to setting concentration limits, regulatory policies increasingly emphasize the adoption of advanced technologies and best practices for particulate matter (PM) emission control at the source (see also Section 4.2). Furthermore, policies are evolving to address non-industrial sources of PM, such as emissions from transportation, residential heating, and agriculture [304,305]. Measures like promoting the use of electric vehicles, transitioning to cleaner fuels, and incentivizing sustainable agricultural practices are becoming integral components of comprehensive PM control strategies [306]. These proactive approaches will eventually not only help in meeting healthy air quality standards but also contribute to long-term sustainability goals by reducing the overall environmental footprint.

4.2. Technological Innovations in Emission Reduction

Recent advancements have become a cornerstone of the global effort to mitigate exposure to PM. Approaches in this field focus on reducing PM emissions from key sources such as industrial processes, transportation, and energy production. Innovations such as advanced filtration systems, catalytic converters, and cleaner fuel technologies have significantly reduced PM emissions from industrial processes and vehicles [307]. Such cutting-edge technologies are designed to capture and neutralize fine particles before they are released into the atmosphere. Additionally, the integration of real-time monitoring systems has enhanced the efficiency of emission control by enabling predictive maintenance and optimizing operational processes. These advancements are also necessary to help industries to comply with increasingly stringent environmental regulations. As technology continues to evolve, it therefore holds immense potential to further reduce PM emissions and address the challenges of air pollution on a global scale.
Another promising area of innovation in emission reduction is the development and deployment of nature-based and bio-inspired technologies. For example, biofiltration systems, which use living microorganisms to capture and degrade particulate matter and other pollutants, are gaining traction as sustainable alternatives to traditional filtration methods [308]. Similarly, urban greening initiatives, such as the strategic planting of trees and vegetation, have been shown to naturally filter airborne particles while providing additional benefits like carbon sequestration and urban cooling [309,310]. Emerging research is also exploring the use of nanotechnology to create advanced materials capable of capturing ultrafine particles with unprecedented efficiency [311]. These approaches could complement existing technological solutions and also align with broader environmental goals by promoting ecosystem health and reducing reliance on energy-intensive processes.

4.3. Approaches to Maintaining High Air Quality

This might represent one of the most critical factors in maintaining human health. The development of specialized filtration systems for different settings—including homes, workplaces, schools, and healthcare facilities—has emerged as a vital tool in mitigating health risks by removing particulate matter from the air [312,313,314,315]. High-efficiency particulate air (HEPA) filters have already been developed as a useful tool to remove PM, hence improving indoor air quality [316]. Other studies investigated the effects of portable air filtration systems on the exposure to PM and the correlation to blood pressure among residents of a low-income senior facility [317]. A comprehensive review dealt with various individual- and household-level interventions to reduce air pollution exposures and minimize the associated health risks [318]. The Reducing Air Pollution in Detroit Intervention Study was designed to evaluate the cardiovascular health benefits and personal fine particulate matter (PM2.5) exposure reductions achieved via portable air filtration units among older adults in Detroit, Michigan. It was also shown that commercially available HEPA-type and true HEPA personal air filters mitigated the median indoor PM2.5 concentrations by 58% and 65%, respectively [319].

4.4. Degradation of Xenobiotic Pollutants

Recent technological progress achieved in this area might represent the most effective and useful metabolic approach to degrading xenobiotic compounds like azo dyes, phenolics, PAHs, halogenated compounds, pesticides, nitroaromatic compounds, triazines, and chlorinated compounds [320,321]. Xenobiotic toxicant substances represent a major environmental pollution threat originating from both natural and human-made processes. The ability of different microorganisms to survive and thrive in the most diverse environments and also produce enzymes and metabolites that break down and inactivate pollutants could facilitate contaminated areas recovering naturally [322,323,324]. Additionally, emerging technologies like nanotechnology and photocatalysis are being explored to enhance the efficiency of degradation processes [325]. Genetic engineering of microbes to target specific xenobiotics and the development of bioaugmentation strategies further expand the potential for effective pollutant removal [326,327]. By harnessing these innovative approaches, it could eventually become possible to mitigate the long-term environmental and health impacts of xenobiotic pollutants, paving the way for cleaner and more sustainable ecosystems.

4.5. Role of Nutrition in Mitigating PM Effects

Components that are effective in countering the toxic effects of chemical contaminations in food include antioxidants, phytochemicals, essential fatty acids, and detoxification-supporting compounds. Together, nutritional components can work synergistically to support the protection of the body from the harmful effects of chemical contaminants in food. Antioxidants such as vitamins C and E, as well as selenium in the form of selenoproteins, play a crucial role in neutralizing ROS generated by chemical toxins, thereby reducing oxidative stress and preventing cellular damage [328]. Also, phytochemicals like flavonoids, carotenoids, and polyphenols, found in fruits, vegetables, and green tea, exhibit strong free-radical-scavenging properties and modulate detoxification pathways [329]. Sulfur-containing compounds, such as those found in garlic, onions, and cruciferous vegetables, enhance the activity of phase II detoxification enzymes in the liver, facilitating the conjugation and excretion of harmful xenobiotics [330]. Omega-3 fatty acids, present in fatty fish, flaxseeds, and walnuts, have anti-inflammatory properties that help mitigate inflammation caused by chemical contaminants [331]. Additionally, dietary fiber, found in whole grains, legumes, and fruits, binds to toxins in the gastrointestinal tract, promoting their elimination and reducing their absorption into the bloodstream [332]. Minerals also play a vital role in the detoxification processes of the human body by supporting various enzymatic functions. For example, Zn and Mg support enzymatic functions involved in liver detoxification pathways, particularly in neutralizing free radicals and repairing damaged tissues [333]. Se acts as a cofactor for glutathione peroxidase, a key antioxidant enzyme that helps eliminate harmful peroxides generated during detoxification [334]. Additionally, Fe and Cu are involved in the metabolism of toxins and the synthesis of detoxifying enzymes [335,336].

4.6. Applications of Artificial Intelligence in PM Management

The use of artificial intelligence (AI) will also prove helpful in the near future by playing a pivotal role in predicting potentially dangerous exposure to PM to minimize negative health effects for people or populations at risk. By leveraging machine learning algorithms and large datasets, AI has been already been described to analyze historical air quality data, meteorological conditions, and real-time sensor readings to forecast PM levels with high accuracy [337]. These predictions can help to identify high-risk areas and times, enabling authorities to issue timely warnings to the public. AI can also integrate satellite imagery and traffic data to pinpoint the sources of pollution, such as industrial emissions or vehicular traffic, and assess their impact on air quality [338]. Potential solutions include deploying AI-powered air quality monitoring systems in urban and industrial areas, optimizing traffic flows to reduce emissions, and implementing predictive maintenance for industrial equipment to minimize pollutant release. Additionally, AI can assist in designing personalized health alerts for vulnerable populations, such as children, the elderly, and individuals with respiratory conditions, advising them to take precautions during periods of high PM exposure [339]. Such public health interventions and awareness programs could prove essential for educating communities about the risks of PM exposure and promoting behaviors that reduce exposure. Initiatives such as air quality monitoring, public advisories, and community engagement can help individuals take protective measures. Future research should focus on understanding the long-term health effects of PM exposure and developing more effective mitigation strategies.
Besides systemic risk modeling, the advancements in AI offer transformative opportunities to address the complex interplay between PM exposure, health outcomes, and policy interventions. AI-driven tools, such as digital twins, enable the creation of virtual replicas of urban environments, allowing researchers to simulate the impacts of various emission reduction strategies on air quality and public health in real time [340]. Smart health early warning systems, powered by machine learning algorithms, can integrate air quality data with health surveillance systems to predict and mitigate acute health events, such as asthma exacerbations or cardiovascular emergencies, during high PM episodes [341,342]. Integrative frameworks that combine omics technologies (e.g., genomics, proteomics, and metabolomics), exposure modeling, and policy analysis provide a holistic approach to understanding PM-related health risks. For example, exposome-based frameworks can link molecular-level changes caused by PM exposure to population-level health outcomes, offering insights into both biological mechanisms and societal impacts [343]. By bridging the gap between scientific research, technological innovation, and policy implementation, these forward-looking approaches can drive more effective and equitable solutions to the challenges posed by PM pollution.

5. Conclusions

PM exposure poses significant health concerns and represents a potential epidemiological threat to society. The key findings indicate that PM exposure is associated with a range of adverse health effects, including respiratory diseases such as asthma and COPD, cardiovascular diseases like heart attack and hypertension, neurological effects such as cognitive decline and neurodegenerative diseases, and reproductive and developmental issues, including birth defects and low birth weight. These observations highlight the risks of potential epidemiological threats, particularly due to the effects of xenobiotic toxicants often associated with PM. These substances, such as heavy metals, PAHs, and VOCs, exacerbate the harmful effects of PM by amplifying oxidative stress, inflammation and systemic toxicity. These effects ultimately contribute to increased morbidity and mortality rates, particularly among vulnerable populations such as the elderly, children, and individuals with pre-existing health conditions.
PM pollution remains a pressing global challenge, with profound implications for public health, environmental sustainability, and social equity. The financial burden on the healthcare system can also be substantial, as treating PM-related illnesses requires significant medical resources and leads to higher healthcare costs. Addressing the multifaceted risks associated with PM exposure therefore requires a paradigm shift toward integrative frameworks that combine advanced scientific methodologies, predictive modeling, and evidence-based policy interventions. Omics technologies, such as genomics, proteomics, and metabolomics, offer unprecedented insights into the molecular mechanisms underlying PM-induced health effects, enabling the identification of biomarkers for exposure and susceptibility. When coupled with exposure modeling tools, such as land-use regression, satellite-based monitoring, and computational toxicology models, these approaches provide a comprehensive understanding of the pathways linking PM sources to health outcomes. Furthermore, the integration of these scientific advancements into policy frameworks is essential for translating research findings into actionable solutions. Policies informed by source-specific toxicity data, systemic risk modeling, and environmental justice considerations can prioritize interventions that maximize public health benefits while addressing disparities in exposure and vulnerability. By fostering collaboration across disciplines and sectors, integrative frameworks can bridge the gap between science and policy, driving innovative strategies to mitigate PM pollution and protect human health. This holistic approach is not only critical for managing current challenges but also for building resilience against future environmental and public health crises.
The importance of continued research and policy development to reduce PM exposure cannot be overstated. Effective regulatory policies, such as the EPA’s NAAQS, are crucial for setting limits on PM concentrations and protecting public health. Technological advancements in emission control, including advanced filtration systems and cleaner fuel technologies, play a vital role in reducing PM emissions. Public health interventions and awareness programs are essential for educating communities and promoting protective behaviors. Future research should focus on understanding the long-term health effects of PM exposure, developing more effective mitigation strategies, and assessing the economic benefits of reducing PM levels.

Author Contributions

Conceptualization, writing, review, and editing, T.L., A.-M.L., and C.W. All authors have read and agreed to the published version of the manuscript.

Funding

C.W. received financial support from the Medical Faculty of the Johannes Kepler University Linz through the internal funding program Impetus No. I-02-23.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

Supported by Johannes Kepler University Open Access Publishing Fund and the Federal State of Upper Austria.

Conflicts of Interest

The authors declare no competing interests.

Abbreviations

The following abbreviations are used in this manuscript:
ADHDAttention-deficit/hyperactivity disorder
AIArtificial intelligence
ASDAutism spectrum disorder
CNSCentral nervous system
COPDChronic obstructive pulmonary disease
DCDendritic cell
DDTDichlordiphenyltrichlorethan
EPAEnvironmental Protection Agency
HEPAHigh-efficiency particulate air
IL-1βInterleukin-1 beta
IL-6Interleukin-6
IL-8Interleukin-8
NAAQSNational Ambient Air Quality Standards
NKNatural killer cell
PAHPolycyclic aromatic hydrocarbons
PCBPolychlorinated biphenyl
PMParticulate matter
ROSReactive oxygen species
Th17T helper 17 cell
Th2T helper 2 cell
TLRToll-like receptor
TNF-αTumor necrosis factor-alpha
TregRegulatory T cell
UFPUltrafine particle
VOCVolatile organic compound

References

  1. Schraufnagel, D.E. The health effects of ultrafine particles. Exp. Mol. Med. 2020, 52, 311–317. [Google Scholar] [CrossRef] [PubMed]
  2. Farley, R.; Zhou, S.; Collier, S.; Jiang, W.; Onasch, T.B.; Shilling, J.E.; Kleinman, L.; Sedlacek Iii, A.J.; Zhang, Q. Chemical Evolution of Biomass Burning Aerosols across Wildfire Plumes in the Western U.S.: From Near-Source to Regional Scales. ACS EST Air 2025, 2, 677–691. [Google Scholar] [CrossRef] [PubMed]
  3. Shahpoury, P.; Schuster, J.; Harner, T. Oxidative potential of ambient particulate matter from community sites in Alberta’s oil sands region. Chemosphere 2025, 378, 144374. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, C.; Zhao, Y.; Liu, B.; Luo, Z.; Zhou, G.; Mao, K. PM2.5 from automobile exhaust induces apoptosis in male rat germ cells via the ROS-UPRmt signaling pathway. PLoS ONE 2025, 20, e0313803. [Google Scholar] [CrossRef]
  5. Ramadan, B.S.; Rosmalina, R.T.; Syafrudin; Munawir; Khair, H.; Widiyanti, A.; Rachman, I.; Matsumoto, T. Potential exposure to metals-bound particulate during open burning of different waste materials. Environ. Monit. Assess. 2025, 197, 495. [Google Scholar] [CrossRef]
  6. Sharma, K.; Kumar, P. Environmental threats posed by xenobiotics. In Bioremediation of Emerging Contaminants from Soils: Soil Health Conservation for Improved Ecology and Food Security; Kumar, P., Chaudhary, V., van Hullebusch, E.D., Busquets, R., Srivastav, A.L., Eds.; Elsevier: Amsterdam, The Netherlands; London, UK; Cambridge, MA, USA, 2024; pp. 183–201. ISBN 9780443139932. [Google Scholar]
  7. Balachandar, R.; Viramgami, A.; Singh, D.P.; Kulkarni, N.; Chudasama, B.; Sivaperumal, P.; Upadhyay, K. Association between chronic PM2.5 exposure and neurodegenerative biomarkers in adults from critically polluted area. BMC Public Health 2025, 25, 1413. [Google Scholar] [CrossRef]
  8. Jung, Y.S.; Johnson, M.M.; Burke, M.; Heft-Neal, S.; Bondy, M.L.; Chinthrajah, R.S.; Cullen, M.R.; Nelson, L.; Dresser, C.; Nadeau, K.C. Fine Particulate Matter From 2020 California Wildfires and Mental Health-Related Emergency Department Visits. JAMA Netw. Open 2025, 8, e253326. [Google Scholar] [CrossRef]
  9. Keleb, A.; Abeje, E.T.; Daba, C.; Endawkie, A.; Tsega, Y.; Abere, G.; Mamaye, Y.; Bezie, A.E. The odds of developing asthma and wheeze among children and adolescents exposed to particulate matter: Asystematic review and meta-analysis. BMC Public Health 2025, 25, 1225. [Google Scholar] [CrossRef]
  10. Sidwell, A.; Smith, S.C.; Roper, C. A comparison of fine particulate matter (PM2.5) in vivo exposure studies incorporating chemical analysis. J. Toxicol. Environ. Health B Crit. Rev. 2022, 25, 422–444. [Google Scholar] [CrossRef]
  11. León-Mejía, G.; Cappetta, M.; Letícia Hilário Garcia, A.; Fiorillo-Moreno, O.; Rohr, P.; Muñoz-Acevedo, A.; Miranda-Guevara, A.; Quintana-Sosa, M.; Martinez-Lopez, W.; Pêgas Henriques, J.A.; et al. Global DNA methylation and its association with genetic instability and exposure to inorganic elements and polycyclic aromatic hydrocarbons in coal mining dust. Mutagenesis 2025, 1–9. [Google Scholar] [CrossRef]
  12. Saldanha, Í.S.; Rocha, C.A.; Pontes, F.; Santos, R.P.; Nascimento, R.F.; Costa, A.B.; Bertoncini, B.; Cavalcante, R.M. Influence of source contributions and seasonal variations on particle-associated polycyclic aromatic hydrocarbons and cancer risks in a rapidly urbanizing South American City (Fortaleza, Brazil). Environ. Toxicol. Chem. 2025, 44, 1378–1388. [Google Scholar] [CrossRef]
  13. Wechselberger, C.; Messner, B.; Bernhard, D. The Role of Trace Elements in Cardiovascular Diseases. Toxics 2023, 11, 956. [Google Scholar] [CrossRef]
  14. Badami, M.M.; Aghaei, Y.; Sioutas, C. Impact of Emission Standards on Fine Particulate Matter Toxicity: A Long-Term Analysis in Los Angeles. Toxics 2025, 13, 140. [Google Scholar] [CrossRef]
  15. Faruqui, N.; Orell, S.; Dondi, C.; Leni, Z.; Kalbermatter, D.M.; Gefors, L.; Rissler, J.; Vasilatou, K.; Mudway, I.S.; Kåredal, M.; et al. Differential Cytotoxicity and Inflammatory Responses to Particulate Matter Components in Airway Structural Cells. Int. J. Mol. Sci. 2025, 26, 830. [Google Scholar] [CrossRef] [PubMed]
  16. Burnett, R.T.; Pope, C.A.; Ezzati, M.; Olives, C.; Lim, S.S.; Mehta, S.; Shin, H.H.; Singh, G.; Hubbell, B.; Brauer, M.; et al. An integrated risk function for estimating the global burden of disease attributable to ambient fine particulate matter exposure. Environ. Health Perspect. 2014, 122, 397–403. [Google Scholar] [CrossRef] [PubMed]
  17. Lushchak, V.I.; Matviishyn, T.M.; Husak, V.V.; Storey, J.M.; Storey, K.B. Pesticide toxicity: A mechanistic approach. EXCLI J. 2018, 17, 1101–1136. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, X.; Sial, M.U.; Bashir, M.A.; Bilal, M.; Raza, Q.-U.-A.; Ali Raza, H.M.; Rehim, A.; Geng, Y. Pesticides Xenobiotics in Soil Ecosystem and Their Remediation Approaches. Sustainability 2022, 14, 3353. [Google Scholar] [CrossRef]
  19. Anzenbacher, P. Metabolism of Drugs and Other Xenobiotics; John Wiley & Sons: Hoboken, NJ, USA, 2012; ISBN 9783527329038. [Google Scholar]
  20. Stapleton, P.A.; Minarchick, V.C.; McCawley, M.; Knuckles, T.L.; Nurkiewicz, T.R. Xenobiotic particle exposure and microvascular endpoints: A call to arms. Microcirculation 2012, 19, 126–142. [Google Scholar] [CrossRef]
  21. Mussalo, L.; Avesani, S.; Shahbaz, M.A.; Závodná, T.; Saveleva, L.; Järvinen, A.; Lampinen, R.; Belaya, I.; Krejčík, Z.; Ivanova, M.; et al. Emissions from modern engines induce distinct effects in human olfactory mucosa cells, depending on fuel and aftertreatment. Sci. Total Environ. 2023, 905, 167038. [Google Scholar] [CrossRef]
  22. Patterson, A.D.; Gonzalez, F.J.; Idle, J.R. Xenobiotic metabolism: A view through the metabolometer. Chem. Res. Toxicol. 2010, 23, 851–860. [Google Scholar] [CrossRef]
  23. Zhu, Y.; Boye, A.; Body-Malapel, M.; Herkovits, J. The Toxic Effects of Xenobiotics on the Health of Humans and Animals. BioMed Res. Int. 2017, 2017, 4627872. [Google Scholar] [CrossRef] [PubMed]
  24. Ramaka, S. Xenobiotics in Health and Disease: The Two Sides of a Coin: A Clinician’s Perspective. Open Acc. J. Toxicol. 2020, 4, 555641. [Google Scholar] [CrossRef]
  25. Ghio, A.J.; Kummarapurugu, S.T.; Tong, H.; Soukup, J.M.; Dailey, L.A.; Boykin, E.; Ian Gilmour, M.; Ingram, P.; Roggli, V.L.; Goldstein, H.L.; et al. Biological effects of desert dust in respiratory epithelial cells and a murine model. Inhal. Toxicol. 2014, 26, 299–309. [Google Scholar] [CrossRef]
  26. Hammond, J.; Maher, B.A.; Gonet, T.; Bautista, F.; Allsop, D. Oxidative Stress, Cytotoxic and Inflammatory Effects of Urban Ultrafine Road-Deposited Dust from the UK and Mexico in Human Epithelial Lung (Calu-3) Cells. Antioxidants 2022, 11, 1814. [Google Scholar] [CrossRef]
  27. Dahl, A.R.; Lewis, J.L. Respiratory tract uptake of inhalants and metabolism of xenobiotics. Annu. Rev. Pharmacol. Toxicol. 1993, 33, 383–407. [Google Scholar] [CrossRef]
  28. Brittebo, E.B.; Brandt, I. Interactions of xenobiotics in the respiratory tract following non-inhalation routes of exposure. Int. J. Epidemiol. 1990, 19 (Suppl. S1), S24–S31. [Google Scholar] [CrossRef]
  29. Collins, S.L.; Patterson, A.D. The gut microbiome: An orchestrator of xenobiotic metabolism. Acta Pharm. Sin. B 2020, 10, 19–32. [Google Scholar] [CrossRef]
  30. Thomson, E.M.; Vladisavljevic, D.; Mohottalage, S.; Kumarathasan, P.; Vincent, R. Mapping acute systemic effects of inhaled particulate matter and ozone: Multiorgan gene expression and glucocorticoid activity. Toxicol. Sci. 2013, 135, 169–181. [Google Scholar] [CrossRef]
  31. Thangavel, P.; Park, D.; Lee, Y.-C. Recent Insights into Particulate Matter (PM2.5)-Mediated Toxicity in Humans: An Overview. Int. J. Environ. Res. Public Health 2022, 19, 7511. [Google Scholar] [CrossRef]
  32. Gonzalez, F.J.; Kimura, S. Understanding the role of xenobiotic-metabolism in chemical carcinogenesis using gene knockout mice. Mutat. Res. 2001, 477, 79–87. [Google Scholar] [CrossRef]
  33. Stradtman, S.C.; Freeman, J.L. Mechanisms of Neurotoxicity Associated with Exposure to the Herbicide Atrazine. Toxics 2021, 9, 207. [Google Scholar] [CrossRef]
  34. Xing, Y.-F.; Xu, Y.-H.; Shi, M.-H.; Lian, Y.-X. The impact of PM2.5 on the human respiratory system. J. Thorac. Dis. 2016, 8, E69–E74. [Google Scholar] [CrossRef] [PubMed]
  35. Hamanaka, R.B.; Mutlu, G.M. Particulate Matter Air Pollution: Effects on the Cardiovascular System. Front. Endocrinol. 2018, 9, 680. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, H.; Zhang, H.; Li, J.; Liao, J.; Liu, J.; Hu, C.; Sun, X.; Zheng, T.; Xia, W.; Xu, S.; et al. Prenatal and early postnatal exposure to ambient particulate matter and early childhood neurodevelopment: A birth cohort study. Environ. Res. 2022, 210, 112946. [Google Scholar] [CrossRef] [PubMed]
  37. Brook, R.D.; Rajagopalan, S.; Pope, C.A.; Brook, J.R.; Bhatnagar, A.; Diez-Roux, A.V.; Holguin, F.; Hong, Y.; Luepker, R.V.; Mittleman, M.A.; et al. Particulate matter air pollution and cardiovascular disease: An update to the scientific statement from the American Heart Association. Circulation 2010, 121, 2331–2378. [Google Scholar] [CrossRef]
  38. Thompson, J.E. Airborne Particulate Matter: Human Exposure and Health Effects. J. Occup. Environ. Med. 2018, 60, 392–423. [Google Scholar] [CrossRef]
  39. Iqbal, S.; Ramini, A.; Kaja, S. Impact of particulate matter and air pollution on ocular surface disease: A systematic review of preclinical and clinical evidence. Ocul. Surf. 2025, 35, 100–116. [Google Scholar] [CrossRef]
  40. Löndahl, J.; Möller, W.; Pagels, J.H.; Kreyling, W.G.; Swietlicki, E.; Schmid, O. Measurement techniques for respiratory tract deposition of airborne nanoparticles: A critical review. J. Aerosol Med. Pulm. Drug Deliv. 2014, 27, 229–254. [Google Scholar] [CrossRef]
  41. Tian, Y.; Li, Y.; Sun, S.; Dong, Y.; Tian, Z.; Zhan, L.; Wang, X. Effects of urban particulate matter on the quality of erythrocytes. Chemosphere 2023, 313, 137560. [Google Scholar] [CrossRef]
  42. Block, M.L.; Calderón-Garcidueñas, L. Air pollution: Mechanisms of neuroinflammation and CNS disease. Trends Neurosci. 2009, 32, 506–516. [Google Scholar] [CrossRef]
  43. Nemmar, A.; Hoet, P.H.M.; Vanquickenborne, B.; Dinsdale, D.; Thomeer, M.; Hoylaerts, M.F.; Vanbilloen, H.; Mortelmans, L.; Nemery, B. Passage of inhaled particles into the blood circulation in humans. Circulation 2002, 105, 411–414. [Google Scholar] [CrossRef]
  44. Kreyling, W.G.; Semmler, M.; Erbe, F.; Mayer, P.; Takenaka, S.; Schulz, H.; Oberdörster, G.; Ziesenis, A. Translocation of ultrafine insoluble iridium particles from lung epithelium to extrapulmonary organs is size dependent but very low. J. Toxicol. Environ. Health A 2002, 65, 1513–1530. [Google Scholar] [CrossRef] [PubMed]
  45. Takenaka, S.; Karg, E.; Roth, C.; Schulz, H.; Ziesenis, A.; Heinzmann, U.; Schramel, P.; Heyder, J. Pulmonary and systemic distribution of inhaled ultrafine silver particles in rats. Environ. Health Perspect. 2001, 109 (Suppl. S4), 547–551. [Google Scholar] [CrossRef] [PubMed]
  46. Georgopoulos, P.G.; Wang, S.-W.; Vyas, V.M.; Sun, Q.; Burke, J.; Vedantham, R.; McCurdy, T.; Ozkaynak, H. A source-to-dose assessment of population exposures to fine PM and ozone in Philadelphia, PA, during a summer 1999 episode. J. Expo. Anal. Environ. Epidemiol. 2005, 15, 439–457. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, Z.; Zhu, Q.; Song, E.; Song, Y. Characterization of blood protein adsorption on PM2.5 and its implications on cellular uptake and cytotoxicity of PM2.5. J. Hazard. Mater. 2021, 414, 125499. [Google Scholar] [CrossRef]
  48. Michael, S.; Montag, M.; Dott, W. Pro-inflammatory effects and oxidative stress in lung macrophages and epithelial cells induced by ambient particulate matter. Environ. Pollut. 2013, 183, 19–29. [Google Scholar] [CrossRef]
  49. Oberdörster, G.; Oberdörster, E.; Oberdörster, J. Nanotoxicology: An emerging discipline evolving from studies of ultrafine particles. Environ. Health Perspect. 2005, 113, 823–839. [Google Scholar] [CrossRef]
  50. Peters, A.; Veronesi, B.; Calderón-Garcidueñas, L.; Gehr, P.; Chen, L.C.; Geiser, M.; Reed, W.; Rothen-Rutishauser, B.; Schürch, S.; Schulz, H. Translocation and potential neurological effects of fine and ultrafine particles a critical update. Part. Fibre Toxicol. 2006, 3, 13. [Google Scholar] [CrossRef]
  51. Noh, K.; Thi, L.T.; Jeong, B.R. Particulate matter in the cultivation area may contaminate leafy vegetables with heavy metals above safe levels in Korea. Environ. Sci. Pollut. Res. Int. 2019, 26, 25762–25774. [Google Scholar] [CrossRef]
  52. Kong, H.K.; Yoon, D.K.; Lee, H.W.; Lee, C.M. Evaluation of particulate matter concentrations according to cooking activity in a residential environment. Environ. Sci. Pollut. Res. Int. 2021, 28, 2443–2456. [Google Scholar] [CrossRef]
  53. Hussain, N.; Jaitley, V.; Florence, A.T. Recent advances in the understanding of uptake of microparticulates across the gastrointestinal lymphatics. Adv. Drug Deliv. Rev. 2001, 50, 107–142. [Google Scholar] [CrossRef]
  54. Delon, L.; Gibson, R.J.; Prestidge, C.A.; Thierry, B. Mechanisms of uptake and transport of particulate formulations in the small intestine. J. Control. Release 2022, 343, 584–599. [Google Scholar] [CrossRef]
  55. Sun, D.; Liu, C.; Zhu, Y.; Yu, C.; Guo, Y.; Sun, D.; Pang, Y.; Pei, P.; Du, H.; Yang, L.; et al. Long-Term Exposure to Fine Particulate Matter and Incidence of Esophageal Cancer: A Prospective Study of 0.5 Million Chinese Adults. Gastroenterology 2023, 165, 61–70.e5. [Google Scholar] [CrossRef]
  56. Zhang, Q.; Wang, X.; Chen, Y.; Song, G.; Zhang, H.; Huang, K.; Luo, Y.; Cheng, N. Discovery and solution for microplastics: New risk carriers in food. Food Chem. 2025, 471, 142784. [Google Scholar] [CrossRef]
  57. Balasch, A.; Moreno, T.; Eljarrat, E. Assessment of Daily Exposure to Organophosphate Esters through PM2.5 Inhalation, Dust Ingestion, and Dermal Contact. Environ. Sci. Technol. 2023, 57, 20669–20677. [Google Scholar] [CrossRef]
  58. Fernando, P.D.S.M.; Piao, M.J.; Zhen, A.X.; Ahn, M.J.; Yi, J.M.; Choi, Y.H.; Hyun, J.W. Extract of Cornus officinalis Protects Keratinocytes from Particulate Matter-induced Oxidative Stress. Int. J. Med. Sci. 2020, 17, 63–70. [Google Scholar] [CrossRef]
  59. Piao, M.J.; Ahn, M.J.; Kang, K.A.; Ryu, Y.S.; Hyun, Y.J.; Shilnikova, K.; Zhen, A.X.; Jeong, J.W.; Choi, Y.H.; Kang, H.K.; et al. Particulate matter 2.5 damages skin cells by inducing oxidative stress, subcellular organelle dysfunction, and apoptosis. Arch. Toxicol. 2018, 92, 2077–2091. [Google Scholar] [CrossRef]
  60. Yang, Q.; Li, K.; Li, D.; Zhang, Y.; Liu, X.; Wu, K. Effects of fine particulate matter on the ocular surface: An in vitro and in vivo study. Biomed. Pharmacother. 2019, 117, 109177. [Google Scholar] [CrossRef]
  61. Yu, D.; Cai, W.; Shen, T.; Wu, Y.; Ren, C.; Li, T.; Hu, C.; Zhu, M.; Yu, J. PM2.5 exposure increases dry eye disease risks through corneal epithelial inflammation and mitochondrial dysfunctions. Cell Biol. Toxicol. 2023, 39, 2615–2630. [Google Scholar] [CrossRef]
  62. Ranjan, A.K.; Patra, A.K.; Gorai, A.K. A Review on Estimation of Particulate Matter from Satellite-Based Aerosol Optical Depth: Data, Methods, and Challenges. Asia-Pac. J. Atmos. Sci. 2021, 57, 679–699. [Google Scholar] [CrossRef]
  63. Batista, T.; Jamal, S.A.; Isbaex, C. Sentinel Data for Monitoring of Pollutant Emissions by Maritime Transport—A Literature Review. Remote Sens. 2025, 17, 2202. [Google Scholar] [CrossRef]
  64. Sarigiannis, D.A.; Saisana, M. Multi-objective optimization of air quality monitoring. Environ. Monit. Assess. 2008, 136, 87–99. [Google Scholar] [CrossRef]
  65. Schäfer, K.; Fömmel, G.; Hoffmann, H.; Briz, S.; Junkermann, W.; Emeis, S.; Jahn, C.; Leipold, S.; Sedlmaier, A.; Dinev, S.; et al. Three-Dimensional Ground-Based Measurements of Urban Air Quality to Evaluate Satellite Derived Interpretations for Urban Air Pollution. Water Air Soil Pollut. Focus 2002, 2, 91–102. [Google Scholar] [CrossRef]
  66. van Donkelaar, A.; Martin, R.V.; Pasch, A.N.; Szykman, J.J.; Zhang, L.; Wang, Y.X.; Chen, D. Improving the accuracy of daily satellite-derived ground-level fine aerosol concentration estimates for North America. Environ. Sci. Technol. 2012, 46, 11971–11978. [Google Scholar] [CrossRef]
  67. Terenzi, V.; Tratzi, P.; Paolini, V.; Ianniello, A.; Barnaba, F.; Bassani, C. Comprehensive Validation of MODIS-MAIAC Aerosol Products and Long-Term Aerosol Detection over an Urban–Rural Area Around Rome in Central Italy. Remote Sens. 2025, 17, 2051. [Google Scholar] [CrossRef]
  68. Liu, Y.; Cao, G.; Zhao, N. Integrate machine learning and geostatistics for high-resolution mapping of ground-level PM2.5 concentrations. In Spatiotemporal Analysis of Air Pollution and Its Application in Public Health; Li, L., Ed.; Elsevier: San Diego, CA, USA, 2020; pp. 135–151. ISBN 9780128158227. [Google Scholar]
  69. Yadollahi, R.; Vasilev, K.; Simovic, S. Nanosuspension Technologies for Delivery of Poorly Soluble Drugs. J. Nanomater. 2015, 2015, 216375. [Google Scholar] [CrossRef]
  70. Nikam, T.; Nikam, R.; Sharma, Y.; Patil, D. Applications Of Nanoparticle System In Drug Delivery Technology. Int. J. Pharm. Sci. 2024, 2, 103–118. [Google Scholar] [CrossRef]
  71. Chaiwong, W.; Liwsrisakun, C.; Inchai, J.; Duangjit, P.; Bumroongkit, C.; Deesomchok, A.; Theerakittikul, T.; Limsukon, A.; Tajarernmuang, P.; Niyatiwatchanchai, N.; et al. Biomarkers of Oxidative Stress, Systemic Inflammation and Thrombosis in Adult Asthmatic Patients Treated with Inhaled Corticosteroids During Exposure to Fine Particulate Matter. J. Clin. Med. 2025, 14, 2360. [Google Scholar] [CrossRef]
  72. Yuan, X.; Yang, Y.; Liu, C.; Tian, Y.; Xia, D.; Liu, Z.; Pan, L.; Xiong, M.; Xiong, J.; Meng, L.; et al. Fine Particulate Matter Triggers α-Synuclein Fibrillization and Parkinson-like Neurodegeneration. Mov. Disord. 2022, 37, 1817–1830. [Google Scholar] [CrossRef]
  73. Jiang, L.; Shao, M.; Song, C.; Zhou, L.; Nie, W.; Yu, H.; Wang, S.; Liu, Y.; Yu, L. The Role of Epigenetic Mechanisms in the Development of PM2.5-Induced Cognitive Impairment. Toxics 2025, 13, 119. [Google Scholar] [CrossRef]
  74. Xia, Z.; Liu, Y.; Liu, C.; Dai, Z.; Liang, X.; Zhang, N.; Wu, W.; Wen, J.; Zhang, H. The causal effect of air pollution on the risk of essential hypertension: A Mendelian randomization study. Front. Public Health 2024, 12, 1247149. [Google Scholar] [CrossRef]
  75. Jung, G.S.; Lee, J.H.; Lee, M.J.; Lee, I.; Park, H.; Kim, N.; Kim, J.-Y.; Im, W.; Cho, S.; Choi, Y.S. Effects of chronic particulate matter exposure on endometriosis-associated signaling pathways and disease progression. Mol. Hum. Reprod. 2025, 31, gaaf013. [Google Scholar] [CrossRef]
  76. Wang, H.; Xu, J.; Huang, C.; Chen, L.; Zhang, X.; Tian, W. The association between particulate matter 2.5 and thyroid function and thyroid cancer: A meta-analysis. Int. Arch. Occup. Environ. Health 2025, 98, 267–282. [Google Scholar] [CrossRef]
  77. Li, F.; Duan, X.; Li, M.; Gao, Y.; Kang, Y.; Zheng, W.; Guo, X.; Chen, Y. Environmental pollution and human fertility: Investigating the relationship between PM2.5 exposure and assisted reproductive technology outcomes. BMC Public Health 2025, 25, 1357. [Google Scholar] [CrossRef]
  78. Chapizanis, D.; Karakitsios, S.; Gotti, A.; Sarigiannis, D.A. Assessing personal exposure using Agent Based Modelling informed by sensors technology. Environ. Res. 2021, 192, 110141. [Google Scholar] [CrossRef]
  79. Karakoltzidis, A.; Agalliadou, A.; Kermenidou, M.; Nikiforou, F.; Chatzimpaloglou, A.; Feleki, E.; Karakitsios, S.; Gotti, A.; Sarigiannis, D.A. Agent-based modelling: A stochastic approach to assessing personal exposure to environmental pollutants-Insights from the URBANOME project. Sci. Total Environ. 2025, 967, 178804. [Google Scholar] [CrossRef]
  80. Syama, K.P.; Blais, E.; Kumarathasan, P. Maternal mechanisms in air pollution exposure-related adverse pregnancy outcomes: A systematic review. Sci. Total Environ. 2025, 970, 178999. [Google Scholar] [CrossRef]
  81. Rivera Rivera, N.Y.; Tamayo-Ortiz, M.; Mercado García, A.; Just, A.C.; Kloog, I.; Téllez-Rojo, M.M.; Wright, R.O.; Wright, R.J.; Rosa, M.J. Prenatal and early life exposure to particulate matter, environmental tobacco smoke and respiratory symptoms in Mexican children. Environ. Res. 2021, 192, 110365. [Google Scholar] [CrossRef]
  82. Gheissari, R.; Liao, J.; Garcia, E.; Pavlovic, N.; Gilliland, F.D.; Xiang, A.H.; Chen, Z. Health Outcomes in Children Associated with Prenatal and Early-Life Exposures to Air Pollution: A Narrative Review. Toxics 2022, 10, 458. [Google Scholar] [CrossRef]
  83. Sarigiannis, D.A.; Karakitsios, S.P.; Kermenidou, M.; Nikolaki, S.; Zikopoulos, D.; Semelidis, S.; Papagiannakis, A.; Tzimou, R. Total exposure to airborne particulate matter in cities: The effect of biomass combustion. Sci. Total Environ. 2014, 493, 795–805. [Google Scholar] [CrossRef]
  84. Ma, X.; Zou, B.; Deng, J.; Gao, J.; Longley, I.; Xiao, S.; Guo, B.; Wu, Y.; Xu, T.; Xu, X.; et al. A comprehensive review of the development of land use regression approaches for modeling spatiotemporal variations of ambient air pollution: A perspective from 2011 to 2023. Environ. Int. 2024, 183, 108430. [Google Scholar] [CrossRef]
  85. Wan Azmi, W.N.F.; Pillai, T.R.; Latif, M.T.; Shaharudin, R.; Koshy, S. Development of land use regression model to estimate particulate matter (PM10) and nitrogen dioxide (NO2) concentrations in Peninsular Malaysia. Atmos. Environ. X 2024, 21, 100244. [Google Scholar] [CrossRef]
  86. Cai, J.; Ge, Y.; Li, H.; Yang, C.; Liu, C.; Meng, X.; Wang, W.; Niu, C.; Kan, L.; Schikowski, T.; et al. Application of land use regression to assess exposure and identify potential sources in PM2.5, BC, NO2 concentrations. Atmos. Environ. 2020, 223, 117267. [Google Scholar] [CrossRef]
  87. Gilmour, M.I.; McGee, J.; Duvall, R.M.; Dailey, L.; Daniels, M.; Boykin, E.; Cho, S.-H.; Doerfler, D.; Gordon, T.; Devlin, R.B. Comparative toxicity of size-fractionated airborne particulate matter obtained from different cities in the United States. Inhal. Toxicol. 2007, 19 (Suppl. S1), 7–16. [Google Scholar] [CrossRef]
  88. Bakr, S.; Sayed, M.A.; Salem, K.M.; Morsi, E.M.; Masoud, M.; Ezzat, E.M. Lead (Pb) and cadmium (Cd) blood levels and potential hematological health risk among inhabitants of the claimed hazardous region around Qaroun Lake in Egypt. BMC Public Health 2023, 23, 1071. [Google Scholar] [CrossRef]
  89. Kelly, F.J.; Fussell, J.C. Size, source and chemical composition as determinants of toxicity attributable to ambient particulate matter. Atmos. Environ. 2012, 60, 504–526. [Google Scholar] [CrossRef]
  90. Jomova, K.; Alomar, S.Y.; Nepovimova, E.; Kuca, K.; Valko, M. Heavy metals: Toxicity and human health effects. Arch. Toxicol. 2025, 99, 153–209. [Google Scholar] [CrossRef] [PubMed]
  91. Vidrio, E.; Jung, H.; Anastasio, C. Generation of Hydroxyl Radicals from Dissolved Transition Metals in Surrogate Lung Fluid Solutions. Atmos. Environ. 2008, 42, 4369–4379. [Google Scholar] [CrossRef] [PubMed]
  92. Montano, L.; Baldini, G.M.; Piscopo, M.; Liguori, G.; Lombardi, R.; Ricciardi, M.; Esposito, G.; Pinto, G.; Fontanarosa, C.; Spinelli, M.; et al. Polycyclic Aromatic Hydrocarbons (PAHs) in the Environment: Occupational Exposure, Health Risks and Fertility Implications. Toxics 2025, 13, 151. [Google Scholar] [CrossRef]
  93. Zhang, H.; Han, Y.; Qiu, X.; Wang, Y.; Li, W.; Liu, J.; Chen, X.; Li, R.; Xu, F.; Chen, W.; et al. Association of internal exposure to polycyclic aromatic hydrocarbons with inflammation and oxidative stress in prediabetic and healthy individuals. Chemosphere 2020, 253, 126748. [Google Scholar] [CrossRef]
  94. Ju, S.; Lim, L.; Ki, Y.-J.; Choi, D.-H.; Song, H. Oxidative stress generated by polycyclic aromatic hydrocarbons from ambient particulate matter enhance vascular smooth muscle cell migration through MMP upregulation and actin reorganization. Part. Fibre Toxicol. 2022, 19, 29. [Google Scholar] [CrossRef]
  95. Bowman, J.D.; Surani, S.; Horseman, M.A. Endotoxin, Toll-like Receptor-4, and Atherosclerotic Heart Disease. Curr. Cardiol. Rev. 2017, 13, 86–93. [Google Scholar] [CrossRef]
  96. Alissa, E.M.; Ferns, G.A. Heavy metal poisoning and cardiovascular disease. J. Toxicol. 2011, 2011, 870125. [Google Scholar] [CrossRef] [PubMed]
  97. Neal, A.P.; Guilarte, T.R. Mechanisms of lead and manganese neurotoxicity. Toxicol. Res. 2013, 2, 99–114. [Google Scholar] [CrossRef]
  98. Bakulski, K.M.; Seo, Y.A.; Hickman, R.C.; Brandt, D.; Vadari, H.S.; Hu, H.; Park, S.K. Heavy Metals Exposure and Alzheimer’s Disease and Related Dementias. J. Alzheimers. Dis. 2020, 76, 1215–1242. [Google Scholar] [CrossRef] [PubMed]
  99. Jager, T.; Ashauer, R. How to Evaluate the Quality of Toxicokinetic-Toxicodynamic Models in the Context of Environmental Risk Assessment. Integr. Environ. Assess. Manag. 2018, 14, 604–614. [Google Scholar] [CrossRef] [PubMed]
  100. Zhen, Z.; Yin, Y.; Chen, K.; Zhen, X.; Zhang, X.; Jiang, H.; Wang, H.; Kuang, X.; Cui, Y.; Dai, M.; et al. Concentration and atmospheric transport of PM2.5-bound polycyclic aromatic hydrocarbons at Mount Tai, China. Sci. Total Environ. 2021, 786, 147513. [Google Scholar] [CrossRef]
  101. Butler, J.D.; Crossley, P. Reactivity of polycyclic aromatic hydrocarbons adsorbed on soot particles. Atmos. Environ. (1967) 1981, 15, 91–94. [Google Scholar] [CrossRef]
  102. Turner, A.; Ip, K.-H. Bioaccessibility of metals in dust from the indoor environment: Application of a physiologically based extraction test. Environ. Sci. Technol. 2007, 41, 7851–7856. [Google Scholar] [CrossRef]
  103. Guo, Y.; Li, Y.; Zhu, T.; Wang, J.; Ye, M. Modeling of dioxin adsorption on activated carbon. Chem. Eng. J. 2016, 283, 1210–1215. [Google Scholar] [CrossRef]
  104. Islam, N.; Saikia, B.K. An overview on atmospheric carbonaceous particulate matter into carbon nanomaterials: A new approach for air pollution mitigation. Chemosphere 2022, 303, 135027. [Google Scholar] [CrossRef]
  105. Barsanti, K.C.; Pankow, J.F. Thermodynamics of the formation of atmospheric organic particulate matter by accretion reactions—Part 3: Carboxylic and dicarboxylic acids. Atmos. Environ. 2006, 40, 6676–6686. [Google Scholar] [CrossRef]
  106. Das, D.; Gaur, V.; Verma, N. Removal of volatile organic compound by activated carbon fiber. Carbon 2004, 42, 2949–2962. [Google Scholar] [CrossRef]
  107. Obeng, G.M.; Aram, S.A.; Agyei, D.; Saalidong, B.M. Exposure to particulate matter (PM2.5) and volatile organic compounds (VOCs), and self-reported health symptoms among fish smokers: A case study in the Western Region of Ghana. PLoS ONE 2023, 18, e0283438. [Google Scholar] [CrossRef] [PubMed]
  108. Shiru, G.A.; Auta, M.; Mustapha, S.A. Removal of Phosphate and Ammonium Ion from Domestic Wastewater Using Clay Feldspar in Wupa Sewage Treatment Plant Abuja. Univ. J. Green Chem. 2024, 2, 218–228. [Google Scholar] [CrossRef]
  109. Hammond, C.M.; Root, R.A.; Maier, R.M.; Chorover, J. Arsenic and iron speciation and mobilization during phytostabilization of pyritic mine tailings. Geochim. Cosmochim. Acta 2020, 286, 306–323. [Google Scholar] [CrossRef]
  110. Berro, Y.; Gueddida, S.; Lebègue, S.; Pasc, A.; Canilho, N.; Kassir, M.; Hassan, F.E.H.; Badawi, M. Atomistic description of phenol, CO and H2O adsorption over crystalline and amorphous silica surfaces for hydrodeoxygenation applications. Appl. Surf. Sci. 2019, 494, 721–730. [Google Scholar] [CrossRef]
  111. Zaady, E.; Sarig, S.; Katra, I. Dust Particles as a Pesticide’s Carrier in Agro-Ecosystems; Qualitative and Quantitative Analysis. Agronomy 2022, 12, 1826. [Google Scholar] [CrossRef]
  112. Malusá, E.; Tartanus, M.; Danelski, W.; Miszczak, A.; Szustakowska, E.; Kicińska, J.; Furmanczyk, E.M. Monitoring of DDT in Agricultural Soils under Organic Farming in Poland and the Risk of Crop Contamination. Environ. Manag. 2020, 66, 916–929. [Google Scholar] [CrossRef]
  113. Balasubramani, A.; Howell, N.L.; Rifai, H.S. Polychlorinated biphenyls (PCBs) in industrial and municipal effluents: Concentrations, congener profiles, and partitioning onto particulates and organic carbon. Sci. Total Environ. 2014, 473–474, 702–713. [Google Scholar] [CrossRef]
  114. Mataix, M.; Rodríguez-Luna, A.; Gutiérrez-Pérez, M.; Milani, M.; Gandarillas, A.; Espada, J.; Pérez-Davó, A. Deschampsia antarctica extract (Edafence®) as a powerful skin protection tool against the aging exposome. Plast. Aesthet. Res. 2020, 7, 69. [Google Scholar] [CrossRef]
  115. Barouki, R. Linking long-term toxicity of xeno-chemicals with short-term biological adaptation. Biochimie 2010, 92, 1222–1226. [Google Scholar] [CrossRef] [PubMed]
  116. Sharma, P.; Alakesh, A.; Jhunjhunwala, S. The consequences of particle uptake on immune cells. Trends Pharmacol. Sci. 2022, 43, 305–320. [Google Scholar] [CrossRef]
  117. Pagán, A.J.; Ramakrishnan, L. The Formation and Function of Granulomas. Annu. Rev. Immunol. 2018, 36, 639–665. [Google Scholar] [CrossRef] [PubMed]
  118. Woodard, B.H.; Rosenberg, S.I.; Farnham, R.; Adams, D.O. Incidence and nature of primary granulomatous inflammation in surgically removed material. Am. J. Surg. Pathol. 1982, 6, 119–129. [Google Scholar] [CrossRef] [PubMed]
  119. Pierik, M.; de Hertogh, G.; Vermeire, S.; van Assche, G.; van Eyken, P.; Joossens, S.; Claessens, G.; Vlietinck, R.; Rutgeerts, P.; Geboes, K. Epithelioid granulomas, pattern recognition receptors, and phenotypes of Crohn’s disease. Gut 2005, 54, 223–227. [Google Scholar] [CrossRef]
  120. Rosen, Y. Pathology of Granulomatous Pulmonary Diseases. Arch. Pathol. Lab. Med. 2022, 146, 233–251. [Google Scholar] [CrossRef]
  121. Nair, N.; Patrick, H.; Narula, J. Particulate matter granulomas masquerading as sarcoidosis: A diagnostic dilemma. Biomol. Concepts 2015, 6, 229–233. [Google Scholar] [CrossRef]
  122. Shah, K.K.; Pritt, B.S.; Alexander, M.P. Histopathologic review of granulomatous inflammation. J. Clin. Tuberc. Other Mycobact. Dis. 2017, 7, 1–12. [Google Scholar] [CrossRef]
  123. Nakamizo, S.; Kabashima, K. Cutaneous granulomas: Mechanisms, cellular interactions and therapeutic insights. Br. J. Dermatol. 2025, 192, 974–982. [Google Scholar] [CrossRef]
  124. Grammatikos, A.; Gennery, A.R. Inflammatory Complications in Chronic Granulomatous Disease. J. Clin. Med. 2024, 13, 1092. [Google Scholar] [CrossRef] [PubMed]
  125. Di Francesco, A.M.; Pasciuto, G.; Verrecchia, E.; Sicignano, L.L.; Gerardino, L.; Massaro, M.G.; Urbani, A.; Manna, R. Sarcoidosis and Cancer: The Role of the Granulomatous Reaction as a Double-Edged Sword. J. Clin. Med. 2024, 13, 5232. [Google Scholar] [CrossRef] [PubMed]
  126. Faurschou, M.; Omland, L.H.; Obel, N.; Lindhardsen, J.; Baslund, B. Risk of Cancer Among Sarcoidosis Patients With Biopsy-verified Nonnecrotizing Granulomatous Inflammation: Population-based Cohort Study. J. Rheumatol. 2022, 49, 186–191. [Google Scholar] [CrossRef] [PubMed]
  127. Xuan, L.; Ju, Z.; Skonieczna, M.; Zhou, P.-K.; Huang, R. Nanoparticles-induced potential toxicity on human health: Applications, toxicity mechanisms, and evaluation models. MedComm 2023, 4, e327. [Google Scholar] [CrossRef]
  128. Winiarska, E.; Jutel, M.; Zemelka-Wiacek, M. The potential impact of nano- and microplastics on human health: Understanding human health risks. Environ. Res. 2024, 251, 118535. [Google Scholar] [CrossRef]
  129. Krause, S.; Ouellet, V.; Allen, D.; Allen, S.; Moss, K.; Nel, H.A.; Manaseki-Holland, S.; Lynch, I. The potential of micro- and nanoplastics to exacerbate the health impacts and global burden of non-communicable diseases. Cell Rep. Med. 2024, 5, 101581. [Google Scholar] [CrossRef]
  130. Rafa, N.; Ahmed, B.; Zohora, F.; Bakya, J.; Ahmed, S.; Ahmed, S.F.; Mofijur, M.; Chowdhury, A.A.; Almomani, F. Microplastics as carriers of toxic pollutants: Source, transport, and toxicological effects. Environ. Pollut. 2024, 343, 123190. [Google Scholar] [CrossRef]
  131. Alijagic, A.; Suljević, D.; Fočak, M.; Sulejmanović, J.; Šehović, E.; Särndahl, E.; Engwall, M. The triple exposure nexus of microplastic particles, plastic-associated chemicals, and environmental pollutants from a human health perspective. Environ. Int. 2024, 188, 108736. [Google Scholar] [CrossRef]
  132. Vignesh, A.; Menaka, C.; Amal, T.C.; Selvakumar, S.; Vasanth, K. Exploring the dual impact of nanoparticles on human well-being: A comprehensive review of risks and benefits. Next Nanotechnol. 2025, 8, 100223. [Google Scholar] [CrossRef]
  133. Liu, Z.J.; Roy, A.; Zheng, Y.; Annabi, N. Harnessing carbon nanomaterials for reactive oxygen species regulation: Insights into generation, scavenging, and sensing. Adv. Drug Deliv. Rev. 2025, 224, 115651. [Google Scholar] [CrossRef]
  134. Juan, C.A.; La Pérez de Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef]
  135. Hu, J.; Zeng, Y.; Tang, L.; Ye, L.; Chen, C.; Ling, Q.; Wang, X.; He, L.; Chen, X.; Wang, Y.; et al. Topical Application of VitB6 Ameliorates PM2.5-Induced Dry Eye via NFκB Pathway in a Murine Model. Biomedicines 2025, 13, 541. [Google Scholar] [CrossRef]
  136. Herath, H.M.U.L.; Piao, M.J.; Kang, K.A.; Fernando, P.D.S.M.; Hyun, C.; Cho, S.J.; Hyun, J.W. The mitigation mechanism of morin on PM2.5-induced apoptosis by inhibiting mitochondrial damage via the ROS/ERK signaling pathway in human HaCaT keratinocytes. Int. J. Environ. Health Res. 2025, 1–16. [Google Scholar] [CrossRef]
  137. Lu, Y.; Qiu, W.; Liao, R.; Cao, W.; Huang, F.; Wang, X.; Li, M.; Li, Y. Subacute PM2.5 Exposure Induces Hepatic Insulin Resistance Through Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2025, 26, 812. [Google Scholar] [CrossRef] [PubMed]
  138. Li, X.; Ma, X.; Zhang, L.; Wu, W.; Zhou, R.; Li, S.; Liu, Y.; Tan, F.; Han, X.; Wang, Q.; et al. PM2.5 regulates TGF-β1/Smads-mediated pulmonary fibrosis via ROS/SnoN in vitro and in vivo. Int. Immunopharmacol. 2025, 153, 114477. [Google Scholar] [CrossRef] [PubMed]
  139. Cheng, P.-P.; He, X.-L.; Jia, Z.-H.; Hu, S.-H.; Feng, X.; Jiang, Y.-H.; Li, Q.; Zhao, L.-Q.; Cui, X.-L.; Ye, S.-Y.; et al. Midkine, a novel MCP-1 activator mediated PM2.5-aggravated experimental pulmonary fibrosis. Environ. Int. 2025, 197, 109354. [Google Scholar] [CrossRef]
  140. Zhao, C.; Pu, W.; Wazir, J.; Jin, X.; Wei, L.; Song, S.; Su, Z.; Li, J.; Deng, Y.; Wang, H. Long-term exposure to PM2.5 aggravates pulmonary fibrosis and acute lung injury by disrupting Nrf2-mediated antioxidant function. Environ. Pollut. 2022, 313, 120017. [Google Scholar] [CrossRef]
  141. Lelieveld, S.; Wilson, J.; Dovrou, E.; Mishra, A.; Lakey, P.S.J.; Shiraiwa, M.; Pöschl, U.; Berkemeier, T. Hydroxyl Radical Production by Air Pollutants in Epithelial Lining Fluid Governed by Interconversion and Scavenging of Reactive Oxygen Species. Environ. Sci. Technol. 2021, 55, 14069–14079. [Google Scholar] [CrossRef]
  142. Libalova, H.; Milcova, A.; Cervena, T.; Vrbova, K.; Rossnerova, A.; Novakova, Z.; Topinka, J.; Rossner, P. Kinetics of ROS generation induced by polycyclic aromatic hydrocarbons and organic extracts from ambient air particulate matter in model human lung cell lines. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2018, 827, 50–58. [Google Scholar] [CrossRef]
  143. Weber, S.A.; Insaf, T.Z.; Hall, E.S.; Talbot, T.O.; Huff, A.K. Assessing the impact of fine particulate matter (PM2.5) on respiratory-cardiovascular chronic diseases in the New York City Metropolitan area using Hierarchical Bayesian Model estimates. Environ. Res. 2016, 151, 399–409. [Google Scholar] [CrossRef]
  144. Liu, K.; Shi, M.; Li, X.; Zeng, X.; Liu, X. Curcumin Modulates the PTEN/PI3K/AKT Pathway to Alleviate Inflammation and Oxidative Stress in PM2.5-Induced Chronic Obstructive Pulmonary Disease. Food Chem. Toxicol. 2025, 201, 115460. [Google Scholar] [CrossRef]
  145. Shi, M.; Liu, K.; Li, X.; Zeng, X.-L.; Liu, X.-J. Melatonin ameliorates PM2.5-induced airway inflammation and apoptosis by PERK/eIF2α/ATF4/CHOP in chronic obstructive pulmonary disease mice. Toxicol. Appl. Pharmacol. 2025, 499, 117314. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, Y.; Zhang, J.; Zhang, T.; Wu, Y.; Xi, Y.; Wu, T.; Li, M.; Li, Y.; Zhou, S.; Wu, M.; et al. Circulating Interleukin-6 Mediates PM2.5-Induced Ovarian Injury by Suppressing the PPARγ Pathway. Research 2024, 7, 538. [Google Scholar] [CrossRef] [PubMed]
  147. Yuan, G.; Liu, Y.; Wang, Z.; Wang, X.; Han, Z.; Yan, X.; Meng, A. PM2.5 activated NLRP3 inflammasome and IL-1β release in MH-S cells by facilitating autophagy via activating Wnt5a. Int. J. Immunopathol. Pharmacol. 2022, 36, 3946320221137464. [Google Scholar] [CrossRef] [PubMed]
  148. Mozaffari, S.; Hassanvand, M.S.; Baeeri, M.; Gholami, M.; Bayrami, Z.; Yunesian, M.; Sahraian, M.A.; Nikfar, S.; Abdollahi, M. Exploring the impact of ambient air PM2.5 on multiple sclerosis: An experimental dive into neuroinflammation. Toxicol. Mech. Methods 2025, 35, 581–591. [Google Scholar] [CrossRef]
  149. Zeng, F.; Pang, G.; Hu, L.; Sun, Y.; Peng, W.; Chen, Y.; Xu, D.; Xia, Q.; Zhao, L.; Li, Y.; et al. Subway Fine Particles (PM2.5)-Induced Pro-Inflammatory Response Triggers Airway Epithelial Barrier Damage Through the TLRs/NF-κB-Dependent Pathway In Vitro. Environ. Toxicol. 2024, 39, 5296–5308. [Google Scholar] [CrossRef]
  150. Le, Y.; Hu, X.; Zhu, J.; Wang, C.; Yang, Z.; Lu, D. Ambient fine particulate matter induces inflammatory responses of vascular endothelial cells through activating TLR-mediated pathway. Toxicol. Ind. Health 2019, 35, 670–678. [Google Scholar] [CrossRef]
  151. Chen, P.; Ning, X.; Feng, W.; Li, Y.; Chen, G.; Shi, X.; Pan, Y.; Shi, X.; Xiao, Y.; Liu, Y.; et al. Chronic Exposure to Bioaerosols in PM2.5 from Garbage Stations Accelerates Vascular Aging via the NF-κB/NLRP3 Pathway. Adv. Sci. 2024, 11, e2404142. [Google Scholar] [CrossRef]
  152. Li, Y.; Sun, B.; Shi, Y.; Jiang, J.; Du, Z.; Chen, R.; Duan, J.; Sun, Z. Subacute exposure of PM2.5 induces airway inflammation through inflammatory cell infiltration and cytokine expression in rats. Chemosphere 2020, 251, 126423. [Google Scholar] [CrossRef]
  153. Sun, N.; Shi, H.; Li, X.; Gao, C.; Liu, R. Combined toxicity of micro/nanoplastics loaded with environmental pollutants to organisms and cells: Role, effects, and mechanism. Environ. Int. 2023, 171, 107711. [Google Scholar] [CrossRef]
  154. Lagunas-Rangel, F.A.; Linnea-Niemi, J.V.; Kudłak, B.; Williams, M.J.; Jönsson, J.; Schiöth, H.B. Role of the Synergistic Interactions of Environmental Pollutants in the Development of Cancer. Geohealth 2022, 6, e2021GH000552. [Google Scholar] [CrossRef] [PubMed]
  155. Grytting, V.S.; Chand, P.; Låg, M.; Øvrevik, J.; Refsnes, M. The pro-inflammatory effects of combined exposure to diesel exhaust particles and mineral particles in human bronchial epithelial cells. Part. Fibre Toxicol. 2022, 19, 14. [Google Scholar] [CrossRef] [PubMed]
  156. Schraufnagel, D.E.; Balmes, J.R.; Cowl, C.T.; de Matteis, S.; Jung, S.-H.; Mortimer, K.; Perez-Padilla, R.; Rice, M.B.; Riojas-Rodriguez, H.; Sood, A.; et al. Air Pollution and Noncommunicable Diseases: A Review by the Forum of International Respiratory Societies’ Environmental Committee, Part 1: The Damaging Effects of Air Pollution. Chest 2019, 155, 409–416. [Google Scholar] [CrossRef] [PubMed]
  157. Nagappan, A.; Park, S.B.; Lee, S.-J.; Moon, Y. Mechanistic Implications of Biomass-Derived Particulate Matter for Immunity and Immune Disorders. Toxics 2021, 9, 18. [Google Scholar] [CrossRef]
  158. Holme, J.A.; Låg, M.; Skuland, T.; Parenicová, M.; Ciganek, M.; Penciková, K.; Grytting, V.S.; Neca, J.; Øvrevik, J.; Mariussen, E.; et al. Characterization of elements, PAHs, AhR-activity and pro-inflammatory responses of road tunnel-derived particulate matter in human hepatocyte-like and bronchial epithelial cells. Toxicol. In Vitr. 2023, 90, 105611. [Google Scholar] [CrossRef]
  159. Balali-Mood, M.; Naseri, K.; Tahergorabi, Z.; Khazdair, M.R.; Sadeghi, M. Toxic Mechanisms of Five Heavy Metals: Mercury, Lead, Chromium, Cadmium, and Arsenic. Front. Pharmacol. 2021, 12, 643972. [Google Scholar] [CrossRef]
  160. Moretti, J.; Blander, J.M. Insights into phagocytosis-coupled activation of pattern recognition receptors and inflammasomes. Curr. Opin. Immunol. 2014, 26, 100–110. [Google Scholar] [CrossRef]
  161. Park, S.-H.; Yoon, S.-J.; Choi, S.; Jung, J.; Park, J.-Y.; Park, Y.-H.; Seo, J.; Lee, J.; Lee, M.-S.; Lee, S.-J.; et al. Particulate matter promotes cancer metastasis through increased HBEGF expression in macrophages. Exp. Mol. Med. 2022, 54, 1901–1912. [Google Scholar] [CrossRef]
  162. Valderrama, A.; Zapata, M.I.; Hernandez, J.C.; Cardona-Arias, J.A. Systematic review of preclinical studies on the neutrophil-mediated immune response to air pollutants, 1980–2020. Heliyon 2022, 8, e08778. [Google Scholar] [CrossRef]
  163. Taylor-Blair, H.C.; Siu, A.C.W.; Haysom-McDowell, A.; Kokkinis, S.; Bani Saeid, A.; Chellappan, D.K.; Oliver, B.G.G.; Paudel, K.R.; de Rubis, G.; Dua, K. The impact of airborne particulate matter-based pollution on the cellular and molecular mechanisms in chronic obstructive pulmonary disease (COPD). Sci. Total Environ. 2024, 954, 176413. [Google Scholar] [CrossRef]
  164. Matthews, N.C.; Pfeffer, P.E.; Mann, E.H.; Kelly, F.J.; Corrigan, C.J.; Hawrylowicz, C.M.; Lee, T.H. Urban Particulate Matter-Activated Human Dendritic Cells Induce the Expansion of Potent Inflammatory Th1, Th2, and Th17 Effector Cells. Am. J. Respir. Cell Mol. Biol. 2016, 54, 250–262. [Google Scholar] [CrossRef]
  165. Pfeffer, P.E.; Ho, T.R.; Mann, E.H.; Kelly, F.J.; Sehlstedt, M.; Pourazar, J.; Dove, R.E.; Sandstrom, T.; Mudway, I.S.; Hawrylowicz, C.M. Urban particulate matter stimulation of human dendritic cells enhances priming of naive CD8 T lymphocytes. Immunology 2018, 153, 502–512. [Google Scholar] [CrossRef]
  166. Gałuszka, A.; Stec, M.; Węglarczyk, K.; Kluczewska, A.; Siedlar, M.; Baran, J. Transition Metal Containing Particulate Matter Promotes Th1 and Th17 Inflammatory Response by Monocyte Activation in Organic and Inorganic Compounds Dependent Manner. Int. J. Environ. Res. Public Health 2020, 17, 1227. [Google Scholar] [CrossRef]
  167. Zambrano-Zaragoza, J.F.; Romo-Martínez, E.J.; Durán-Avelar, M.d.J.; García-Magallanes, N.; Vibanco-Pérez, N. Th17 cells in autoimmune and infectious diseases. Int. J. Inflam. 2014, 2014, 651503. [Google Scholar] [CrossRef] [PubMed]
  168. Hafkamp, F.M.J.; Groot Kormelink, T.; de Jong, E.C. Targeting DCs for Tolerance Induction: Don’t Lose Sight of the Neutrophils. Front. Immunol. 2021, 12, 732992. [Google Scholar] [CrossRef] [PubMed]
  169. Mpakosi, A.; Cholevas, V.; Tzouvelekis, I.; Passos, I.; Kaliouli-Antonopoulou, C.; Mironidou-Tzouveleki, M. Autoimmune Diseases Following Environmental Disasters: A Narrative Review of the Literature. Healthcare 2024, 12, 1767. [Google Scholar] [CrossRef] [PubMed]
  170. Müller, L.; Chehrazi, C.V.E.; Henderson, M.W.; Noah, T.L.; Jaspers, I. Diesel exhaust particles modify natural killer cell function and cytokine release. Part. Fibre Toxicol. 2013, 10, 16. [Google Scholar] [CrossRef]
  171. Tuazon, J.A.; Kilburg-Basnyat, B.; Oldfield, L.M.; Wiscovitch-Russo, R.; Dunigan-Russell, K.; Fedulov, A.V.; Oestreich, K.J.; Gowdy, K.M. Emerging Insights into the Impact of Air Pollution on Immune-Mediated Asthma Pathogenesis. Curr. Allergy Asthma Rep. 2022, 22, 77–92. [Google Scholar] [CrossRef]
  172. Kayalar, Ö.; Rajabi, H.; Konyalilar, N.; Mortazavi, D.; Aksoy, G.T.; Wang, J.; Bayram, H. Impact of particulate air pollution on airway injury and epithelial plasticity; underlying mechanisms. Front. Immunol. 2024, 15, 1324552. [Google Scholar] [CrossRef]
  173. Ma, J.; Chiu, Y.-F.; Kao, C.-C.; Chuang, C.-N.; Chen, C.-Y.; Lai, C.-H.; Kuo, M.-L. Fine particulate matter manipulates immune response to exacerbate microbial pathogenesis in the respiratory tract. Eur. Respir. Rev. 2024, 33, 230259. [Google Scholar] [CrossRef]
  174. Genc, S.; Zadeoglulari, Z.; Fuss, S.H.; Genc, K. The adverse effects of air pollution on the nervous system. J. Toxicol. 2012, 2012, 782462. [Google Scholar] [CrossRef] [PubMed]
  175. Lee, J.; Weerasinghe-Mudiyanselage, P.D.E.; Kim, B.; Kang, S.; Kim, J.-S.; Moon, C. Particulate matter exposure and neurodegenerative diseases: A comprehensive update on toxicity and mechanisms. Ecotoxicol. Environ. Saf. 2023, 266, 115565. [Google Scholar] [CrossRef] [PubMed]
  176. Becker, S.; Mundandhara, S.; Devlin, R.B.; Madden, M. Regulation of cytokine production in human alveolar macrophages and airway epithelial cells in response to ambient air pollution particles: Further mechanistic studies. Toxicol. Appl. Pharmacol. 2005, 207, 269–275. [Google Scholar] [CrossRef]
  177. Farina, F.; Sancini, G.; Mantecca, P.; Gallinotti, D.; Camatini, M.; Palestini, P. The acute toxic effects of particulate matter in mouse lung are related to size and season of collection. Toxicol. Lett. 2011, 202, 209–217. [Google Scholar] [CrossRef]
  178. Pope, C.A.; Ezzati, M.; Dockery, D.W. Fine-particulate air pollution and life expectancy in the United States. N. Engl. J. Med. 2009, 360, 376–386. [Google Scholar] [CrossRef]
  179. Künzli, N.; Kaiser, R.; Medina, S.; Studnicka, M.; Chanel, O.; Filliger, P.; Herry, M.; Horak, F.; Puybonnieux-Texier, V.; Quénel, P.; et al. Public-health impact of outdoor and traffic-related air pollution: A European assessment. Lancet 2000, 356, 795–801. [Google Scholar] [CrossRef]
  180. Schins, R.P.F.; Knaapen, A.M. Genotoxicity of poorly soluble particles. Inhal. Toxicol. 2007, 19 (Suppl. S1), 189–198. [Google Scholar] [CrossRef]
  181. Weng, C.-M.; Wang, C.-H.; Lee, M.-J.; He, J.-R.; Huang, H.-Y.; Chao, M.-W.; Chung, K.F.; Kuo, H.-P. Aryl hydrocarbon receptor activation by diesel exhaust particles mediates epithelium-derived cytokines expression in severe allergic asthma. Allergy 2018, 73, 2192–2204. [Google Scholar] [CrossRef]
  182. de Homdedeu, M.; Cruz, M.; Sanchez-Díez, S.; Ojanguren, I.; Romero-Mesones, C.; Vanoirbeek, J.; Velde, G.V.; Muñoz, X. The immunomodulatory effects of diesel exhaust particles in asthma. Environ. Pollut. 2020, 263, 114600. [Google Scholar] [CrossRef]
  183. Takano, H.; Yoshikawa, T.; Ichinose, T.; Miyabara, Y.; Imaoka, K.; Sagai, M. Diesel exhaust particles enhance antigen-induced airway inflammation and local cytokine expression in mice. Am. J. Respir. Crit. Care Med. 1997, 156, 36–42. [Google Scholar] [CrossRef]
  184. Diaz-Sanchez, D.; Dotson, A.R.; Takenaka, H.; Saxon, A. Diesel exhaust particles induce local IgE production in vivo and alter the pattern of IgE messenger RNA isoforms. J. Clin. Investig. 1994, 94, 1417–1425. [Google Scholar] [CrossRef]
  185. Li, N.; Sioutas, C.; Cho, A.; Schmitz, D.; Misra, C.; Sempf, J.; Wang, M.; Oberley, T.; Froines, J.; Nel, A. Ultrafine particulate pollutants induce oxidative stress and mitochondrial damage. Environ. Health Perspect. 2003, 111, 455–460. [Google Scholar] [CrossRef] [PubMed]
  186. Nadeau, K.; McDonald-Hyman, C.; Noth, E.M.; Pratt, B.; Hammond, S.K.; Balmes, J.; Tager, I. Ambient air pollution impairs regulatory T-cell function in asthma. J. Allergy Clin. Immunol. 2010, 126, 845–852.e10. [Google Scholar] [CrossRef] [PubMed]
  187. Hiraiwa, K.; van Eeden, S.F. Contribution of lung macrophages to the inflammatory responses induced by exposure to air pollutants. Mediat. Inflamm. 2013, 2013, 619523. [Google Scholar] [CrossRef] [PubMed]
  188. Chen, Y.-W.; Huang, M.-Z.; Chen, C.-L.; Kuo, C.-Y.; Yang, C.-Y.; Chiang-Ni, C.; Chen, Y.-Y.M.; Hsieh, C.-M.; Wu, H.-Y.; Kuo, M.-L.; et al. PM2.5 impairs macrophage functions to exacerbate pneumococcus-induced pulmonary pathogenesis. Part. Fibre Toxicol. 2020, 17, 37. [Google Scholar] [CrossRef]
  189. Zhao, H.; Li, W.; Gao, Y.; Li, J.; Wang, H. Exposure to particular matter increases susceptibility to respiratory Staphylococcus aureus infection in rats via reducing pulmonary natural killer cells. Toxicology 2014, 325, 180–188. [Google Scholar] [CrossRef]
  190. Arias-Pérez, R.D.; Taborda, N.A.; Gómez, D.M.; Narvaez, J.F.; Porras, J.; Hernandez, J.C. Inflammatory effects of particulate matter air pollution. Environ. Sci. Pollut. Res. Int. 2020, 27, 42390–42404. [Google Scholar] [CrossRef]
  191. Glencross, D.A.; Ho, T.-R.; Camiña, N.; Hawrylowicz, C.M.; Pfeffer, P.E. Air pollution and its effects on the immune system. Free Radic. Biol. Med. 2020, 151, 56–68. [Google Scholar] [CrossRef]
  192. Baccarelli, A.; Zanobetti, A.; Martinelli, I.; Grillo, P.; Hou, L.; Lanzani, G.; Mannucci, P.M.; Bertazzi, P.A.; Schwartz, J. Air pollution, smoking, and plasma homocysteine. Environ. Health Perspect. 2007, 115, 176–181. [Google Scholar] [CrossRef]
  193. Oberdörster, G.; Sharp, Z.; Atudorei, V.; Elder, A.; Gelein, R.; Kreyling, W.; Cox, C. Translocation of inhaled ultrafine particles to the brain. Inhal. Toxicol. 2004, 16, 437–445. [Google Scholar] [CrossRef]
  194. Braakhuis, H.M.; Park, M.V.D.Z.; Gosens, I.; de Jong, W.H.; Cassee, F.R. Physicochemical characteristics of nanomaterials that affect pulmonary inflammation. Part. Fibre Toxicol. 2014, 11, 18. [Google Scholar] [CrossRef] [PubMed]
  195. Thomas, R.J. Particle size and pathogenicity in the respiratory tract. Virulence 2013, 4, 847–858. [Google Scholar] [CrossRef] [PubMed]
  196. Wen, J.; Yang, Y.; Zhang, H.; Wu, W.; Dai, Z.; Liang, X.; Chen, S. PM2.5 increases the risk of early-onset COPD mediated by smoking and shared genes: A large-scale genetic analysis. Clin. Exp. Med. 2025, 25, 116. [Google Scholar] [CrossRef] [PubMed]
  197. Zhao, L.; Ding, X.; Zhou, L.; Song, C.; Kang, T.; Xu, Y.; Liu, Y.; Han, Y.; Zhao, W.; Zhang, B.; et al. Effect of PM2.5 exposure on susceptibility to allergic asthma in elderly rats treated with allergens. Sci. Rep. 2025, 15, 5594. [Google Scholar] [CrossRef]
  198. Wallis, R.S.; Broder, M.S.; Wong, J.Y.; Hanson, M.E.; Beenhouwer, D.O. Granulomatous infectious diseases associated with tumor necrosis factor antagonists. Clin. Infect. Dis. 2004, 38, 1261–1265. [Google Scholar] [CrossRef]
  199. Fallahi-Sichani, M.; El-Kebir, M.; Marino, S.; Kirschner, D.E.; Linderman, J.J. Multiscale computational modeling reveals a critical role for TNF-α receptor 1 dynamics in tuberculosis granuloma formation. J. Immunol. 2011, 186, 3472–3483. [Google Scholar] [CrossRef]
  200. Mohan, V.P.; Scanga, C.A.; Yu, K.; Scott, H.M.; Tanaka, K.E.; Tsang, E.; Tsai, M.M.; Flynn, J.L.; Chan, J. Effects of tumor necrosis factor alpha on host immune response in chronic persistent tuberculosis: Possible role for limiting pathology. Infect. Immun. 2001, 69, 1847–1855. [Google Scholar] [CrossRef]
  201. Im Kim, D.; Song, M.-K.; Yuk, J.E.; Seo, H.J.; Lee, K. Establishment of an artificial particulate matter-induced lung disease model through analyzing pathological changes and transcriptomic profiles in mice. Sci. Rep. 2023, 13, 5955. [Google Scholar] [CrossRef]
  202. Makrufardi, F.; Bai, K.-J.; Suk, C.-W.; Rusmawatiningtyas, D.; Chung, K.F.; Chuang, H.-C. Alveolar deposition of inhaled fine particulate matter increased risk of severity of pulmonary tuberculosis in the upper and middle lobes. ERJ Open Res. 2023, 9, 00064-2023. [Google Scholar] [CrossRef]
  203. Pan, Y.; Mei, J.; Jiang, J.; Xu, K.; Gao, X.; Jiang, S.; Liu, Y. PFAS in PMs might be the escalating hazard to the lung health. Nano Res. 2023, 16, 13113–13133. [Google Scholar] [CrossRef]
  204. Koyama, H.; Kamogashira, T.; Yamasoba, T. Heavy Metal Exposure: Molecular Pathways, Clinical Implications, and Protective Strategies. Antioxidants 2024, 13, 76. [Google Scholar] [CrossRef]
  205. Pal, S.; Firdous, S.M. Unraveling the role of heavy metals xenobiotics in cancer: A critical review. Discov. Oncol. 2024, 15, 615. [Google Scholar] [CrossRef] [PubMed]
  206. Liu, J.; Chen, X.; Qiu, X.; Zhang, H.; Lu, X.; Li, H.; Chen, W.; Zhang, L.; Que, C.; Zhu, T. Association between exposure to polycyclic aromatic hydrocarbons and lipid peroxidation in patients with chronic obstructive pulmonary disease. Sci. Total Environ. 2021, 780, 146660. [Google Scholar] [CrossRef] [PubMed]
  207. Ma, Y.; Huang, Y.; Li, L.; Yu, L.; Xiao, P.; Wang, Q. Time trends in coronary heart disease mortality attributed to outdoor PM2.5 in China: An age-period-cohort analysis using the Global Burden of Disease Study 2019. Front. Public Health 2025, 13, 1517507. [Google Scholar] [CrossRef]
  208. Zhang, Z.; An, R.; Guo, H.; Yang, X. Effects of PM2.5 exposure and air temperature on risk of cardiovascular disease: Evidence from a prospective cohort study. Front. Public Health 2024, 12, 1487034. [Google Scholar] [CrossRef]
  209. Pota, P.; Suwannasom, P.; Chattipakorn, S.C.; Chattipakorn, N. From smog to scarred hearts: Unmasking the detrimental impact of air pollution on myocardial ischemia-reperfusion injury. Cell. Mol. Life Sci. 2025, 82, 65. [Google Scholar] [CrossRef]
  210. Miller, M.R. The cardiovascular effects of air pollution: Prevention and reversal by pharmacological agents. Pharmacol. Ther. 2022, 232, 107996. [Google Scholar] [CrossRef]
  211. Song, B.; Bie, Y.; Feng, H.; Xie, B.; Liu, M.; Zhao, F. Inflammatory Factors Driving Atherosclerotic Plaque Progression New Insights. J. Transl. Int. Med. 2022, 10, 36–47. [Google Scholar] [CrossRef]
  212. Bezsonov, E.; Khotina, V.; Glanz, V.; Sobenin, I.; Orekhov, A. Lipids and Lipoproteins in Atherosclerosis. Biomedicines 2023, 11, 1424. [Google Scholar] [CrossRef]
  213. Afshar-Kharghan, V.; Thiagarajan, P. Leukocyte adhesion and thrombosis. Curr. Opin. Hematol. 2006, 13, 34–39. [Google Scholar] [CrossRef]
  214. Chanda, F.; Lin, K.-X.; Chaurembo, A.I.; Huang, J.-Y.; Zhang, H.-J.; Deng, W.-H.; Xu, Y.-J.; Li, Y.; Fu, L.-D.; Cui, H.-D.; et al. PM2.5-mediated cardiovascular disease in aging: Cardiometabolic risks, molecular mechanisms and potential interventions. Sci. Total Environ. 2024, 954, 176255. [Google Scholar] [CrossRef]
  215. Wróblewski, M.; Miłek, J.; Godlewski, A.; Wróblewska, J. The Impact of Arsenic, Cadmium, Lead, Mercury, and Thallium Exposure on the Cardiovascular System and Oxidative Mechanisms in Children. Curr. Issues Mol. Biol. 2025, 47, 483. [Google Scholar] [CrossRef] [PubMed]
  216. Qu, F.; Zheng, W. Cadmium Exposure: Mechanisms and Pathways of Toxicity and Implications for Human Health. Toxics 2024, 12, 388. [Google Scholar] [CrossRef] [PubMed]
  217. Kumar, S.V.; Bhattacharya, S. In vitro toxicity of mercury, cadmium, and arsenic to platelet aggregation: Influence of adenylate cyclase and phosphodiesterase activity. In Vitr. Mol. Toxicol. 2000, 13, 137–144. [Google Scholar] [CrossRef] [PubMed]
  218. Kumar, S.V.; Bose, R.; Bhattacharya, S. Low doses of heavy metals disrupt normal structure and function of rat platelets. J. Environ. Pathol. Toxicol. Oncol. 2001, 20, 65–75. [Google Scholar] [CrossRef]
  219. Solenkova, N.V.; Newman, J.D.; Berger, J.S.; Thurston, G.; Hochman, J.S.; Lamas, G.A. Metal pollutants and cardiovascular disease: Mechanisms and consequences of exposure. Am. Heart J. 2014, 168, 812–822. [Google Scholar] [CrossRef]
  220. Liu, Y.; Wang, L.; Wang, F.; Li, C. Effect of Fine Particulate Matter (PM2.5) on Rat Placenta Pathology and Perinatal Outcomes. Med. Sci. Monit. 2016, 22, 3274–3280. [Google Scholar] [CrossRef]
  221. Nagiah, S.; Phulukdaree, A.; Naidoo, D.; Ramcharan, K.; Naidoo, R.N.; Moodley, D.; Chuturgoon, A. Oxidative stress and air pollution exposure during pregnancy: A molecular assessment. Hum. Exp. Toxicol. 2015, 34, 838–847. [Google Scholar] [CrossRef]
  222. Vornic, I.; Buciu, V.; Furau, C.G.; Gaje, P.N.; Ceausu, R.A.; Dumitru, C.-S.; Barb, A.C.; Novacescu, D.; Cumpanas, A.A.; Latcu, S.C.; et al. Oxidative Stress and Placental Pathogenesis: A Contemporary Overview of Potential Biomarkers and Emerging Therapeutics. Int. J. Mol. Sci. 2024, 25, 12195. [Google Scholar] [CrossRef]
  223. Duan, S.; Zheng, Y.; Tian, J.; Zhang, L. Single-cell RNA sequencing of estrual mice reveals PM2.5-induced uterine cell heterogeneity and reproductive toxicity. Ecotoxicol. Environ. Saf. 2024, 284, 116968. [Google Scholar] [CrossRef]
  224. Zhang, X.; Wang, J.; Wu, Y.; Li, X.; Zheng, D.; Sun, L. Personal exposure to polycyclic aromatic hydrocarbons-bound particulate matter during pregnancy and umbilical inflammation and oxidative stress. Ecotoxicol. Environ. Saf. 2025, 291, 117896. [Google Scholar] [CrossRef]
  225. Techapichetvanich, P.; Sillapaprayoon, S.; Vivithanaporn, P.; Pimtong, W.; Khemawoot, P. Assessing developmental and transcriptional effects of PM2.5 on zebrafish embryos. Toxicol. Rep. 2024, 12, 397–403. [Google Scholar] [CrossRef] [PubMed]
  226. Li, Y.; Xie, T.; Cardoso Melo, R.D.; de Vries, M.; Lakerveld, J.; Zijlema, W.; Hartman, C.A. Longitudinal effects of environmental noise and air pollution exposure on autism spectrum disorder and attention-deficit/hyperactivity disorder during adolescence and early adulthood: The TRAILS study. Environ. Res. 2023, 227, 115704. [Google Scholar] [CrossRef] [PubMed]
  227. Pagalan, L.; Bickford, C.; Weikum, W.; Lanphear, B.; Brauer, M.; Lanphear, N.; Hanley, G.E.; Oberlander, T.F.; Winters, M. Association of Prenatal Exposure to Air Pollution With Autism Spectrum Disorder. JAMA Pediatr. 2019, 173, 86–92. [Google Scholar] [CrossRef] [PubMed]
  228. Santos, J.X.; Sampaio, P.; Rasga, C.; Martiniano, H.; Faria, C.; Café, C.; Oliveira, A.; Duque, F.; Oliveira, G.; Sousa, L.; et al. Evidence for an association of prenatal exposure to particulate matter with clinical severity of Autism Spectrum Disorder. Environ. Res. 2023, 228, 115795. [Google Scholar] [CrossRef]
  229. Amnuaylojaroen, T.; Parasin, N.; Saokaew, S. Exploring the association between early-life air pollution exposure and autism spectrum disorders in children: A systematic review and meta-analysis. Reprod. Toxicol. 2024, 125, 108582. [Google Scholar] [CrossRef]
  230. Roy, R.; D’Angiulli, A. Air pollution and neurological diseases, current state highlights. Front. Neurosci. 2024, 18, 1351721. [Google Scholar] [CrossRef]
  231. Du, X.; Guan, L.; Chen, C.; Wang, X.; Geng, X. Long-term exposure to PM2.5 exacerbates dopaminergic neuronal loss through CpG hypermethylation induced down-regulation of PINK1 and DJ-1 genes. Sci. Rep. 2025, 15, 10778. [Google Scholar] [CrossRef]
  232. Calderón-Garcidueñas, L.; Stommel, E.W.; Torres-Jardón, R.; Hernández-Luna, J.; Aiello-Mora, M.; González-Maciel, A.; Reynoso-Robles, R.; Pérez-Guillé, B.; Silva-Pereyra, H.G.; Tehuacanero-Cuapa, S.; et al. Alzheimer and Parkinson diseases, frontotemporal lobar degeneration and amyotrophic lateral sclerosis overlapping neuropathology start in the first two decades of life in pollution exposed urbanites and brain ultrafine particulate matter and industrial nanoparticles, including Fe, Ti, Al, V, Ni, Hg, Co, Cu, Zn, Ag, Pt, Ce, La, Pr and W are key players. Metropolitan Mexico City health crisis is in progress. Front. Hum. Neurosci. 2023, 17, 1297467. [Google Scholar] [CrossRef]
  233. Han, Y.; Yu, Z.; Chen, Y.; Guo, X.; Liu, Y.; Zhang, H.; Li, Z.; Chen, L. PM2.5 induces developmental neurotoxicity in cortical organoids. Environ. Pollut. 2024, 361, 124913. [Google Scholar] [CrossRef]
  234. Kim, M.E.; Lee, J.S. Mechanisms and Emerging Regulators of Neuroinflammation: Exploring New Therapeutic Strategies for Neurological Disorders. Curr. Issues Mol. Biol. 2024, 47, 8. [Google Scholar] [CrossRef]
  235. Lepeta, K.; Lourenco, M.V.; Schweitzer, B.C.; Martino Adami, P.V.; Banerjee, P.; Catuara-Solarz, S.; de La Fuente Revenga, M.; Guillem, A.M.; Haidar, M.; Ijomone, O.M.; et al. Synaptopathies: Synaptic dysfunction in neurological disorders-A review from students to students. J. Neurochem. 2016, 138, 785–805. [Google Scholar] [CrossRef]
  236. Devanand, D.P.; Sano, M.; Tang, M.X.; Taylor, S.; Gurland, B.J.; Wilder, D.; Stern, Y.; Mayeux, R. Depressed mood and the incidence of Alzheimer’s disease in the elderly living in the community. Arch. Gen. Psychiatry 1996, 53, 175–182. [Google Scholar] [CrossRef]
  237. Reed, M.D.; Gigliotti, A.P.; McDonald, J.D.; Seagrave, J.C.; Seilkop, S.K.; Mauderly, J.L. Health effects of subchronic exposure to environmental levels of diesel exhaust. Inhal. Toxicol. 2004, 16, 177–193. [Google Scholar] [CrossRef]
  238. Lee, S.-H.; Chen, Y.-H.; Chien, C.-C.; Yan, Y.-H.; Chen, H.-C.; Chuang, H.-C.; Hsieh, H.-I.; Cho, K.-H.; Kuo, L.-W.; Chou, C.C.-K.; et al. Three month inhalation exposure to low-level PM2.5 induced brain toxicity in an Alzheimer’s disease mouse model. PLoS ONE 2021, 16, e0254587. [Google Scholar] [CrossRef] [PubMed]
  239. Calderón-Garcidueñas, L.; Solt, A.C.; Henríquez-Roldán, C.; Torres-Jardón, R.; Nuse, B.; Herritt, L.; Villarreal-Calderón, R.; Osnaya, N.; Stone, I.; García, R.; et al. Long-term air pollution exposure is associated with neuroinflammation, an altered innate immune response, disruption of the blood-brain barrier, ultrafine particulate deposition, and accumulation of amyloid beta-42 and alpha-synuclein in children and young adults. Toxicol. Pathol. 2008, 36, 289–310. [Google Scholar] [CrossRef] [PubMed]
  240. Levesque, S.; Surace, M.J.; McDonald, J.; Block, M.L. Air pollution & the brain: Subchronic diesel exhaust exposure causes neuroinflammation and elevates early markers of neurodegenerative disease. J. Neuroinflamm. 2011, 8, 105. [Google Scholar] [CrossRef]
  241. Yan, Y.; Guo, F.; Liu, K.; Ding, R.; Wang, Y. The effect of endocrine-disrupting chemicals on placental development. Front. Endocrinol. 2023, 14, 1059854. [Google Scholar] [CrossRef]
  242. Silva-Adaya, D.; Garza-Lombó, C.; Gonsebatt, M.E. Xenobiotic transport and metabolism in the human brain. Neurotoxicology 2021, 86, 125–138. [Google Scholar] [CrossRef]
  243. Kim, A.C.; Lim, S.; Kim, Y.K. Metal Ion Effects on Aβ and Tau Aggregation. Int. J. Mol. Sci. 2018, 19, 128. [Google Scholar] [CrossRef]
  244. Murata, H.; Barnhill, L.M.; Bronstein, J.M. Air Pollution and the Risk of Parkinson’s Disease: A Review. Mov. Disord. 2022, 37, 894–904. [Google Scholar] [CrossRef] [PubMed]
  245. Olasehinde, T.A.; Olaniran, A.O. Neurotoxicity of Polycyclic Aromatic Hydrocarbons: A Systematic Mapping and Review of Neuropathological Mechanisms. Toxics 2022, 10, 417. [Google Scholar] [CrossRef] [PubMed]
  246. Pino, J.S.; Alvarado, P.N.; Larrea, A.M.; Rojas, W.; Gomez-Lopera, N. Analysis of cytotoxicity and genotoxicity of diesel exhaust PM2.5 generated from diesel and dual natural gas-diesel engines. Environ. Toxicol. Pharmacol. 2025, 114, 104638. [Google Scholar] [CrossRef] [PubMed]
  247. Mu, Q.; Jiang, G.; Chen, L.; Zhou, H.; Fourches, D.; Tropsha, A.; Yan, B. Chemical basis of interactions between engineered nanoparticles and biological systems. Chem. Rev. 2014, 114, 7740–7781. [Google Scholar] [CrossRef]
  248. Arif, I.; Adams, M.D.; Johnson, M.T.J. A meta-analysis of the carcinogenic effects of particulate matter and polycyclic aromatic hydrocarbons. Environ. Pollut. 2024, 351, 123941. [Google Scholar] [CrossRef]
  249. Abbas, I.; Badran, G.; Verdin, A.; Ledoux, F.; Roumie, M.; Lo Guidice, J.-M.; Courcot, D.; Garçon, G. In vitro evaluation of organic extractable matter from ambient PM2.5 using human bronchial epithelial BEAS-2B cells: Cytotoxicity, oxidative stress, pro-inflammatory response, genotoxicity, and cell cycle deregulation. Environ. Res. 2019, 171, 510–522. [Google Scholar] [CrossRef]
  250. Zhang, Y.; Yang, D.; Yang, B.; Li, B.; Guo, J.; Xiao, C. PM2.5 induces cell cycle arrest through regulating mTOR/P70S6K1 signaling pathway. Exp. Ther. Med. 2019, 17, 4371–4378. [Google Scholar] [CrossRef]
  251. Wang, Y.; Wen, Y.; Chen, Q.; Huang, Y.; Zhou, D.; Yang, W.; Yang, L.; Xiong, J.; Gao, K.; Sun, L.; et al. Downregulation of tRNA methyltransferase FTSJ1 by PM2.5 promotes glycolysis and malignancy of NSCLC via facilitating PGK1 expression and translation. Cell Death Dis. 2024, 15, 911. [Google Scholar] [CrossRef]
  252. Zeng, Y.; Zhang, R.; Jiang, Y.; Li, D.; Chen, L.; Dong, G.; Zhang, R.; Niu, Y.; Chen, W.; Chen, S. Interactions between fibroblasts and monocyte-derived cells in chronic lung injuries induced by real-ambient particulate matter exposure. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2024, 899, 503807. [Google Scholar] [CrossRef]
  253. Cui, A.; Xiang, M.; Xu, M.; Lu, P.; Wang, S.; Zou, Y.; Qiao, K.; Jin, C.; Li, Y.; Lu, M.; et al. VCAM-1-mediated neutrophil infiltration exacerbates ambient fine particle-induced lung injury. Toxicol. Lett. 2019, 302, 60–74. [Google Scholar] [CrossRef]
  254. Jan, A.T.; Azam, M.; Siddiqui, K.; Ali, A.; Choi, I.; Haq, Q.M.R. Heavy Metals and Human Health: Mechanistic Insight into Toxicity and Counter Defense System of Antioxidants. Int. J. Mol. Sci. 2015, 16, 29592–29630. [Google Scholar] [CrossRef]
  255. An, X.; Yu, W.; Liu, J.; Tang, D.; Yang, L.; Chen, X. Oxidative cell death in cancer: Mechanisms and therapeutic opportunities. Cell Death Dis. 2024, 15, 556. [Google Scholar] [CrossRef] [PubMed]
  256. Bhattacharjee, P.; Banerjee, M.; Giri, A.K. Role of genomic instability in arsenic-induced carcinogenicity. A review. Environ. Int. 2013, 53, 29–40. [Google Scholar] [CrossRef] [PubMed]
  257. Sakai, A.; Nakanishi, M.; Yoshiyama, K.; Maki, H. Impact of reactive oxygen species on spontaneous mutagenesis in Escherichia coli. Genes Cells 2006, 11, 767–778. [Google Scholar] [CrossRef] [PubMed]
  258. Tarantini, A.; Maitre, A.; Lefebvre, E.; Marques, M.; Marie, C.; Ravanat, J.-L.; Douki, T. Relative contribution of DNA strand breaks and DNA adducts to the genotoxicity of benzoapyrene as a pure compound and in complex mixtures. Mutat. Res. 2009, 671, 67–75. [Google Scholar] [CrossRef]
  259. Wise, J.T.F.; Lu, H.; Meaza, I.; Wise, S.S.; Williams, A.R.; Wise, J.Y.; Mason, M.D.; Wise, J.P. Prolonged Particulate Hexavalent Chromium Exposure Induces DNA Double-Strand Breaks and Inhibits Homologous Recombination Repair in Primary Rodent Lung Cells. Biol. Trace Elem. Res. 2024, 202, 5653–5663. [Google Scholar] [CrossRef]
  260. Kay, J.; Thadhani, E.; Samson, L.; Engelward, B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair 2019, 83, 102673. [Google Scholar] [CrossRef]
  261. Wu, R.; Högberg, J.; Adner, M.; Stenius, U.; Zheng, H. Crystalline silica particles induce DNA damage in respiratory epithelium by ATX secretion and Rac1 activation. Biochem. Biophys. Res. Commun. 2021, 548, 91–97. [Google Scholar] [CrossRef]
  262. Miller, J.L.; Grant, P.A. The role of DNA methylation and histone modifications in transcriptional regulation in humans. Epigenetics Dev. Dis. 2013, 61, 289–317. [Google Scholar] [CrossRef]
  263. Wang, B.; Li, Y.; Shao, C.; Tan, Y.; Cai, L. Cadmium and its epigenetic effects. Curr. Med. Chem. 2012, 19, 2611–2620. [Google Scholar] [CrossRef]
  264. Islam, R.; Zhao, L.; Wang, Y.; Lu-Yao, G.; Liu, L.-Z. Epigenetic Dysregulations in Arsenic-Induced Carcinogenesis. Cancers 2022, 14, 4502. [Google Scholar] [CrossRef] [PubMed]
  265. Sobanski, T.; Rose, M.; Suraweera, A.; O’Byrne, K.; Richard, D.J.; Bolderson, E. Cell Metabolism and DNA Repair Pathways: Implications for Cancer Therapy. Front. Cell Dev. Biol. 2021, 9, 633305. [Google Scholar] [CrossRef]
  266. Fang, Y.; Lu, L.; Liang, Y.; Peng, D.; Aschner, M.; Jiang, Y. Signal transduction associated with lead-induced neurological disorders: A review. Food Chem. Toxicol. 2021, 150, 112063. [Google Scholar] [CrossRef] [PubMed]
  267. Salim, S.Y.; Kaplan, G.G.; Madsen, K.L. Air pollution effects on the gut microbiota: A link between exposure and inflammatory disease. Gut Microbes 2014, 5, 215–219. [Google Scholar] [CrossRef] [PubMed]
  268. Vieceli, T.; Tejada, S.; Martinez-Reviejo, R.; Pumarola, T.; Schrenzel, J.; Waterer, G.W.; Rello, J. Impact of air pollution on respiratory microbiome: A narrative review. Intensive Crit. Care Nurs. 2023, 74, 103336. [Google Scholar] [CrossRef]
  269. Mutlu, E.A.; Comba, I.Y.; Cho, T.; Engen, P.A.; Yazıcı, C.; Soberanes, S.; Hamanaka, R.B.; Niğdelioğlu, R.; Meliton, A.Y.; Ghio, A.J.; et al. Inhalational exposure to particulate matter air pollution alters the composition of the gut microbiome. Environ. Pollut. 2018, 240, 817–830. [Google Scholar] [CrossRef]
  270. 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]
  271. Arca-Lafuente, S.; Nuñez-Corcuera, B.; Ramis, R.; Karakitsios, S.; Sarigiannis, D.; García Dos Santos, S.; Fernández-Rodríguez, A.; Briz, V. Effects of urban airborne particulate matter exposure on the human upper respiratory tract microbiome: A systematic review. Respir. Res. 2025, 26, 118. [Google Scholar] [CrossRef]
  272. Xue, Y.; Chu, J.; Li, Y.; Kong, X. The influence of air pollution on respiratory microbiome: A link to respiratory disease. Toxicol. Lett. 2020, 334, 14–20. [Google Scholar] [CrossRef]
  273. Kaplan, G.G.; Hubbard, J.; Korzenik, J.; Sands, B.E.; Panaccione, R.; Ghosh, S.; Wheeler, A.J.; Villeneuve, P.J. The inflammatory bowel diseases and ambient air pollution: A novel association. Am. J. Gastroenterol. 2010, 105, 2412–2419. [Google Scholar] [CrossRef]
  274. Ananthakrishnan, A.N.; McGinley, E.L.; Binion, D.G.; Saeian, K. Ambient air pollution correlates with hospitalizations for inflammatory bowel disease: An ecologic analysis. Inflamm. Bowel Dis. 2011, 17, 1138–1145. [Google Scholar] [CrossRef] [PubMed]
  275. Ahmed, H.; Leyrolle, Q.; Koistinen, V.; Kärkkäinen, O.; Layé, S.; Delzenne, N.; Hanhineva, K. Microbiota-derived metabolites as drivers of gut-brain communication. Gut Microbes 2022, 14, 2102878. [Google Scholar] [CrossRef] [PubMed]
  276. Leblhuber, F.; Ehrlich, D.; Steiner, K.; Geisler, S.; Fuchs, D.; Lanser, L.; Kurz, K. The Immunopathogenesis of Alzheimer’s Disease Is Related to the Composition of Gut Microbiota. Nutrients 2021, 13, 361. [Google Scholar] [CrossRef]
  277. Fouladi, F.; Bailey, M.J.; Patterson, W.B.; Sioda, M.; Blakley, I.C.; Fodor, A.A.; Jones, R.B.; Chen, Z.; Kim, J.S.; Lurmann, F.; et al. Air pollution exposure is associated with the gut microbiome as revealed by shotgun metagenomic sequencing. Environ. Int. 2020, 138, 105604. [Google Scholar] [CrossRef] [PubMed]
  278. Bailey, M.J.; Naik, N.N.; Wild, L.E.; Patterson, W.B.; Alderete, T.L. Exposure to air pollutants and the gut microbiota: A potential link between exposure, obesity, and type 2 diabetes. Gut Microbes 2020, 11, 1188–1202. [Google Scholar] [CrossRef]
  279. Alderete, T.L.; Jones, R.B.; Chen, Z.; Kim, J.S.; Habre, R.; Lurmann, F.; Gilliland, F.D.; Goran, M.I. Exposure to traffic-related air pollution and the composition of the gut microbiota in overweight and obese adolescents. Environ. Res. 2018, 161, 472–478. [Google Scholar] [CrossRef]
  280. Ghosh, S.; Nukavarapu, S.P.; Jala, V.R. Effects of heavy metals on gut barrier integrity and gut microbiota. Microbiota Host 2023, 2, e230015. [Google Scholar] [CrossRef]
  281. Teffera, M.; Veith, A.C.; Ronnekleiv-Kelly, S.; Bradfield, C.A.; Nikodemova, M.; Tussing-Humphreys, L.; Malecki, K. Diverse mechanisms by which chemical pollutant exposure alters gut microbiota metabolism and inflammation. Environ. Int. 2024, 190, 108805. [Google Scholar] [CrossRef]
  282. Chezganova, E.; Efimova, O.; Sakharova, V.; Efimova, A.; Sozinov, S.; Kutikhin, A.; Ismagilov, Z.; Brusina, E. Ventilation-Associated Particulate Matter Is a Potential Reservoir of Multidrug-Resistant Organisms in Health Facilities. Life 2021, 11, 639. [Google Scholar] [CrossRef]
  283. Utembe, W.; Kamng’ona, A.W. Inhalation exposure to chemicals, microbiota dysbiosis and adverse effects on humans. Sci. Total Environ. 2024, 955, 176938. [Google Scholar] [CrossRef]
  284. Yadav, B.; Bhattacharya, S.S.; Rosen, L.; Nagpal, R.; Yadav, H.; Yadav, J.S. Oro-Respiratory Dysbiosis and Its Modulatory Effect on Lung Mucosal Toxicity during Exposure or Co-Exposure to Carbon Nanotubes and Cigarette Smoke. Nanomaterials 2024, 14, 314. [Google Scholar] [CrossRef] [PubMed]
  285. Hiemstra, P.S.; Grootaers, G.; van der Does, A.M.; Krul, C.A.M.; Kooter, I.M. Human lung epithelial cell cultures for analysis of inhaled toxicants: Lessons learned and future directions. Toxicol. In Vitr. 2018, 47, 137–146. [Google Scholar] [CrossRef] [PubMed]
  286. Camilleri, M. Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut 2019, 68, 1516–1526. [Google Scholar] [CrossRef] [PubMed]
  287. Abrami, M.; Biasin, A.; Tescione, F.; Tierno, D.; Dapas, B.; Carbone, A.; Grassi, G.; Conese, M.; Di Gioia, S.; Larobina, D.; et al. Mucus Structure, Viscoelastic Properties, and Composition in Chronic Respiratory Diseases. Int. J. Mol. Sci. 2024, 25, 1933. [Google Scholar] [CrossRef]
  288. 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]
  289. Winans, B.; Humble, M.C.; Lawrence, B.P. Environmental toxicants and the developing immune system: A missing link in the global battle against infectious disease? Reprod. Toxicol. 2011, 31, 327–336. [Google Scholar] [CrossRef]
  290. Marín-Palma, D.; Fernandez, G.J.; Ruiz-Saenz, J.; Taborda, N.A.; Rugeles, M.T.; Hernandez, J.C. Particulate matter impairs immune system function by up-regulating inflammatory pathways and decreasing pathogen response gene expression. Sci. Rep. 2023, 13, 12773. [Google Scholar] [CrossRef]
  291. Sarigiannis, D.A.; Karakitsios, S.P.; Handakas, E.; Simou, K.; Solomou, E.; Gotti, A. Integrated exposure and risk characterization of bisphenol-A in Europe. Food Chem. Toxicol. 2016, 98, 134–147. [Google Scholar] [CrossRef]
  292. Ajisafe, O.M.; Adekunle, Y.A.; Egbon, E.; Ogbonna, C.E.; Olawade, D.B. The role of machine learning in predictive toxicology: A review of current trends and future perspectives. Life Sci. 2025, 378, 123821. [Google Scholar] [CrossRef]
  293. Li, L.; Westgate, J.N.; Hughes, L.; Zhang, X.; Givehchi, B.; Toose, L.; Armitage, J.M.; Wania, F.; Egeghy, P.; Arnot, J.A. A Model for Risk-Based Screening and Prioritization of Human Exposure to Chemicals from Near-Field Sources. Environ. Sci. Technol. 2018, 52, 14235–14244. [Google Scholar] [CrossRef]
  294. Batool, M.; Zamir, A.; Alqhtani, H.; Saeed, H.; Rasool, M.F. A Mechanistic Physiologically Based Pharmacokinetic (PBPK) modeling approach for fexofenadine: Predictive pharmacokinetic insights in humans. Saudi Pharm. J. 2025, 33, 24. [Google Scholar] [CrossRef]
  295. Hartmanshenn, C.; Scherholz, M.; Androulakis, I.P. Physiologically-based pharmacokinetic models: Approaches for enabling personalized medicine. J. Pharmacokinet. Pharmacodyn. 2016, 43, 481–504. [Google Scholar] [CrossRef]
  296. Escher, B.I.; Hackermüller, J.; Polte, T.; Scholz, S.; Aigner, A.; Altenburger, R.; Böhme, A.; Bopp, S.K.; Brack, W.; Busch, W.; et al. From the exposome to mechanistic understanding of chemical-induced adverse effects. Environ. Int. 2017, 99, 97–106. [Google Scholar] [CrossRef]
  297. Neo, E.X.; Hasikin, K.; Mokhtar, M.I.; Lai, K.W.; Azizan, M.M.; Razak, S.A.; Hizaddin, H.F. Towards Integrated Air Pollution Monitoring and Health Impact Assessment Using Federated Learning: A Systematic Review. Front. Public Health 2022, 10, 851553. [Google Scholar] [CrossRef]
  298. Sarigiannis, D.A.; Karakitsios, S.P.; Kermenidou, M.V. Health impact and monetary cost of exposure to particulate matter emitted from biomass burning in large cities. Sci. Total Environ. 2015, 524–525, 319–330. [Google Scholar] [CrossRef] [PubMed]
  299. Thaveevong, P.; Ahmad, M.; Panyametheekul, S.; Chetwittayachan, T.; Zhang, Y. Chemical composition and oxidative potential of fine particulate matter (PM2.5) in urban parks of Bangkok, Thailand: Implication for public health. Environ. Chall. 2025, 20, 101194. [Google Scholar] [CrossRef]
  300. Luo, X.-S.; Huang, W.; Shen, G.; Pang, Y.; Tang, M.; Li, W.; Zhao, Z.; Li, H.; Wei, Y.; Xie, L.; et al. Source differences in the components and cytotoxicity of PM 2.5 from automobile exhaust, coal combustion, and biomass burning contributing to urban aerosol toxicity. Atmos. Chem. Phys. 2024, 24, 1345–1360. [Google Scholar] [CrossRef]
  301. Kim Oanh, N.T.; Pongkiatkul, P.; Upadhyay, N.; Hopke, P.P. Designing ambient particulate matter monitoring program for source apportionment study by receptor modeling. Atmos. Environ. 2009, 43, 3334–3344. [Google Scholar] [CrossRef]
  302. Kuklinska, K.; Wolska, L.; Namiesnik, J. Air quality policy in the U.S. and the EU—A review. Atmos. Pollut. Res. 2015, 6, 129–137. [Google Scholar] [CrossRef]
  303. Cromar, K.R.; Lee, A.G.; Harkema, J.R.; Annesi-Maesano, I. Science-based Policy Recommendations for Fine Particulate Matter in the United States. Am. J. Respir. Crit. Care Med. 2022, 206, 1067–1069. [Google Scholar] [CrossRef]
  304. Glazener, A.; Khreis, H. Transforming Our Cities: Best Practices Towards Clean Air and Active Transportation. Curr. Environ. Health Rep. 2019, 6, 22–37. [Google Scholar] [CrossRef]
  305. Bouscasse, H.; Gabet, S.; Kerneis, G.; Provent, A.; Rieux, C.; Ben Salem, N.; Dupont, H.; Troude, F.; Mathy, S.; Slama, R. Designing local air pollution policies focusing on mobility and heating to avoid a targeted number of pollution-related deaths: Forward and backward approaches combining air pollution modeling, health impact assessment and cost-benefit analysis. Environ. Int. 2022, 159, 107030. [Google Scholar] [CrossRef]
  306. Filigrana, P.; Levy, J.I.; Gauthier, J.; Batterman, S.; Adar, S.D. Health benefits from cleaner vehicles and increased active transportation in Seattle, Washington. J. Expo. Sci. Environ. Epidemiol. 2022, 32, 538–544. [Google Scholar] [CrossRef] [PubMed]
  307. Park, J.-H.; Son, Y.-S.; Kim, K.-H. A review of traditional and advanced technologies for the removal of particulate matter in subway systems. Indoor Air 2019, 29, 177–191. [Google Scholar] [CrossRef] [PubMed]
  308. Pachaiappan, R.; Cornejo-Ponce, L.; Rajendran, R.; Manavalan, K.; Femilaa Rajan, V.; Awad, F. A review on biofiltration techniques: Recent advancements in the removal of volatile organic compounds and heavy metals in the treatment of polluted water. Bioengineered 2022, 13, 8432–8477. [Google Scholar] [CrossRef]
  309. Zhao, H.; Zhao, D.; Jiang, X.; Zhang, S.; Lin, Z. Assessment of Urban Forest Ecological Benefit Based on the i-Tree Eco Model—A Case Study of Changchun Central City. Forests 2023, 14, 1304. [Google Scholar] [CrossRef]
  310. Kacprzak, M.J.; Ellis, A.; Fijałkowski, K.; Kupich, I.; Gryszpanowicz, P.; Greenfield, E.; Nowak, D. Urban forest species selection for improvement of ecological benefits in Polish cities-The actual and forecast potential. J. Environ. Manag. 2024, 366, 121732. [Google Scholar] [CrossRef]
  311. Yin, L.; Yang, X.; Xing, Y.; Lu, P.; Tsai, C.-J.; Attoui, M.; Cui, Y.; Liu, Y.; Li, Z. Removal of ultrafine particles by porous nanomaterials with varied morphologies. Powder Technol. 2019, 342, 380–387. [Google Scholar] [CrossRef]
  312. Tong, Z.; Li, Y.; Westerdahl, D.; Freeman, R.B. The impact of air filtration units on primary school students’ indoor exposure to particulate matter in China. Environ. Pollut. 2020, 266, 115107. [Google Scholar] [CrossRef]
  313. Vijayan, V.K.; Paramesh, H.; Salvi, S.S.; Dalal, A.A.K. Enhancing indoor air quality—The air filter advantage. Lung India 2015, 32, 473–479. [Google Scholar] [CrossRef]
  314. Thompson, M.; Castorina, R.; Chen, W.; Moore, D.; Peerless, K.; Hurley, S. Effectiveness of Air Filtration in Reducing PM2.5 Exposures at a School in a Community Heavily Impacted by Air Pollution. Atmosphere 2024, 15, 901. [Google Scholar] [CrossRef]
  315. Chen, C.-F.; Hsu, C.-H.; Chang, Y.-J.; Lee, C.-H.; Lee, D.L. Efficacy of HEPA Air Cleaner on Improving Indoor Particulate Matter 2.5 Concentration. Int. J. Environ. Res. Public Health 2022, 19, 11517. [Google Scholar] [CrossRef]
  316. Dubey, S.; Rohra, H.; Taneja, A. Assessing effectiveness of air purifiers (HEPA) for controlling indoor particulate pollution. Heliyon 2021, 7, e07976. [Google Scholar] [CrossRef] [PubMed]
  317. Morishita, M.; Adar, S.D.; D’Souza, J.; Ziemba, R.A.; Bard, R.L.; Spino, C.; Brook, R.D. Effect of Portable Air Filtration Systems on Personal Exposure to Fine Particulate Matter and Blood Pressure Among Residents in a Low-Income Senior Facility: A Randomized Clinical Trial. JAMA Intern. Med. 2018, 178, 1350–1357. [Google Scholar] [CrossRef] [PubMed]
  318. Allen, R.W.; Barn, P. Individual- and Household-Level Interventions to Reduce Air Pollution Exposures and Health Risks: A Review of the Recent Literature. Curr. Environ. Health Rep. 2020, 7, 424–440. [Google Scholar] [CrossRef]
  319. Klaver, Z.M.; Crane, R.C.; Ziemba, R.A.; Bard, R.L.; Adar, S.D.; Brook, R.D.; Morishita, M. Reduction of Outdoor and Indoor PM2.5 Source Contributions via Portable Air Filtration Systems in a Senior Residential Facility in Detroit, Michigan. Toxics 2023, 11, 1019. [Google Scholar] [CrossRef]
  320. Miglani, R.; Parveen, N.; Kumar, A.; Ansari, M.A.; Khanna, S.; Rawat, G.; Panda, A.K.; Bisht, S.S.; Upadhyay, J.; Ansari, M.N. Degradation of Xenobiotic Pollutants: An Environmentally Sustainable Approach. Metabolites 2022, 12, 818. [Google Scholar] [CrossRef]
  321. Mishra, S.; Lin, Z.; Pang, S.; Zhang, W.; Bhatt, P.; Chen, S. Recent Advanced Technologies for the Characterization of Xenobiotic-Degrading Microorganisms and Microbial Communities. Front. Bioeng. Biotechnol. 2021, 9, 632059. [Google Scholar] [CrossRef]
  322. Priya, A.K.; Muruganandam, M.; Kumar, A.; Senthilkumar, N.; Shkir, M.; Pandit, B.; Imran, M.; Prakash, C.; Ubaidullah, M. Recent advances in microbial-assisted degradation and remediation of xenobiotic contaminants; challenges and future prospects. J. Water Process Eng. 2024, 60, 105106. [Google Scholar] [CrossRef]
  323. Bora, B.; Kauser, H.; Geed, S.R. Bioremediation strategies for xenobiotic degradation in petroleum-impacted industrial ecosystems: Practical challenges and future directions. J. Water Process Eng. 2025, 70, 106877. [Google Scholar] [CrossRef]
  324. Rathore, S.; Varshney, A.; Mohan, S.; Dahiya, P. An innovative approach of bioremediation in enzymatic degradation of xenobiotics. Biotechnol. Genet. Eng. Rev. 2022, 38, 1–32. [Google Scholar] [CrossRef]
  325. Heshammuddin, N.A.; Al-Gheethi, A.; Saphira Radin Mohamed, R.M.; Bin Khamidun, M.H. Eliminating xenobiotics organic compounds from greywater through green synthetic nanoparticles. Environ. Res. 2023, 222, 115316. [Google Scholar] [CrossRef]
  326. Rafeeq, H.; Afsheen, N.; Rafique, S.; Arshad, A.; Intisar, M.; Hussain, A.; Bilal, M.; Iqbal, H.M.N. Genetically engineered microorganisms for environmental remediation. Chemosphere 2023, 310, 136751. [Google Scholar] [CrossRef]
  327. Sharma, P.; Bano, A.; Singh, S.P.; Sharma, S.; Xia, C.; Nadda, A.K.; Lam, S.S.; Tong, Y.W. Engineered microbes as effective tools for the remediation of polyaromatic aromatic hydrocarbons and heavy metals. Chemosphere 2022, 306, 135538. [Google Scholar] [CrossRef] [PubMed]
  328. Hondal, R.J. Selenium vitaminology: The connection between selenium, vitamin C, vitamin E, and ergothioneine. Curr. Opin. Chem. Biol. 2023, 75, 102328. [Google Scholar] [CrossRef] [PubMed]
  329. Andrés, C.M.C.; La Pérez de Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences. Int. J. Mol. Sci. 2024, 25, 2600. [Google Scholar] [CrossRef] [PubMed]
  330. Hodges, R.E.; Minich, D.M. Modulation of Metabolic Detoxification Pathways Using Foods and Food-Derived Components: A Scientific Review with Clinical Application. J. Nutr. Metab. 2015, 2015, 760689. [Google Scholar] [CrossRef]
  331. Zivkovic, A.M.; Telis, N.; German, J.B.; Hammock, B.D. Dietary omega-3 fatty acids aid in the modulation of inflammation and metabolic health. Calif. Agric. 2011, 65, 106–111. [Google Scholar] [CrossRef]
  332. Guo, J.; Knol, L.L.; Yang, X.; Kong, L. Dietary fiber intake is inversely related to serum heavy metal concentrations among US adults consuming recommended amounts of seafood: NHANES 2013–2014. Food Front. 2022, 3, 142–149. [Google Scholar] [CrossRef]
  333. Espinosa-Vellarino, F.L.; Garrido, I.; Casimiro, I.; Silva, A.C.; Espinosa, F.; Ortega, A. Enzymes Involved in Antioxidant and Detoxification Processes Present Changes in the Expression Levels of Their Coding Genes under the Stress Caused by the Presence of Antimony in Tomato. Plants 2024, 13, 609. [Google Scholar] [CrossRef]
  334. Bermingham, E.N.; Hesketh, J.E.; Sinclair, B.R.; Koolaard, J.P.; Roy, N.C. Selenium-enriched foods are more effective at increasing glutathione peroxidase (GPx) activity compared with selenomethionine: A meta-analysis. Nutrients 2014, 6, 4002–4031. [Google Scholar] [CrossRef]
  335. Xu, W.; Barrientos, T.; Andrews, N.C. Iron and copper in mitochondrial diseases. Cell Metab. 2013, 17, 319–328. [Google Scholar] [CrossRef] [PubMed]
  336. Begum, S.Z.; Nizam, N.S.M.; Muhamad, A.; Saiman, M.I.; Crouse, K.A.; Abdul Rahman, M.B. Imidazole-rich copper peptides as catalysts in xenobiotic degradation. PLoS ONE 2020, 15, e0238147. [Google Scholar] [CrossRef] [PubMed]
  337. Chadalavada, S.; Faust, O.; Salvi, M.; Seoni, S.; Raj, N.; Raghavendra, U.; Gudigar, A.; Barua, P.D.; Molinari, F.; Acharya, R. Application of artificial intelligence in air pollution monitoring and forecasting: A systematic review. Environ. Model. Softw. 2025, 185, 106312. [Google Scholar] [CrossRef]
  338. Rowley, A.; Karakuş, O. Predicting air quality via multimodal AI and satellite imagery. Remote Sens. Environ. 2023, 293, 113609. [Google Scholar] [CrossRef]
  339. Olawade, D.B.; Wada, O.Z.; Ige, A.O.; Egbewole, B.I.; Olojo, A.; Oladapo, B.I. Artificial intelligence in environmental monitoring: Advancements, challenges, and future directions. Hyg. Environ. Health Adv. 2024, 12, 100114. [Google Scholar] [CrossRef]
  340. Naveed, K.; Umer, T.; Asghar, A.B.; Aslam, M.; Ejsmont, K.; Mohammed Metwally, A.S.; Thanh, K.N. Machine learning assisted predictive urban digital twin for intelligent monitoring of air quality index for smart city environment. Environ. Model. Softw. 2025, 192, 106559. [Google Scholar] [CrossRef]
  341. Subramaniam, S.; Raju, N.; Ganesan, A.; Rajavel, N.; Chenniappan, M.; Prakash, C.; Pramanik, A.; Basak, A.K.; Dixit, S. Artificial Intelligence Technologies for Forecasting Air Pollution and Human Health: A Narrative Review. Sustainability 2022, 14, 9951. [Google Scholar] [CrossRef]
  342. Molfino, N.A.; Turcatel, G.; Riskin, D. Machine Learning Approaches to Predict Asthma Exacerbations: A Narrative Review. Adv. Ther. 2024, 41, 534–552. [Google Scholar] [CrossRef]
  343. Sarigiannis, D.; Karakitsios, S.; Anesti, O.; Stem, A.; Valvi, D.; Sumner, S.C.J.; Chatzi, L.; Snyder, M.P.; Thompson, D.C.; Vasiliou, V. Advancing translational exposomics: Bridging genome, exposome and personalized medicine. Hum. Genom. 2025, 19, 48. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the uptake, systemic distribution, and health impacts of particulate matter (PM) based on the particle size. (A) Fine (PM2.5) and ultrafine particles (PM0.1) are inhaled into the lungs, where their size determines their deposition and interaction with lung tissue. Larger particles are primarily deposited in the upper airways, while smaller particles penetrate deeper into the alveoli. (B) Ultrafine particles can translocate across the alveolar–capillary barrier into the bloodstream, enabling systemic distribution to distant organs. The toxin-binding capacity of particles increases with a decreasing size, as smaller particles have a larger surface area-to-volume ratio, allowing them to carry more adsorbed toxic substances. (C) Systemic circulation of particles leads to inflammation, oxidative stress, and tissue damage in multiple organs, including the brain (neurodegeneration) and the heart (cardiovascular disease). Chronic exposure to particulate matter is associated with adverse health outcomes, including respiratory diseases, cardiovascular disorders, neurodegenerative diseases, and malignant transformation of cells, ultimately leading to cancer. (Created in BioRender. Reingruber, S. (2025), https://BioRender.com/5rrwxa2, accessed on 11 August 2025).
Figure 1. Schematic representation of the uptake, systemic distribution, and health impacts of particulate matter (PM) based on the particle size. (A) Fine (PM2.5) and ultrafine particles (PM0.1) are inhaled into the lungs, where their size determines their deposition and interaction with lung tissue. Larger particles are primarily deposited in the upper airways, while smaller particles penetrate deeper into the alveoli. (B) Ultrafine particles can translocate across the alveolar–capillary barrier into the bloodstream, enabling systemic distribution to distant organs. The toxin-binding capacity of particles increases with a decreasing size, as smaller particles have a larger surface area-to-volume ratio, allowing them to carry more adsorbed toxic substances. (C) Systemic circulation of particles leads to inflammation, oxidative stress, and tissue damage in multiple organs, including the brain (neurodegeneration) and the heart (cardiovascular disease). Chronic exposure to particulate matter is associated with adverse health outcomes, including respiratory diseases, cardiovascular disorders, neurodegenerative diseases, and malignant transformation of cells, ultimately leading to cancer. (Created in BioRender. Reingruber, S. (2025), https://BioRender.com/5rrwxa2, accessed on 11 August 2025).
Jox 15 00131 g001
Table 1. Comparison of classical xenobiotic toxicants and particulate matter: nature, composition, exposure, and health impacts.
Table 1. Comparison of classical xenobiotic toxicants and particulate matter: nature, composition, exposure, and health impacts.
Classical Xenobiotic ToxicantsParticulate Matter
Nature and OriginChemicals not naturally produced by the body or commonly found in the environment (e.g., pesticides, industrial chemicals, and pharmaceuticals) [17,18,19].Complex mixture of tiny particles and liquid droplets suspended in the air, originating from both natural sources (e.g., sand, dust, pollen, sea salts) and anthropogenic sources (e.g., emissions from vehicles, industrial activities) [20,21].
CompositionSpecific chemical substances with defined molecular structures [22].Often contains a variety of substances, including xenobiotic and toxic pollutants like heavy metals and organic compounds; particles themselves are not necessarily toxic but exert a variety of physiological and pathophysiological effects [13].
Health EffectsDirectly cause adverse health effects due to their chemical properties and interactions with biological systems [23,24].Exerts effects through mechanisms such as the generation of ROS and oxidative stress, leading to cellular damage, inflammation, and disruption of normal cellular functions [25,26].
Exposure PathwaysTypically enter the body through ingestion, inhalation, or dermal absorption [27,28,29].Primarily enters the body through inhalation and ingestion, depositing in the respiratory system and the gastrointestinal tract and potentially causing systemic effects [30,31].
Health OutcomesAssociated with specific toxicological effects depending on the substance (e.g., neurotoxicity, carcinogenicity) [32,33].Linked to a broad range of health issues, including respiratory diseases (e.g., asthma, COPD), cardiovascular diseases (e.g., heart attacks, hypertension), neurological effects (e.g., cognitive decline, neurodegenerative diseases), and reproductive and developmental issues (e.g., birth defects, low birth weight) [34,35,36].
ROS, reactive oxygen species; COPD, chronic obstructive pulmonary disease.
Table 2. Toxicokinetics and toxicodynamics of key compounds in particulate matter: mechanisms of absorption, distribution, and potential health effects.
Table 2. Toxicokinetics and toxicodynamics of key compounds in particulate matter: mechanisms of absorption, distribution, and potential health effects.
CompoundToxicokineticsToxicodynamics
Heavy Metals (e.g., Pb, Cd, As)Soluble metals can cross biological barriers (e.g., alveolar–capillary membrane) and enter the bloodstream, leading to systemic distribution; may accumulate in organs such as the liver and kidneys [88,89].Can interfere with cellular processes by binding to proteins and enzymes; induces oxidative stress and inflammation, contributing to organ damage and chronic diseases [90].
Transition Metals (e.g., Fe, Cu, Ni)Catalyze the production of ROS via Fenton-like reactions; may remain in lung tissues, prolonging exposure [91].ROS generation leads to oxidative damage to lipids, proteins, and DNA; contributes to respiratory and cardiovascular diseases [91].
Polycyclic Aromatic Hydrocarbons (PAHs)Lipophilic nature allows absorption through the respiratory tract and distribution into fatty tissues; metabolized in the liver via cytochrome P450 enzymes, forming reactive intermediates [92].Reactive intermediates can form DNA adducts, leading to mutations and increased cancer risk; induces oxidative stress and inflammation [93].
Nitro-PAHsSimilar to PAHs, nitro-PAHs are metabolized into reactive intermediates; can persist in tissues due to their chemical stability.Forms DNA adducts, increasing genotoxicity and cancer risk; potent inducers of oxidative stress [94].
Endotoxins (Biological Sources)Endotoxins are not absorbed systemically but remain in the respiratory tract, where they interact with immune cells.Activates TLRs on immune cells, triggering inflammation and release of pro-inflammatory cytokines; exacerbates respiratory diseases like asthma and COPD [95].
Soluble Metals (e.g., Ni, V)Soluble metals are absorbed into the bloodstream and distributed systemically; can accumulate in cardiovascular tissues.Causes systemic inflammation, endothelial dysfunction, and increased blood coagulation; contributes to cardiovascular diseases [96].
Neurotoxic Metals (e.g., Mn)Can cross the blood–brain barrier after systemic absorption; accumulates in brain tissues, leading to prolonged exposure [97].Induces neuroinflammation and oxidative stress in the brain; associated with neurodegenerative diseases like Parkinson’s and Alzheimer’s [98].
ROS, reactive oxygen species; PAH, polycyclic aromatic hydrocarbon; TLR, toll-like receptor; COPD, chronic obstructive pulmonary disease.
Table 3. Mechanisms of interaction between xenobiotic toxicants and particulate matter and implications for distribution, bioavailability, and toxicity.
Table 3. Mechanisms of interaction between xenobiotic toxicants and particulate matter and implications for distribution, bioavailability, and toxicity.
MechanismExamples
Adsorption via Van der Waals ForcesWeak intermolecular forces allow xenobiotic and toxic substances to adhere to the surface of particulate matter.PAHs adsorbing onto soot particles in urban air pollution [100,101].
Electrostatic AttractionCharged xenobiotic toxicant molecules bind to oppositely charged particulate matter.Heavy metals like Pb or Cd binding to negatively charged fine dust particles [102].
Hydrophobic InteractionsNonpolar xenobiotic toxicant substances bind to hydrophobic surfaces of particulate matter.Persistent organic pollutants, such as dioxins, binding to carbonaceous PM [103,104].
Covalent Bond FormationXenobiotic and toxic substances form covalent bonds with reactive functional groups on particulate matter.Reactive aldehydes or ketones forming covalent bonds with organic matter in PM [105].
Physical EntrapmentXenobiotic and toxic substances become physically trapped within porous particulate matter.VOCs trapped in porous volcanic ash or activated carbon particles [106,107].
Ion ExchangeIonic xenobiotic toxicants exchange with ions on the surface of particulate matter.Ammonium ions (NH4+) from fertilizers binding to mineral dust particles [108].
Complexation with Metal OxidesXenobiotic and toxic substances form complexes with metal oxides present on particulate matter.Forming complexes with iron oxides in PM from mining activities [109].
Hydrogen BondingHydrogen bonds form between xenobiotic toxicants and functional groups on particulate matter.Phenolic compounds binding to hydroxyl groups on silica particles [110].
Surface CoatingXenobiotic and toxic substances coat the surface of particulate matter, forming a thin layer.Pesticides like DDT coating soil dust particles during agricultural spraying [111,112].
Aggregation with Organic MatterXenobiotic and toxic substances aggregate with organic matter present in particulate matter.PCBs binding to organic carbon in PM from industrial emissions [113].
PAH, polycyclic aromatic hydrocarbon; VOC, volatile organic compound; DDT dichlordiphenyltrichlorethan; PCB, polychlorinated biphenyl.
Table 4. Interactions between particulate matter and different immune cell types—possible effects on human health and disease.
Table 4. Interactions between particulate matter and different immune cell types—possible effects on human health and disease.
Immune Cell TypeType of InteractionEffects in the Human Body
MacrophagesPhagocytosis of particles; activation via pattern recognition receptors (e.g., TLRs) [160].Release of pro-inflammatory cytokines (e.g., IL-6, TNF-α, IL-1β); ROS generation; NLRP3 inflammasome activation; chronic inflammation and tissue damage in the lungs; cancer metastasis [160,161].
NeutrophilsChemotaxis in response to cytokines; direct activation by particles [162].Increased neutrophil infiltration, NETosis, ROS release, tissue damage, acute lung injury; exacerbation of respiratory diseases (e.g., asthma, COPD) [163].
Dendritic Cells (DCs)Uptake of particulate matter and presentation of associated antigens; activation by associated components; AhR-mediated signaling [164].Impaired antigen presentation and T cell activation; skewing of adaptive immune responses (e.g., Th2 or Th17 cell dominance) [165].
T Helper Cells (Th Cells)Skewing of Th cell subsets by associated components; Th2 and Th17 activation by particle-induced cytokines [166].Th2 dominance promotes allergic inflammation (e.g., asthma); Th17 activation contributes to autoimmune diseases and chronic inflammation [167].
Regulatory T Cells (Tregs)Inhibited induction via tolerogenic DCs; suppression of Treg activity; impaired immune regulation due to oxidative stress and inflammation [168].Reduced immune tolerance, leading to exacerbation of autoimmune diseases; increased risk of chronic inflammation [169].
Natural Killer (NK) CellsIndirect modulation via cytokine shifts and particle-induced stress [31].Impaired cytotoxicity; increased susceptibility to infection and tumor growth [170].
B cellsAntigen exposure and co-stimulation [157].Elevated IgE production; exacerbation of allergic responses [157,171].
Epithelial CellsDirect interaction with particles deposited in the respiratory tract; activation via ROS and cytokines [172].Release of pro-inflammatory mediators (e.g., IL-8, granulocyte–macrophage colony-stimulating factor); recruitment of immune cells to the site of exposure; damage to epithelial barriers, increasing susceptibility to infections [173].
Microglia (Brain Immune Cells)Activation by particles translocated to the brain via the olfactory nerve or systemic circulation [174].Neuroinflammation and oxidative stress in the brain; linked to neurodegenerative diseases (e.g., Alzheimer’s, Parkinson’s) [175].
TLR, toll-like receptor; IL, interleukin; TNF, tumor necrosis factor; ROS, reactive oxygen species; NLRP, nucleotide-binding domain, leucine-rich-containing family, pyrin domain; COPD, chronic obstructive pulmonary disease; AhR, aryl hydrocarbon receptor; Th, T helper; DC, dendritic cell; Treg, regulatory T cell; NK, natural killer cell.
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

Lang, T.; Lipp, A.-M.; Wechselberger, C. Xenobiotic Toxicants and Particulate Matter: Effects, Mechanisms, Impacts on Human Health, and Mitigation Strategies. J. Xenobiot. 2025, 15, 131. https://doi.org/10.3390/jox15040131

AMA Style

Lang T, Lipp A-M, Wechselberger C. Xenobiotic Toxicants and Particulate Matter: Effects, Mechanisms, Impacts on Human Health, and Mitigation Strategies. Journal of Xenobiotics. 2025; 15(4):131. https://doi.org/10.3390/jox15040131

Chicago/Turabian Style

Lang, Tamara, Anna-Maria Lipp, and Christian Wechselberger. 2025. "Xenobiotic Toxicants and Particulate Matter: Effects, Mechanisms, Impacts on Human Health, and Mitigation Strategies" Journal of Xenobiotics 15, no. 4: 131. https://doi.org/10.3390/jox15040131

APA Style

Lang, T., Lipp, A.-M., & Wechselberger, C. (2025). Xenobiotic Toxicants and Particulate Matter: Effects, Mechanisms, Impacts on Human Health, and Mitigation Strategies. Journal of Xenobiotics, 15(4), 131. https://doi.org/10.3390/jox15040131

Article Metrics

Back to TopTop