Abstract
Chronic obstructive pulmonary disease (COPD) continues to be a leading cause of mortality worldwide. Emerging evidence increasingly identifies exposure to fine particulate matter (PM2.5) as a significant risk factor, in addition to cigarette smoking, highlighting a critical gap in available treatment options. This review synthesizes epidemiological and experimental data that documents the correlation between airborne PM2.5 and the incidence and exacerbations of COPD. Numerous studies have demonstrated that PM2.5 exposure exacerbates COPD, manifesting in reduced lung function, lung and systemic inflammation, oxidative stress, mitochondrial dysfunction, cell death, emphysema, and small airway remodeling, all of which involve complex molecular pathways. Understanding the cellular and molecular mechanisms underlying these effects may inform the development of potential therapeutic interventions, mainly including reactive oxygen species scavengers, anti-inflammatory agents, specific pathway regulators, and traditional Chinese medicine. Future research should prioritize longitudinal exposure assessments and the development of personalized mitigation strategies to address the increasing burden of COPD in regions with high-pollution levels. This review serves as a stark reminder of the urgent necessity for substantive measures to combat air pollution, with the aim of reducing the associated health burden on our expanding global population.
Keywords
Particulate matter, COPD, Epidemiological evidence, Experimental evidence, Inflammation, Oxidative stress
Introduction
Chronic obstructive pulmonary disease (COPD) is a widely prevalent, preventable, and treatable chronic respiratory disorder characterized by persistent airflow limitation and progressive respiratory symptoms [1]. According to the World Health Organization (WHO), COPD has been the third leading cause of death globally, accounting for over 3 million deaths annually, with mortality rates continuing to rise, particularly in low- and middle-income countries. The Global Burden of Disease (GBD) Study estimates that COPD affects approximately 392 million individuals globally, significantly contributing to years lived with disability (YLDs) and disability-adjusted life years (DALYs) [2]. For the past decades, cigarette smoking (CS) has been identified as the primary risk factor for the development of COPD. However, recent data suggest that only a small proportion of smokers (15–20%) comprise the total COPD patient population, underscoring the critical role of other harmful exposures, such as ambient and household air pollution, occupational hazards, and dust exposure, in the development and progression of the disease [3]. The rising prevalence of COPD among non-smokers is a worrying concern globally.
Since the onset of the Industrial Revolution in the 18th century, anthropogenic activities have significantly degraded air quality, with particulate matter (PM) pollution emerging as a major environmental health concern. As of 2024, WHO reports that 99% of the global population lives in regions where air pollution levels surpass recommended safety limits, resulting in approximately 6.7 million premature deaths annually due to exposure to both ambient (outdoor) and household air pollution. Among the most hazardous air pollutants is fine particulate matter (PM2.5), defined as airborne particles with an aerodynamic diameter of ≤2.5 micrometers (μm). Due to their ultrafine size and high specific surface area, PM2.5 particles efficiently adsorb toxic compounds, such as heavy metals, polycyclic aromatic hydrocarbons (PAHs), and sulfates, thereby enhancing their biological reactivity when inhaled [4].
Upon inhalation, PM2.5 particles evade mucociliary clearance and penetrate deeply into the alveolar regions, where they induce sustained pulmonary inflammation, oxidative stress, and cellular damage through mechanisms such as mitochondrial dysfunction, ferroptosis, and epigenetic modifications [5]. Epidemiological studies have demonstrated a dose-dependent correlation between PM2.5 exposure and the incidence, exacerbation, and mortality of respiratory diseases, including asthma, COPD, and lung cancer [6]. Importantly, long-term exposure to PM2.5 is linked to accelerated decline in lung function, increased hospitalization rates for COPD exacerbations, and heightened risk of lung cancer, particularly in urban populations and vulnerable subgroups, such as children, the elderly, and individuals with pre-existing cardiopulmonary conditions [7]. Given its widespread presence and profound pathological impacts, PM2.5 pollution remains a critical public health concern, necessitating the implementation of stringent regulatory policies and targeted mitigation strategies to reduce its disease burden.
Methodology
We conducted a comprehensive literature search up to June 10, 2025, utilizing PubMed, Web of Science, CINAHL, and CNKI databases. We employed paired keyword searches, including "PM2.5", "particulate matter", "air pollutants", "environmental pollution", "COPD", and "respiratory diseases". A total of 2,714 articles were retrieved from the above database. After removing duplicates and excluding review articles, 80 studies were selected for inclusion in the present review.
PM Characterization
Classification of PM
Atmospheric particulate matter (PM) represents a complex mixture of solid and liquid particles suspended in the air, varying significantly in size, composition, and origin [8]. Based on aerodynamic diameter, PM is categorized into three principal classes: coarse particles (PM10, 2.5–10 μm), fine particles (PM2.5, 0.1–2.5 μm), and ultrafine particles (UFPs, ≤0.1 μm). These fractions exhibit distinct deposition patterns within the respiratory tract, determining their pathogenic potential [9]. Coarse particles predominantly deposit in the nasopharyngeal and tracheobronchial regions, where mucociliary clearance mechanisms provide partial protection [10]. In contrast, PM2.5 penetrates more deeply into the bronchioles and alveolar spaces, while UFPs—due to their nanoscale dimensions—achieve nearly complete alveolar deposition and even systemic translocation across the air-blood barrier [11]. Although the lungs are the primary target for exposure to ambient pollution, particles can also be detected in other organs, including the liver [12], kidneys [13], heart [14], and brain [15]. Moreover, this characteristic of PM2.5 enhances its adsorption of toxic compounds, including transition metals and organic carcinogens. Mounting studies have suggested that smaller PM can induce more significant mitochondrial dysfunction and DNA damage at considerably lower mass concentrations than larger PM, highlighting their unique toxicological profile [16]. Both short- and long-term exposure to PM significantly impacts human health and is closely associated with a range of acute and chronic disorders.
Sources and composition of PM2.5
PM2.5 originates from both anthropogenic and natural emission sources, with its chemical composition varying markedly by geographic region and pollution type. Primary anthropogenic sources encompass fossil fuel combustion, such as vehicular exhaust and coal-fired power plants, as well as industrial processes like smelting and cement production, and biomass burning. Secondary formation occurs through atmospheric reactions involving sulfur dioxide (SO2), nitrogen oxides (NOx), and volatile organic compounds (VOCs), significantly contributing to regional PM2.5 burdens, particularly in the form of ammonium sulfate and nitrate aerosols [17].
Geochemical signatures reveal distinct compositional profiles: Urban PM2.5 in East Asia is typically sulfate-dominated, indicative of coal combustion emissions, whereas European cities exhibit higher nitrate fractions due to the prevalence of diesel vehicles [18,19]. Industrial areas are characterized by an enrichment of heavy metals, such as lead, cadmium, and arsenic, while rural regions affected by agricultural burning show elevated levels of potassium-containing particles and organic carbon [20]. Notably, traffic-derived PM2.5 contains abundant elemental carbon (EC) and polycyclic aromatic hydrocarbons (PAHs), components that are strongly associated with oxidative stress generation.
The toxic potential of PM2.5 is also composition-dependent. Water-soluble ions, such as sulfates and nitrates, facilitate radical generation through aqueous-phase chemical reactions, whereas transition metals like iron and copper are involved in Fenton reactions. Organic constituents, including PAHs and quinones, undergo metabolic activation to form DNA-adducting species [21]. This chemical heterogeneity necessitates source-specific risk assessment, as demonstrated by cohort studies that link traffic-related PM2.5 with stronger associations with COPD than crustal dust particles of equivalent size.
Epidemiological and Experimental Evidence for PM2.5-induced COPD
Epidemiological evidence
COPD is a major public health concern in China, with its prevalence among individuals aged ≥40 years increasing markedly from approximately 8.2% during 2002–2004 to 13.7% in 2012–2015. This 5.5 percentage point rise highlights the escalating burden of COPD, with ambient air pollution—particularly PM2.5—emerging as a critical environmental risk factor [22]. The COPD burden attributable to ambient PM2.5 exposure has shown a substantial increase from 1990 to 2019. Over the past two decades, both epidemiological and toxicological studies have progressively established a robust association between PM2.5 exposure and the incidence, severity, and mortality of COPD, thereby informing current understanding and policy interventions [23].
Initial research in the early 2000s primarily focused on smoking as the leading cause of COPD. However, with the rapid urbanization in China, air pollution has gained increasing recognition as a significant contributing factor. Early cross-sectional surveys revealed that regions with higher PM2.5 concentrations exhibited elevated COPD prevalence, even after adjusting for smoking rates Meta-analyses conducted during this period confirmed a positive correlation between long-term PM2.5 exposure and COPD morbidity, with particulate matter identified as a key driver of COPD mortality in low socio-demographic index (SDI) regions [24]. These findings suggested that air pollution could independently exacerbate the respiratory disease burden.
In 2020, a systematic review of 107 epidemiological studies revealed that chronic exposure to PM2.5 at levels below the WHO Air Quality Guidelines (AQG) mean annual exposure limit of 10 μg.m-3 is correlated with heightened mortality rates due to respiratory diseases such as COPD, acute lower respiratory tract infection, and cardiovascular disease. Prolonged exposure to PM2.5 has been shown to provoke pulmonary inflammation, accelerate fibrosis, and impair lung function, particularly among vulnerable populations such as the elderly (≥60 years) and smokers [25]. Additionally, short-term exposure to PM2.5 has also been associated with increased airway inflammation and respiratory symptoms in COPD patients in China. Dose-response analyses have indicated that each 10 μg.m-³ increase in PM2.5 concentration elevates the risk of hospitalization for COPD by 2.5%, with acute exacerbations (AECOPD) emerging as a critical concern [26]. Epidemiological data have demonstrated that COPD patients experience between 0.5 to 3.5 acute exacerbations annually, substantially contributing to mortality rates [27]. Currently, an expanding body of evidence suggests that exposure to even very low levels of PM2.5 is a genuine public health concern. Considering this new evidence, the WHO revised its AQG in 2021 to a recommended maximal annual average PM2.5 exposure of 5 μg.m-3 [28].
Furthermore, the aging population in China has further exacerbated COPD mortality, as older adults with pre-existing conditions exhibit heightened susceptibility to PM2.5 toxicity. Immunological decline and reduced physical activity in this demographic further intensify the adverse effects of air pollution [29]. Meanwhile, global reductions in smoking rates have partially offset COPD mortality growth, underscoring the multifactorial nature of the disease. Predictive models suggest that the interaction between persistent air pollution and aging may lead to an epidemiological turning point in COPD mortality by 2030 [30]. Key epidemiological studies examining the association between PM2.5 and COPD are presented in Table 1.
|
Location |
Study design |
Sample size |
Health effects |
Research Quality Assessment |
Outcome |
References |
|
Beijing |
Cohort |
75 |
Prevalence |
This study employed an iron tracer method to apportion indoor PM2.5 sources. Source separation was achieved through synchronized indoor/outdoor sampling over five consecutive days, chemical characterization of 21 components, and exposure assessment using infiltration factor modeling. Limitations include strong model dependency and the absence of gaseous pollutant monitoring. |
Exposure to PM2.5 in indoor and outdoor environments exerts differential effects on the cardiopulmonary function of healthy individuals and patients with COPD. |
[29] |
|
World (1990-2019) |
Cohort |
- |
Premature deaths |
This study employed a satellite-ground fused model coupled with chemical transport modeling to estimate global ambient PM2.5 annual exposure levels. Health effects were characterized using MR-BRT risk curves. Limitations include strong model dependency, failure to account for differential toxicity of PM 2.5 components, and lack of adjustment for indoor-outdoor exposure differences.
|
From 1990 to 2019, both deaths and disability-adjusted life years (DALYs) attributable to ambient PM2.5 among persons with COPD rose by over 90%, with the highest increases observed among those aged 70–89 years and 60–84 years, respectively. |
[24] |
|
World (2010-2022) |
Meta-analysis |
9,466 |
Hospitalization |
This study assessed daily PM2.5 exposure levels (lag days 0–7) using government environmental monitoring station data. Limitations include failure to distinguish individual mobility and indoor-outdoor differences, and inability to incorporate heterogeneity in the toxicity of PM2.5 chemical components. |
Short-term exposure to PM2.5 increases the risk of hospitalization among patients with COPD, with each 10 μg/m³ increase in daily PM2.5concentration associated with a 1.6% rise in COPD hospital admissions. |
[60] |
|
China (1990-2021) |
Decomposition analysis |
- |
Premature deaths |
This study employed a population-level PM2.5 exposure assessment approach based on the Global Burden of Disease (GBD) dataset. Population Attributable Fractions (PAFs) were calculated using the theoretical minimum risk exposure level (TMREL; 2.4–5.9 μg/m³), without integration of individual exposure measurements or adjustment for confounding factors, incurring potential ecological bias. |
The crude mortality rates attributable to PM2.5 increased significantly for LC (500.40%) and COPD (85.26%). 43.0% of the increase in LC mortality was due to population aging, and 57.0% was attributed to changes in other risk factors. For COPD, population aging contributed to an increase, whereas other risk factors reduced mortality. |
[30] |
|
Beijing (2013-2017) |
Ecological analysis |
161,613 |
Hospitalization |
This study utilized daily average concentration data (2013–2017) from 35 official monitoring stations in Beijing to assess exposure levels to pollutants including PM10, PM2.5, NO2, SO2, CO (24-hour mean), and O3 (maximum 8-hour average). Associations with hospital admissions for COPD exacerbations were analyzed using lag models (lag0–lag4) and exposure-response curves. Missing PM10 data were reconstructed based on the annual mean ratio of PM2.5 to PM10. |
During this five-year period, the average ambient concentration of SO2 decreased by 68%, while that of PM2.5 decreased by 33%. Females and patients aged 65 years and older exhibited greater susceptibility to the impact of pollutants on hospitalization risk than males and patients under 65 years of age. |
[22] |
|
Shanxi (2018-2022) |
Cohort |
1284 |
Hospitalization |
This study utilized monthly average PM2.5 concentrations from Shanxi Province (TAP database) as the exposure metric. Admission dates of 1,284 patients hospitalized for AECOPD were temporally matched by season. Collagen deposition in lung tissue (a marker of fibrosis) was validated via Masson’s trichrome staining in relation to PM2.5 exposure. Nevertheless, individual exposure levels were not quantified. |
Elevated PM2.5 exposure significantly increases the risk of AECOPD, linked to heightened inflammation, pulmonary fibrosis, and diminished pulmonary function, and the majority of these patients are over 60 years old or are smokers. |
[27] |
|
South Korea |
Cohort |
1,404,505 |
Hospitalization |
This nationwide ecological study investigated associations between ambient air pollutants (including PM2.5, NO2, among others), meteorological factors (such as temperature and DTR), and acute exacerbation of COPD. However, due to reliance on group-level exposure data and incomplete adjustment for confounding factors, the findings are at moderate risk of bias. |
PM2.5, PM10, CO, NO2, O3, SO2, average temperature, humidity, and DTR affected the incidence of COPD exacerbations in various patterns, up to 10 lag days. |
[61] |
|
North Carolina (2004–2016) |
Cohort |
520 |
Hospitalization |
This study employed a case-crossover design to investigate the impact of short-term PM2.5 exposure on COPD hospitalizations and, for the first time, assessed the modifying effect of long-term exposure. Although overall associations were null or inverse, stratification revealed a potentially elevated risk (OR >1) in the high long-term exposure group. Key limitations include limited power due to the small sample size, selection bias from a single healthcare system, and inadequate control for confounders. |
Among patients residing in areas with higher annual PM2.5 concentrations, the association between short-term PM2.5 exposure and hospital admissions was stronger than among those in areas with lower annual concentrations. |
[62] |
|
Canadian |
Cohort |
1,452 |
Lung function |
Leveraging the Canadian Cohort of Obstructive Lung Disease (CanCOLD) database, this study evaluated intra-urban air-pollution exposure effects while integrating multi-year cumulative exposure estimates and individual time–activity patterns to minimize non-differential exposure misclassification. Covariate adjustment encompassed core demographic variables (age, sex), additional contextual factors, and stratified analyses by sex and smoking status. However, the report provides no information on event counts or formal bias-risk ratings. |
Ambient air pollution exposure was associated with lower lung function, even at relatively low concentrations. Individuals with dysanaptic lung growth might be particularly susceptible to inhaled ambient air pollutants, especially those at the extremes of dysanapsis. |
[63] |
|
Denmark Sweden |
Cohort |
98,058 |
Incidence |
Drawing on three cohorts within the ELAPSE consortium (n=98,058), the study identified 4,928 incident COPD cases over a mean follow-up of 16.6 years. Exposure assessment referenced the year 2010 and employed back-extrapolation as well as time-varying exposure metrics. Covariate adjustment was implemented in two nested models: Model 1 included age, sex, and additional basic covariates; Model 2 further adjusted for smoking status. No formal bias-risk rating was reported. |
Long-term exposure to low-level air pollution is associated with the development of COPD, even below current EU and US limit values and possibly WHO guidelines. |
[64] |
|
Poland |
Ecological Study |
910,372 |
Hospitalization |
Through systematic exposure assessment and stratified analyses, this study corroborates the association between ambient air pollution particularly SO2 and meteorological variability (temperature fluctuations) and emergency COPD hospitalizations in Poland. Owing to its observational design and residual potential for uncontrolled confounding, these findings warrant corroboration by complementary investigations. |
A significant association between air pollution levels, meteorological factors, and the number of emergency admissions of patients diagnosed with COPD. |
[65] |
Experimental evidence
While robust epidemiological evidence establishes a link between ambient PM2.5 exposure and COPD, a detailed mechanistic understanding of the cellular and molecular pathways involved in PM2.5-induced COPD remains nascent. To elucidate the potential mechanistic pathophysiology, researchers have conducted both in vitro and in vivo experimental studies. The characteristic features of chronic inflammation, oxidative stress, and airway remodeling found in COPD have been replicated in experimental models following exposure to PM2.5. These experimental studies are summarized in Table 2 (in vivo) and Table 3 (in vitro), respectively, detailing the type, dose, and duration of PM2.5, along with the major effects observed in each study.
Effect of PM2.5 exposure on inflammation
The involvement of PM2.5 in the pathogenesis of COPD via inflammatory mechanisms has been extensively investigated in recent years. An increasing volume of evidence indicates that inflammation induced by PM2.5 significantly contributes to both the initiation and progression of COPD through complex interactions between innate and adaptive immune responses, ultimately leading to airway remodeling and a decline in lung function [31,32].
Upon inhalation, PM2.5 is recognized by alveolar macrophages and epithelial cells through pattern recognition receptors (PRRs), initiating downstream signaling pathways such as the NF-κB pathway [33]. This activation leads to the nuclear translocation of transcription factors and upregulation of pro-inflammatory cytokines and chemokines, which facilitate the recruitment and activation of neutrophils, eosinophils, and macrophages within the airways. Recent experimental studies have demonstrated that PM2.5 exposure induces an inflammatory phenotype in mice, characterized by immune cell infiltration and pro-inflammatory cytokine release via the NF-κB pathway. In COPD rat models, exposure to PM2.5 has been shown to exacerbate pulmonary inflammation, as evidenced by elevated inflammatory cell counts in bronchoalveolar lavage fluid (BALF) and increased levels of interleukin-1β (IL-1β), granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-4, which collectively contribute to further tissue damage [34]. Moreover, even a drastically lower PM2.5 exposure (5 μg.day-1) was sufficient to induce COPD-like pathology, characterized by marked pulmonary inflammation, lung damage, and airway remodeling in mice [35].
|
Animal species |
Exposure materials |
Exposure method |
Dose and exposure period |
Research Quality Assessment |
Major effects on Lungs |
Reference |
|
C57BL/6 mice |
PM2.5 |
Oropharyngeal aspiration |
6 h/day for 6 days/week for a period of 24 weeks of 28.07±8.84 mg m-3 |
In the experimental design of this study, the exposure parameters for both animal and cell experiments were clearly defined, and the sample preparation and handling procedures were standardized. Partial bias was controlled through randomized allocation and validation of results using multiple methods. The overall experimental quality is methodologically reliable. However, the study lacks standardized rating for bias risk and did not address covariate adjustment. |
In the advanced stage of PM2.5-associated COPD mouse models, M2 macrophages emerge as the dominant phenotype. These macrophages upregulate MMP12 expression via the IL-4/STAT6 pathway, mediating epithelial barrier damage and excessive extracellular matrix (ECM) degradation, thereby driving COPD progression. |
[40] |
|
BALB/c |
PM2.5 and CS |
Intratracheal instillation |
4 weeks CS and 10 mg/kg PM2.5 for 7 days |
In the experimental design of this study, the preparation of exposure sources for both cell and animal experiments were standardized, and the exposure parameters were clearly defined. Partial bias was controlled through randomized allocation, standardized experimental conditions, and validation of results using multiple methods. The overall experimental quality is methodologically reliable. However, the study lacks standardized assessment of bias risk and did not address covariate adjustment. |
Targeting NOX4 inhibits ROS generation, thus suppressing excessive mitophagy activation and ameliorating PM2.5 plus cigarette smoke-induced acute exacerbations of COPD. |
[44] |
|
C57BL/6 mice |
PM2.5 and carbon black |
Inhalation |
2 h/day for 5 days/week for a period of 41 days of 4.5 mg m-3 |
In the experimental design of this study, the preparation of PM2.5 and carbon black exposure sources was standardized, with well-defined concentration and temporal gradients. The combination of cellular and animal experiments, multi-parameter detection, and antibody-based intervention for mechanistic validation contributed to methodologically reliable experimental quality. However, the study lacks standardized assessment of bias risk and did not address covariate adjustment. |
Inhalation of carbon black/PM2.5 induces airway smooth muscle remodeling in vivo. |
[48] |
|
C57BL/6 mice |
PM2.5 |
Intratracheal instillation |
PM2.5 at 100 μg/20 μL concentration, twice a day for 28 consecutive days |
In the experimental design of this study, the preparation of the PM2.5 exposure source was standardized. Animal and cellular experiments validated the involvement of the Wnt5a/β-catenin pathway mechanism through concentration gradient exposure and intervention with the Wnt5a antagonist BOX5. Multi-parameter detection ensured the reliability of the results. However, the study lacks a standardized assessment of bias risk and did not address covariate adjustment in clinical samples. |
PM2.5 exposure elevates α-smooth muscle actin (α-SMA) expression and thickens the basement membrane in murine lung tissue, concurrently with enhanced activation of the Wnt5a/β-catenin signaling pathway. These alterations suggest PM2.5-induced airway remodeling and emphysematous changes. |
[66] |
|
C57BL/6 mice |
PM2.5 and CS |
Intratracheal instillation |
5 weeks CS and PM2.5 at 50 μg/30 μL concentration, twice a day for 28 consecutive days |
In the experimental design of this study, the preparation of PM2.5 and CSE exposure sources were standardized. The clinical research component enrolled patients with COPD and incorporated stratification by smoking status. For animal and cellular experiments, the involvement of the NLRP3/caspase-1 pathway mechanism was validated through interventions using inhibitors and siRNA. Multi-parameter detection ensured the reliability of the results. However, the study lacks a standardized assessment of bias risk, and the clinical analysis did not adjust for covariates such as age and sex. |
Co-exposure to CS and PM significantly elevates cytokine levels in BALF of mice. This exposure simultaneously synergizes to induce cellular apoptosis through the NLRP3/caspase-1 pathway. |
[46] |
Research using healthy human volunteers and primary cells has also provided compelling evidence regarding the adverse respiratory effects associated with PM2.5 exposure. Specifically, primary human nasal epithelial cells exposed to PM2.5 exhibited significantly compromised epithelial integrity, as indicated by reduced expression of tight junction protein, enhanced cellular permeability, and elevated secretion of pro-inflammatory cytokines [36]. These findings suggest that PM2.5-induced epithelial barrier dysfunction may serve as a critical pathogenic mechanism that heightens susceptibility to inflammatory respiratory disorders, such as rhinitis and chronic rhinosinusitis. Consistently, a notable investigation involving 32 volunteers exposed to ambient PM for just 5 hours documented measurable increases in nasal and bronchial inflammation markers [37]. This acute response pattern aligns with findings from animal models, collectively underscoring the immediate pro-inflammatory potential of PM2.5 exposure on the respiratory system.
Beyond the initial innate immune response, PM2.5 exposure has been shown to disrupt adaptive immunity, particularly by influencing the polarization of T helper cells. Experimental evidence suggests that PM2.5 promotes the differentiation of Th1 and Th17 while suppressing the function of regulatory T cells (Treg), leading to an imbalance that favors persistent inflammation. Th17 cells, through the release of their signature cytokine IL-17, stimulate epithelial cells and fibroblasts to produce additional pro-inflammatory mediators and matrix metalloproteinases (MMPs), thereby accelerating airway remodeling [38]. Furthermore, recent studies utilizing human bronchial epithelial cells (HBECs) have revealed that PM2.5 activates the non-canonical Wnt5a/Ror2 signaling pathway, which not only amplifies the production of inflammatory cytokines but also contributes to epithelial-mesenchymal transition (EMT), a critical process in airway wall thickening and fibrosis [39].
|
Cell lines |
Exposure materials |
Dose and exposure period |
Research Quality Assessment |
Major effects |
References |
|
MHS cells and THP-1 cells |
PM2.5 |
40 μg/mL for 24h |
- |
Exposure to PM2.5elevated IL-4, STAT6, and MMP12 levels in MHS and THP-1 cells while reducing E-cadherin protein expression. PM2.5 impairs the alveolar epithelial barrier by promoting MMP12 expression in macrophages. |
[40] |
|
Beas-2B cells |
CSE and PM2.5 |
200 μg/ml PM2.5 and 2.5% CSE |
- |
Co-exposure to PM2.5 and cigarette smoke elevated LC3-II and ATG3 expression while reducing p62 and mTOR levels in Beas-2b cells in a PM2.5 dose-dependent manner, indicating autophagic activation. |
[44] |
|
16HBE cells |
Biomass-related PM2.5 (BRPM) |
40 mg/mL for 24h and 48h |
The experimental design in this study was rigorous, employing a combination of cellular and animal models, treatment at multiple concentration levels and time points, and diverse detection methods. The involvement of the underlying mechanism was further validated using the mitochondria-targeting peptide SS-31. Results demonstrated dose- and time-dependent effects, corroborated in primary cells. Collectively, these aspects contribute to methodologically higher experimental quality. |
Exposure to BRPM reduced cell viability and enhanced proinflammatory responses in 16HBE human bronchial epithelial cells. This exposure simultaneously induced intracellular ROS and mitochondrial ROS (mtROS) generation, concomitant with loss of mitochondrial membrane potential and decreased ATP levels, suggesting mitochondrial dysfunction. |
[43] |
|
Human ASMCs |
Carbon Black and PM2.5 |
25 μg/mL for 24h |
- |
Inhalation of either PM2.5 or carbon black leads to thickening of airway smooth muscle bundles, collagen deposition, and elevated expression of alpha-smooth muscle actin (α-SMA), inducing airway remodeling. |
[48] |
|
HSAEpCs and BEAS-2B |
Diesel PM2.5 |
0–200 μg/mL PM for 24h |
The experimental design in this study is relatively systematic, integrating clinical sample analysis with cellular experiments and employing diverse detection methods. The regulatory mechanism of lnc-IL7R and EZH2 on p21 was validated through overexpression/knockdown experiments. The analysis demonstrated a correlation between PM2.5 exposure and clinical indicators, supported by statistical evidence and relatively comprehensive mechanistic validation. Collectively, these aspects contribute to methodologically higher experimental quality. |
Lnc-IL7R expression was negatively correlated with PM exposure, and PM-mediated cellular senescence genes (such as p21) were regulated by lnc-IL7R. |
[45] |
|
16HBEC |
PM2.5 |
5-20 μg/mL PM2.5 for 3-24h |
The experimental design in this study is sound, integrating animal models with cellular experiments and employing multiple methodologies. The regulatory role of the Wnt5a/Ror2 pathway in PM2.5-induced secretion of inflammatory mediators was validated through siRNA knockdown and intervention with the antagonist BOX5. Concentration and time-dependent effects were clearly established, yielding reliable results indicative of good experimental quality. |
PM2.5 induces upregulation of Wnt5a, which subsequently elevates Ror2 expression. This is accompanied by positive regulation of NF-κB activity, thereby promoting the production of IL-1β, IL-6, and IL-8 in HBECs. |
[39] |
|
BEAS-2B |
CSE and PM2.5 |
20 μg/ml PM2.5 and 3% CES for 24h |
- |
CSE synergistically induces cellular apoptosis through the NLRP3/caspase-1 pathway. |
[46] |
|
ASMC |
PM2.5 |
3 μg/mL for 24h |
The experimental design in this study is relatively comprehensive, integrating clinical samples, animal models, and cellular experiments while employing diverse detection methods. The involvement of the Wnt5a/JNK/NF-κB pathway in PM2.5-induced pulmonary inflammation and fibrosis was validated through interventions including siRNA knockdown, the antagonist BOX5, and the inhibitor SP600125. Concentration-dependent effects and the pathway mechanism were clearly delineated, yielding reliable results indicative of methodologically robust experimental quality. |
PM2.5 elevates Wnt5a production, thereby triggering phosphorylation of JNK and augmenting phosphorylation of NF-κB. This ultimately promotes the generation of inflammatory cytokines and ASMCs. |
[32] |
As PM2.5-induced lung injury progresses, a shift in macrophage polarization becomes evident. In early stages, M1 macrophages predominate, releasing pro-inflammatory cytokines that intensify tissue damage. In contrast, in chronic PM2.5 exposure models, there is a marked transition to M2 macrophages, which facilitate fibrotic changes via the IL-4/STAT6 signaling axis. This pathway upregulates matrix metalloproteinase-12 (MMP12), an enzyme that degrades elastin and other extracellular matrix (ECM) components, leading to the loss of alveolar structure and progressive airway destruction [40]. The resulting ECM remodeling, coupled with ongoing epithelial damage, establishes a self-perpetuating cycle of inflammation and tissue repair that characterizes advanced COPD.
Effect of PM2.5 on oxidative stress
Oxidative stress, characterized by an imbalance where increased levels of oxidants overwhelm the body’s intrinsic antioxidant defense mechanisms, plays a significant role in the pathogenesis of numerous chronic respiratory diseases. Its contribution to the development and progression of COPD resulting from PM2.5 exposure has been very well established.
PM2.5 exposure notably disrupts the pulmonary antioxidant defense system, as demonstrated by significant alterations in key oxidative stress biomarkers. Studies consistently report a depletion of reduced glutathione (GSH), an increase in oxidized glutathione (GSSG) levels, and a subsequent decrease in the GSH/GSSG ratio following PM2.5 exposure [41]. This critical redox couple serves as the primary cellular antioxidant buffer system, and its perturbation signifies severe oxidative stress. Additionally, PM2.5 suppresses the activity of superoxide dismutase (SOD), the first-line enzymatic defense against superoxide radicals, while elevating levels of malondialdehyde (MDA), a reliable marker of lipid peroxidation. These interconnected changes foster a pro-oxidant microenvironment that predisposes pulmonary tissues susceptible to oxidative damage.
The excessive ROS production triggered by PM2.5 initiates a cascade of pro-inflammatory signaling events. In both BALB/c murine models and human bronchial epithelial cells (BEAS-2B), PM2.5-induced ROS have been shown to upregulate the expression of intercellular adhesion molecule-1 (ICAM-1) via the activation of the IL-6/AKT/STAT3/NF-κB signaling axis [42]. This pathway constitutes a crucial connection between oxidative stress and pulmonary inflammation, as ICAM-1 facilitates leukocyte-endothelial adhesion and subsequent transendothelial migration, which are essential processes in the recruitment and infiltration of inflammatory cells into tissues. The persistent activation of this signaling network contributes to chronic inflammation, a hallmark feature of COPD pathogenesis.
Moreover, mitochondria are sensitive targets of PM2.5, and PM2.5 exposure induces profound mitochondrial dysfunction. Damaged mitochondria produce large amounts of ROS, resulting in intracellular redox imbalance and eventually leading to extensive mitochondrial damage. Detailed in vitro studies using human bronchial cells reveal that PM2.5 disrupts mitochondrial ultrastructure, impairs the electron transport chain function, and elevates mitochondrial-specific ROS (mtROS) production [43]. These changes lead to: (1) compromised oxidative phosphorylation and ATP synthesis; (2) disrupted mitochondrial dynamics, characterized by an imbalance in fusion and fission processes; (3) impaired mitophagy leading to the accumulation of damaged mitochondria; and (4) metabolic reprogramming towards glycolysis [44]. Importantly, dysfunctional mitochondria themselves become substantial sources of ROS, perpetuating a cycle of oxidative stress that exacerbates cellular injury. This mitochondrial-ROS feedback loop is particularly relevant in COPD, where persistent oxidative damage accelerates lung aging and contributes to progressive functional decline.
Effect of PM2.5 on cell death
The pathogenesis of COPD under PM2.5 exposure involves complex cellular processes, notably apoptosis and cellular senescence, which significantly contribute to pulmonary tissue damage and airway remodeling. These processes are mediated through distinct yet interconnected molecular pathways that warrant detailed examination.
Programmed cell death is a critical component of PM2.5-induced lung injury. Extensive research employing various model systems, including samples from COPD patients, human small airway epithelial cells (HSAEpCs), and lung tissues, has identified the p21 protein and the lncIL7R-EZH2 signaling axis as key regulators of PM2.5-induced apoptosis. This pathway operates through H3K27 trimethylation (H3K27me3) histone modifications, which concurrently modulate oxidative stress responses and apoptotic processes [45]. In vitro studies using BEAS-2B cells have demonstrated that PM2.5 exposure diminishes cell viability while upregulating apoptosis-related genes, primarily mediated through the activation of the NLRP3/caspase-1 inflammasome pathway. This activation not only facilitates apoptotic cell death but also intensifies pulmonary inflammation, as evidenced by increased inflammatory cell infiltration and cytokine release in COPD mouse models [46].
The PI3K/AKT/mTOR signaling axis emerges as another critical pathway in the pathogenesis of COPD induced by PM2.5 exposure. This pathway, which serves as a key regulator of autophagy, is notably suppressed upon exposure to PM2.5, resulting in disrupted autophagic flux within alveolar epithelial cells [47]. This disruption leads to impaired cellular homeostasis, creating an environment conducive to apoptotic cell death, which in turn contributes to progressive damage of lung tissue. This mechanism explains the observed association between chronic exposure to PM2.5 and accelerated alveolar destruction in COPD.
Beyond apoptosis, PM2.5 exposure induces cellular senescence, a permanent cell cycle arrest that contributes to the progression of COPD through distinct mechanisms. Experimental evidence demonstrates that PM2.5 triggers senescence in both rat and human airway smooth muscle cells (ASMCs) via activation of the autophagy-dependent GATA4/TRAF6/NF-κB signaling cascade. This pathway ultimately leads to the development of a senescence-associated secretory phenotype (SASP), marked by the sustained secretion of pro-inflammatory cytokines, growth factors, and enzymes involved in matrix remodeling [48]. The SASP not only perpetuates local inflammation but also drives pathological airway remodeling, a hallmark feature of advanced COPD.
Recent Developments in Therapeutic Approaches for PM2.5-induced COPD (Table 4)
Conventional medical therapy
Exacerbations of COPD induced by PM2.5 exposure present a substantial clinical challenge, necessitating advanced therapeutic interventions to address inflammation, oxidative stress, and cellular damage. Recent studies have identified several promising pharmacological agents that target specific molecular pathways implicated in the pathogenesis of PM2.5-induced COPD, thereby offering novel approaches to disease management.
Hydrogen sulfide (H2S), an endogenous gaseous signaling molecule with potent antioxidant properties, has been identified as a potential therapeutic candidate for addressing PM2.5-induced COPD. Experimental studies using C57BL/6 mice and BEAS-2B bronchial epithelial cells have revealed that H2S donors, such as sodium hydrosulfide (NaHS), can mitigate oxidative stress and inflammation by modulating the NLRP3/caspase-1 inflammasome pathway and reducing the expression of IL-1β, IL-18, and MMP-9 [49]. Additionally, H2S enhances Klotho/Parkin-mediated mitophagy, thereby preserving mitochondrial function and reducing cellular senescence in airway epithelial cells, ultimately alleviating PM2.5-induced lung injury. Although the clinical application of H2S-based therapies remains under exploration, preclinical evidence supports its potential efficacy in COPD management [50]. It is noteworthy, however, that while high concentrations of H2S are detrimental to human health, exposure to low concentrations confers cytoprotective effects.
Glycogen synthase kinase-3β (GSK-3β) serves as a pivotal regulator of inflammatory responses, influencing NF-κB, STAT, and β-catenin signaling. In murine models and human bronchial epithelial cells (HBECs) exposed to PM2.5, pharmacological inhibition of GSK-3β has been shown to attenuate pulmonary inflammation by suppressing JNK phosphorylation and NF-κB activation, thereby reducing the release of pro-inflammatory cytokines (e.g., TNF-α, IL-6). This strategy may augment existing anti-inflammatory therapies, such as corticosteroids, by targeting upstream signaling mechanisms that drive COPD exacerbations [51].
Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase (NOS), is elevated in COPD patients and contributes to increased oxidative stress and inflammation. Dimethylarginine dimethylaminohydrolase 1 (DDAH1), the enzyme responsible for the degradation of ADMA, exerts protective effects in PM-exposed mice by reducing p65 phosphorylation and Bax-mediated apoptosis, while simultaneously enhancing antioxidant defenses, such as SOD1 and peroxiredoxin 4 (Prdx4). Therapeutic approaches targeting the enhancement of DDAH1 activity or the reduction of ADMA accumulation may improve vascular and airway function in COPD [52].
In addition, melatonin is a naturally occurring hormone with anti-inflammatory and antioxidant properties, which has shown efficacy in ameliorating PM2.5-induced COPD exacerbations. Studies in BEAS-2B cells and murine models reveal that melatonin inhibits the PERK/eIF2α/ATF4/CHOP endoplasmic reticulum (ER) stress pathway, thereby reducing apoptosis and airway inflammation. Given its favorable safety profile, melatonin may serve as an adjunctive therapy for COPD patients experiencing high exposure to PM2.5 [53].
Although these emerging therapies demonstrate potential, their clinical practice necessitates further validation through randomized controlled trials. Key considerations include: (1) the use of combination therapies that targeting multiple pathways (e.g., Nrf2 activation in conjunction with anti-inflammatory agents); (2) personalized medicine approaches tailored to individual genetic and environmental risk factors; (3) comprehensive long-term safety assessments, especially concerning the chronic use of novel agents like H2S donors and GSK-3β inhibitors. The advancing understanding of the pathogenesis of COPD induced by PM2.5 exposure has facilitated the development of targeted pharmacological interventions aimed at mitigating oxidative stress, inflammation, and cellular apoptosis. Future research should focus on optimizing these therapies for clinical application, particularly among high-risk populations with prolonged exposure to PM2.5.
|
International Nonproprietary Name |
Study subjects |
Mechanism(s) of action |
Clinical efficacy |
Drug toxicity |
Reference |
|
H2S |
C57BL/6 mice, BEAS-2B cells |
Inhibition of PM2.5-induced COPD progression is achieved by suppressing cellular senescence via the Klotho/Parkin-mediated mitophagy signaling pathway. |
Not yet investigated |
The biological effects of H2S exhibit concentration-dependent duality: high concentrations induce acute cytotoxicity or lethality, whereas low concentrations elicit cytoprotective responses. |
[49,50] |
|
GSK-3β |
C57BL/6 mice, 16HBE cells |
Attenuation of PM2.5-induced pulmonary inflammation is affected through inhibition of JNK phosphorylation, blockade of NF-κB activation, and suppression of NF-κB-dependent pro-inflammatory gene expression. |
Not yet investigated |
Not yet identified |
[51] |
|
ADMA |
Ddah1 and DDAH1-transgenic mice, RAW264.7 cells |
Alleviates PM2.5 exposure-induced murine lung injury by inhibiting inflammation and oxidative stress. |
Not yet investigated |
Not yet identified |
[52] |
|
Melatonin |
BALB/c mice, BEAS-2B cells |
Ameliorates PM2.5-induced airway inflammation and apoptosis in COPD mice via PERK/eIF2α/ATF4/CHOP. |
Not yet investigated |
Not yet identified |
[53] |
|
Bufei Yishen Formula |
SD rats, BEAS-2B cells |
Attenuates PM2.5-induced oxidative stress in COPD rats via the miR-155/FOXO3a pathway. |
Has been employed in the clinical treatment of COPD patients. |
Not yet identified |
[56] |
|
APS, CPS |
Balb/c mice, alveolar macrophages (AMs) |
Enhances AM phagocytic function; reduces inflammatory mediators in lung tissue and systemic circulation of COPD mice. |
Not yet investigated |
Not yet identified |
[57] |
|
Curcumin |
Balb/c mice, BEAS-2B cells |
Modulates the PTEN/PI3K/AKT pathway to alleviate PM2.5-induced inflammation and oxidative stress in COPD. |
Not yet investigated |
Not yet identified |
[58] |
|
Lentinan |
Beas-2B cells |
Inhibits the GARP/TGF-β/SMAD pathway to attenuate PM2.5 exposure-induced pulmonary epithelial-mesenchymal transition. |
Not yet investigated |
Not yet identified |
[59] |
Traditional Chinese Medicine (TCM) therapy
Although Western medicine primarily provides symptomatic management of COPD through the use of bronchodilators and anti-inflammatory agents, Traditional Chinese Medicine (TCM) has gained recognition as a complementary approach with considerable therapeutic promise. Recent studies highlight the efficacy of TCM formulations and bioactive compounds in alleviating PM2.5-induced COPD through anti-inflammatory, antioxidant, and immunomodulatory mechanisms. This review summarizes key findings regarding the role of TCM in the prevention and treatment of COPD, with an emphasis on its molecular mechanisms and preclinical evidence.
Previous studies have demonstrated that the Bufei Yishen Formula, a clinically prescribed TCM formulation, exerts notable efficacy in the management of COPD. The combination of its active constituents has also been indicated to suppress macrophage activation and reduce PM2.5-induced pulmonary inflammation [54]. Notably, this ingredient combination exhibits beneficial effects on mitochondrial function by inhibiting mitochondrial oxidative stress through the enhancement of SIRT3-mediated deacetylation of FOXO3 [55]. Moreover, it exerts notable anti-oxidative and anti-inflammatory properties by modulating the miR-155/FOXO3a signaling pathway in the COPD rat model [56]. In addition, various natural polysaccharides derived from medicinal herbs, such as astragalus polysaccharides (APS) and codonopsis polysaccharides (CPS), have also been found to offer protective effects against PM2.5-induced COPD. In murine models of COPD, APS, and CPS significantly reduced systemic and pulmonary inflammation by inhibiting the hyperactivation of alveolar macrophages. Furthermore, these polysaccharides have also been shown to mitigate oxidative stress and improve lung function, suggesting their potential as adjunctive therapies for COPD sufferers who are exposed to environmental pollutants [57].
Recently, Curcumin, the principal bioactive compound derived from Curcuma longa, has garnered significant attention due to its extensive pharmacological properties, particularly its anti-inflammatory and antioxidant effects. In both in vivo and in vitro studies, curcumin was shown to counteract PM2.5-induced inflammation and oxidative stress by regulating the PTEN/PI3K/AKT signaling pathway. This modulation not only decreased the release of inflammatory cytokines but also protected against alveolar damage, highlighting curcumin’s potential role in the management of COPD [58]. Lentinan, another polysaccharide extracted from Lentinula edodes, demonstrates strong immunomodulatory and anti-inflammatory properties. Previous studies have revealed that lentinan suppresses PM2.5-induced pulmonary inflammation by inhibiting the GARP/TGF-β/SMAD pathway, thereby reducing EMT and fibrosis. Furthermore, lentinan exhibits metal-chelating effects, neutralizing toxic heavy metals present in PM2.5 and offering additional cytoprotection [59].
Taken together, existing evidence highlights the therapeutic potential of TCM in addressing PM2.5-induced COPD through mechanisms that include the inhibition of oxidative stress, suppression of inflammation, and immunomodulation. Nonetheless, further investigation is required to clarify the specific molecular pathways involved and to optimize clinical applications. Future research should prioritize the standardization of TCM formulations, the execution of large-scale clinical trials, and the exploration of synergistic effects with conventional therapies. Integrating TCM into COPD management could facilitate the development of a more comprehensive and personalized treatment strategy to effectively combat this condition.
Conclusion
In this review, we summarized the existing literature on the cellular and molecular mechanisms linking PM2.5 exposure to the development of COPD, and assessed advancements in innovative therapeutic agents for COPD within both contemporary and traditional medical frameworks. Given the involvement of inflammatory responses, immune dysregulation, oxidative stress, mitochondrial dysfunction, apoptosis, and cellular senescence during COPD progression, alongside the documented influence of PM2.5 on these pathways, it is plausible that PM2.5 contributes to COPD pathogenesis through the concurrent activation of multiple pathways (Figure 1).
Figure 1. Effects of particulate matter (PM) on inflammation, oxidative stress, cell apoptosis, and airway remodeling in lungs.
However, the heterogeneous composition of PM2.5, encompassing metals, organic compounds, and microbial elements, complicates mechanistic studies, and the toxicological contributions of individual components remain ambiguous. Furthermore, the impact of PM2.5 on COPD and the corresponding therapeutic management have been elucidated mostly based on various in vitro and animal studies; nonetheless, large-scale randomized controlled trials (RCTs) are imperative to confirm the clinical benefits. Furthermore, during the development of novel therapeutics, comprehensive assessment of drug toxicity profiles is imperative, necessitating extensive preclinical validation to ensure human safety. PM2.5, as an environmental risk factor for sustained population exposure, has garnered significant academic interest. Moreover, the influence of other contaminants on respiratory disorders is similarly significant and must not be disregarded. An urgent necessity exists to enhance and strengthen pertinent research to clarify their pathogenic mechanisms and investigate effective prevention and control techniques. Additionally, future research should focus on biomarker-guided therapies, incorporating omics technologies to identify patient subgroups that would derive the greatest benefit from TCM interventions. This might unveil novel preventive/therapeutic strategies for mitigating PM2.5-induced COPD and associated exacerbations. Simultaneously, systematic interventions targeting pollution sources are essential, which include the stringent enforcement of air pollution prevention regulations, enhanced control of industrial emissions, the clean retrofitting of mobile sources, and the optimization of the energy structure to achieve synergistic reductions in multi-pollutant emissions.
Consent for Publication
Not Applicable.
Availability of Data and Materials
All data analyzed during this study are included in this published article.
Competing Interests
The authors declare that they have no competing interests.
Funding
This work was financially supported by the National Natural Science Fund of China [82374263].
Authors' Contributions
Yangzi Dong: Investigation, Methodology, Formal analysis, Writing – original draft. Huiyu Yue: Methodology, Writing – original draft. Mengyao Hu: Methodology, Formal analysis. Yingying Li: Methodology, Formal analysis. Tiantian Liu: Funding acquisition. Jing Wang: Conceptualization, Supervision, Writing – review & editing.
References
2. Wang Z, Lin J, Liang L, Huang F, Yao X, Peng K, et al. Global, regional, and national burden of chronic obstructive pulmonary disease and its attributable risk factors from 1990 to 2021: an analysis for the Global Burden of Disease Study 2021. Respir Res. 2025 Jan 2;26(1):2.
3. Pando-Sandoval A, Ruano-Ravina A, Candal-Pedreira C, Rodríguez-García C, Represas-Represas C, Golpe R, et al. Risk factors for chronic obstructive pulmonary disease in never-smokers: A systematic review. Clin Respir J. 2022 Apr;16(4):261–75.
4. Manisalidis I, Stavropoulou E, Stavropoulos A, Bezirtzoglou E. Environmental and Health Impacts of Air Pollution: A Review. Front Public Health. 2020 Feb 20;8:14.
5. Gong FH, Liu L, Wang X, Xiang Q, Yi X, Jiang DS. Ferroptosis induced by environmental pollutants and its health implications. Cell Death Discov. 2025 Jan 24;11(1):20.
6. Pun VC, Kazemiparkouhi F, Manjourides J, Suh HH. Long-Term PM2.5 Exposure and Respiratory, Cancer, and Cardiovascular Mortality in Older US Adults. Am J Epidemiol. 2017 Oct 15;186(8):961–69.
7. Chen CH, Wu CD, Chiang HC, Chu D, Lee KY, Lin WY, et al. The effects of fine and coarse particulate matter on lung function among the elderly. Sci Rep. 2019 Oct 15;9(1):14790.
8. Mukherjee A, Agrawal M. A Global Perspective of Fine Particulate Matter Pollution and Its Health Effects. Rev Environ Contam Toxicol. 2018;244:5–51.
9. Ishihara Y, Tanaka M, Nezu N, Ishihara N, Oguro A, Vogel CFA. Pathways to the Brain: Impact of Fine Particulate Matter Components on the Central Nervous System. Antioxidants (Basel). 2025 Jun 14;14(6):730.
10. Thomas RJ. Particle size and pathogenicity in the respiratory tract. Virulence. 2013 Nov 15;4(8):847–58.
11. Li R, Zhao X, Liu Y, Xu X. [Progress in numerical simulation and experimental study on inhalable particles deposition in human respiratory system]. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi. 2017 Aug 25;34(4):637–42.
12. VoPham T, Kim NJ, Berry K, Mendoza JA, Kaufman JD, Ioannou GN. PM2.5 air pollution exposure and nonalcoholic fatty liver disease in the Nationwide Inpatient Sample. Environ Res. 2022 Oct;213:113611.
13. Leonetti A, Peansukwech U, Charnnarong J, Cha'on U, Suttiprapa S, Anutrakulchai S. Effects of particulate matter (PM2.5) concentration and components on mortality in chronic kidney disease patients: a nationwide spatial-temporal analysis. Sci Rep. 2024 Jul 22;14(1):16810.
14. Alexeeff SE, Deosaransingh K, Van Den Eeden S, Schwartz J, Liao NS, Sidney S. Association of Long-term Exposure to Particulate Air Pollution With Cardiovascular Events in California. JAMA Netw Open. 2023 Feb 1;6(2):e230561.
15. Han Y, Yu Z, Chen Y, Guo X, Liu Y, Zhang H, et al. PM2.5 induces developmental neurotoxicity in cortical organoids. Environ Pollut. 2024 Nov 15;361:124913.
16. Kwon HS, Ryu MH, Carlsten C. Ultrafine particles: unique physicochemical properties relevant to health and disease. Exp Mol Med. 2020 Mar;52(3):318–28.
17. Kuang B, Zhang F, Shen J, Shen Y, Qu F, Jin L, et al. Chemical characterization, formation mechanisms and source apportionment of PM2.5 in north Zhejiang Province: The importance of secondary formation and vehicle emission. Sci Total Environ. 2022 Dec 10;851(Pt 2):158206.
18. Park J, Kim H, Kim Y, Heo J, Kim SW, Jeon K, et al. Source apportionment of PM2.5 in Seoul, South Korea and Beijing, China using dispersion normalized PMF. Sci Total Environ. 2022 Aug 10;833:155056.
19. Paisi N, Kushta J, Pozzer A, Violaris A, Lelieveld J. Health effects of carbonaceous PM2.5 compounds from residential fuel combustion and road transport in Europe. Sci Rep. 2024 Jan 17;14(1):1530.
20. Park SY, Jeon JI, Jung JY, Yoon SW, Kwon J, Lee CM. PM2. 5 and heavy metals in urban and agro-industrial areas: health risk assessment considerations. Asian Journal of Atmospheric Environment. 2024 Jul 26;18(1):16.
21. Hrdina AIH, Kohale IN, Kaushal S, Kelly J, Selin NE, Engelward BP, et al. The Parallel Transformations of Polycyclic Aromatic Hydrocarbons in the Body and in the Atmosphere. Environ Health Perspect. 2022 Feb;130(2):25004.
22. Liang L, Cai Y, Barratt B, Lyu B, Chan Q, Hansell AL, et al. Associations between daily air quality and hospitalisations for acute exacerbation of chronic obstructive pulmonary disease in Beijing, 2013-17: an ecological analysis. Lancet Planet Health. 2019 Jun;3(6):e270–9.
23. Li M, Hanxiang C, Na Z, Ping Z, Shengnan D, Taihua W, et al. Burden of COPD in China and the global from 1990 to 2019: a systematic analysis for the Global Burden of Disease Study 2019. BMJ Open Respir Res. 2023 Jul;10(1):e001698.
24. Yang X, Zhang T, Zhang Y, Chen H, Sang S. Global burden of COPD attributable to ambient PM2.5 in 204 countries and territories, 1990 to 2019: A systematic analysis for the Global Burden of Disease Study 2019. Sci Total Environ. 2021 Nov 20;796:148819.
25. Wang Y, Shi L, Lee M, Liu P, Di Q, Zanobetti A, Schwartz JD. Long-term Exposure to PM2.5 and Mortality Among Older Adults in the Southeastern US. Epidemiology. 2017 Mar;28(2):207–14.
26. Zhu RX, Nie XH, Chen YH, Chen J, Wu SW, Zhao LH. Relationship Between Particulate Matter (PM2.5) and Hospitalizations and Mortality of Chronic Obstructive Pulmonary Disease Patients: A Meta-Analysis. Am J Med Sci. 2020 Jun;359(6):354–64.
27. Cui A, Shao Y, Wang Y, Wang H, Song J, Liu C, Shen N, Yang Z, Zhang W, Luo T, Wang C. Clinicopathological relationship between PM2.5 exposure and acute exacerbations of chronic obstructive pulmonary disease. Ecotoxicol Environ Saf. 2025 Feb;291:117846.
28. WHO global air quality guidelines: Particulate matter (PM2.5 and PM10), ozone, nitrogen dioxide, sulfur dioxide and carbon monoxide [Internet]. Geneva: World Health Organization; 2021.
29. Chi R, Chen C, Li H, Pan L, Zhao B, Deng F, et al. Different health effects of indoor- and outdoor-originated PM2.5 on cardiopulmonary function in COPD patients and healthy elderly adults. Indoor Air. 2019 Mar;29(2):192–201.
30. Liu X, Zhou H, Yi X, Zhang X, Lu Y, Zhou W, et al. Decomposition analysis of lung cancer and COPD mortality attributable to ambient PM2.5 in China (1990-2021). Asia Pac J Oncol Nurs. 2025 Jan 7;12:100653.
31. Xing M, Cui F, Zheng L, Ma Y, Wang J, Tang L, Chen N, Zhao X, Tian Y, Su B. Association of fine particulate matter constituents with chronic obstructive pulmonary disease and the effect modification of genetic susceptibility. npj Climate and Atmospheric Science. 2025 Mar 5;8(1):89.
32. Zou W, Liu S, Ye D, Bai G, Guo M, Sun R, et al. PM2.5 induces lung inflammation and fibrosis via airway smooth muscle cell expression of the Wnt5a/JNK pathway. J Thorac Dis. 2023 Nov 30;15(11):6094–105.
33. Piao CH, Fan Y, Nguyen TV, Shin HS, Kim HT, Song CH, et al. PM2.5 Exacerbates Oxidative Stress and Inflammatory Response through the Nrf2/NF-κB Signaling Pathway in OVA-Induced Allergic Rhinitis Mouse Model. Int J Mol Sci. 2021 Jul 29;22(15):8173.
34. Wang J, Li Y, Zhao P, Tian Y, Liu X, He H, et al. Exposure to Air Pollution Exacerbates Inflammation in Rats with Preexisting COPD. Mediators Inflamm. 2020 May 8;2020:4260204.
35. Fang ZF, Wang ZN, Chen Z, Peng Y, Fu Y, Yang Y, et al. Fine particulate matter contributes to COPD-like pathophysiology: experimental evidence from rats exposed to diesel exhaust particles. Respir Res. 2024 Jan 4;25(1):14.
36. Yuan J, Liao Z, Zhu X, Zhu Y, Wu S, Guo L, et al. PM2.5 exacerbates nasal epithelial barrier dysfunction in allergic rhinitis by inducing NLRP3-mediated pyroptosis via the AhR/CYP1A1/ROS axis. J Hazard Mater. 2025 Jul 15;492:138145.
37. Sun B, Song J, Wang Y, Jiang J, An Z, Li J, et al. Associations of short-term PM2.5 exposures with nasal oxidative stress, inflammation and lung function impairment and modification by GSTT1-null genotype: A panel study of the retired adults. Environ Pollut. 2021 Sep 15;285:117215.
38. Gu XY, Chu X, Zeng XL, Bao HR, Liu XJ. Effects of PM2.5 exposure on the Notch signaling pathway and immune imbalance in chronic obstructive pulmonary disease. Environ Pollut. 2017 Jul;226:163–73.
39. Zou W, Wang X, Hong W, He F, Hu J, Sheng Q, et al. PM2.5 Induces the Expression of Inflammatory Cytokines via the Wnt5a/Ror2 Pathway in Human Bronchial Epithelial Cells. Int J Chron Obstruct Pulmon Dis. 2020 Oct 23;15:2653–62
40. Guo X, Yang S, Zhu H, Liu F, Li K, Li G, et al. Involvement of M2 macrophages polarization in PM2.5-induced COPD by upregulating MMP12 via IL4/STAT6 pathway. Ecotoxicol Environ Saf. 2024 Sep 15;283:116793.
41. Ding S, Jiang J, Li Y. Quercetin alleviates PM2.5-induced chronic lung injury in mice by targeting ferroptosis. PeerJ. 2024 Jan 2;12:e16703.
42. Hou T, Zhu L, Wang Y, Peng L. Oxidative stress is the pivot for PM2.5-induced lung injury. Food Chem Toxicol. 2024 Feb;184:114362.
43. Gao M, Liang C, Hong W, Yu X, Zhou Y, Sun R, et al. Biomass-related PM2.5 induces mitochondrial fragmentation and dysfunction in human airway epithelial cells. Environ Pollut. 2022 Jan 1;292(Pt B):118464.
44. Fan X, Dong T, Yan K, Ci X, Peng L. PM2.5 increases susceptibility to acute exacerbation of COPD via NOX4/Nrf2 redox imbalance-mediated mitophagy. Redox Biol. 2023 Feb;59:102587.
45. Lee KY, Ho SC, Sun WL, Feng PH, Lin CW, Chen KY, et al. Lnc-IL7R alleviates PM2.5-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol Toxicol. 2022 Dec;38(6):1097–120.
46. Chung C, Park SY, Huh JY, Kim NH, Shon C, Oh EY, et al. Fine particulate matter aggravates smoking induced lung injury via NLRP3/caspase-1 pathway in COPD. J Inflamm (Lond). 2024 Apr 24;21(1):13.
47. Zhang F, Ma H, Wang ZL, Li WH, Liu H, Zhao YX. The PI3K/AKT/mTOR pathway regulates autophagy to induce apoptosis of alveolar epithelial cells in chronic obstructive pulmonary disease caused by PM2.5 particulate matter. J Int Med Res. 2020 Jul;48(7):300060520927919.
48. Cheng PP, Yu F, Chen SJ, Feng X, Jia ZH, Hu SH, et al. PM2.5 exposure-induced senescence-associated secretory phenotype in airway smooth muscle cells contributes to airway remodeling. Environ Pollut. 2024 Apr 15;347:123674.
49. Wang Y, Liao S, Pan Z, Jiang S, Fan J, Yu Set al. Hydrogen sulfide alleviates particulate matter-induced emphysema and airway inflammation by suppressing ferroptosis. Free Radic Biol Med. 2022 Jun;186:1–16.
50. Wang Y, Han G, Yang J, Xue L, Chen Y. Hydrogen sulfide attenuates PM2.5-induced COPD by inhibiting cellular senescence via the Klotho/Parkin-mediated mitophagy signaling pathway. Ecotoxicol Environ Saf. 2025 Mar 15;293:117987.
51. Zou W, Ye D, Liu S, Hu J, Zhu T, He F, et al. GSK-3β Inhibitors Attenuate the PM2.5-Induced Inflammatory Response in Bronchial Epithelial Cells. Int J Chron Obstruct Pulmon Dis. 2021 Oct 14;16:2845–56.
52. Gao J, Lei T, Wang H, Luo K, Wang Y, Cui B, et al. Dimethylarginine dimethylaminohydrolase 1 protects PM2.5 exposure-induced lung injury in mice by repressing inflammation and oxidative stress. Part Fibre Toxicol. 2022 Oct 14;19(1):64.
53. Shi M, Liu K, Li X, Zeng XL, Liu XJ. Melatonin ameliorates PM2.5-induced airway inflammation and apoptosis by PERK/eIF2α/ATF4/CHOP in chronic obstructive pulmonary disease mice. Toxicol Appl Pharmacol. 2025 Jun;499:117314.
54. Wang J, He W, Yue H, Zhao P, Li J. Effective-components combination alleviates PM2.5-induced inflammation by evoking macrophage autophagy in COPD. J Ethnopharmacol. 2024 Mar 1;321:117537.
55. Wang J, Yue H, Dong Y, Liu T, Li J. Effective compound combination of Bufei Yishen formula ameliorates PM2.5-induced COPD by inhibiting mitochondrial oxidative stress through SIRT3-mediated FOXO3 deacetylation. Phytomedicine. 2025 May;140:156568.
56. Li J, Wang J, Li Y, Zhao P, Tian Y, Liu X, et al. Effective-component compatibility of Bufei Yishen formula protects COPD rats against PM2.5-induced oxidative stress via miR-155/FOXO3a pathway. Ecotoxicol Environ Saf. 2021 Nov 10;228:112918.
57. Chu X, Liu XJ, Qiu JM, Zeng XL, Bao HR, Shu J. Effects of Astragalus and Codonopsis pilosula polysaccharides on alveolar macrophage phagocytosis and inflammation in chronic obstructive pulmonary disease mice exposed to PM2.5. Environ Toxicol Pharmacol. 2016 Dec;48:76–84.
58. 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 Jul;201:115460.
59. Wang Z, Xu S, Bian B, Hu Z, Wu F, Zhao S, et al. Lentinan Alleviated PM2.5 Exposure-Induced Epithelial-Mesenchymal Transition in Pulmonary Epithelial Cells by Inhibiting the GARP/TGF-β/Smad Pathway. Toxics. 2025 Feb 26;13(3):166.
60. Delavar MA, Jahani MA, Sepidarkish M, Alidoost S, Mehdinezhad H, Farhadi Z. Relationship between fine particulate matter (PM2.5) concentration and risk of hospitalization due to chronic obstructive pulmonary disease: a systematic review and meta-analysis. BMC Public Health. 2023 Nov 13;23(1):2229.
61. Huh JY, Hong J, Han DW, Park YJ, Jung J, Lee SW. The Impact of Air Pollutants and Meteorological Factors on Chronic Obstructive Pulmonary Disease Exacerbations: A Nationwide Study. Ann Am Thorac Soc. 2022 Feb;19(2):214–26.
62. Cowan KN, Wyatt LH, Luben TJ, Sacks JD, Ward-Caviness C, Rappazzo KM. Effect measure modification of the association between short-term exposures to PM2.5 and hospitalizations by longs-term PM2.5 exposure among a cohort of people with Chronic Obstructive Pulmonary Disease (COPD) in North Carolina, 2002-2015. Environ Health. 2023 Jun 29;22(1):49.
63. Bourbeau J, Doiron D, Biswas S, Smith BM, Benedetti A, Brook JR, et al. Ambient Air Pollution and Dysanapsis: Associations with Lung Function and Chronic Obstructive Pulmonary Disease in the Canadian Cohort Obstructive Lung Disease Study. Am J Respir Crit Care Med. 2022 Jul 1;206(1):44–55.
64. Liu S, Jørgensen JT, Ljungman P, Pershagen G, Bellander T, Leander K, et al. Long-term exposure to low-level air pollution and incidence of chronic obstructive pulmonary disease: The ELAPSE project. Environ Int. 2021 Jan;146:106267.
65. Bochenek B, Jankowski M, Wieczorek J, Gruszczyńska M, Sekuła P, Jaczewski A, et al. The impact of ambient air pollution and meteorological factors on emergency hospital admissions of COPD patients in Poland (2012-2019). Sci Rep. 2025 Jul 1;15(1):21915.
66. Zou W, Wang X, Sun R, Hu J, Ye D, Bai G, et al. PM2.5 Induces Airway Remodeling in Chronic Obstructive Pulmonary Diseases via the Wnt5a/β-Catenin Pathway. Int J Chron Obstruct Pulmon Dis. 2021 Dec 3;16:3285–95.
