Abstract

Recently, the advent of health risks due to the cytotoxicity of fine particulate matter (FPM) is concerning. Numerous studies have reported abundant data elucidating the FPM-induced cell death pathways. However, several challenges and knowledge gaps are still confronted nowadays. On one hand, the undefined components of FPM (such as heavy metals, polycyclic aromatic hydrocarbons, and pathogens) are all responsible for detrimental effects, thus rendering it difficult to delineate the specific roles of these copollutants. On the other hand, owing to the crosstalk and interplay among different cell death signaling pathways, precisely determining the threats and risks posed by FPM is difficult. Herein, we recapitulate the current knowledge gaps present in the recent studies regarding FPM-induced cell death, and propose future research directions for policy-making to prevent FPM-induced diseases and improve knowledge concerning the adverse outcome pathways and public health risks of FPM.
Keywords: Air pollution, Fine particulate matter, Cytotoxicity, Cell death pathway, Apoptosis, Autophagy, Inflammation, Health risks
Recently, health issues due to air pollution are becoming increasingly prominent. Particulate matter (PM), a major contributor of air pollution, has gained global attention. Several epidemiological studies have correlated PM pollution with various detrimental health issues, including respiratory diseases, cardiovascular diseases, reproductive system disorders, and even neurological disorders.1−4 Although the harmful effects of heavy PM pollution have been recognized, the adverse outcome pathways (AOPs) involved remain controversial. PM exhibits variable compositions depending on the source of generation and atmospheric conditions.5 The complex physicochemical properties of PM render it difficult to elucidate its cytotoxic mechanisms. In addition, PM, especially fine PM (FPM, such as PM2.5) and ultrafine PM, can enter the respiratory tract and lungs and even cross the air–blood barrier, causing long-term toxicity in secondary organs, such as the heart, liver, kidneys, and brain.6 Thus, the cytotoxicity of FPM is inevitable regardless of the exposure pathway and biodistribution patterns, which warrant further investigation.
Cell death is a distinct genetic and biochemical pathway that is essential for nearly all living organisms. It is not only involved in the developmental processes of multicellular organisms but also critical for immune responses against various diseases, such as infections and cellular dysfunction. In recent years, various forms of cell death patterns, such as apoptosis,7 autophagy,8 pyroptosis,9 ferroptosis,10 necroptosis,11 and cuproptosis,12 have been described in detail. To date, numerous studies have focused on the toxic effects and related mechanisms of FPM. Furthermore, cell death pathways induced by engineered nanoparticles have helped understand FPM-induced cytotoxicity.13−15 Although the corresponding body of knowledge is expanding (Table 1), several questions remain unanswered, particularly questions regarding the cell death pathways and related mechanisms induced by FPM. Owing to the complex network of interacting genes implicated in cell death, specific FPM-related cell death pathways need to be elucidated. Consequently, understanding the FPM-induced cell death pathways may provide further clues that can help modulate these pathways to treat related diseases. Herein, we summarized the current stage of understanding regarding FPM-induced cell death pathways and elaborated the existing knowledge gaps. We expect that a deeper understanding of the physiological and mechanistic aspects of FPM-induced cell death signaling will yield further knowledge regarding AOPs and public health risks.
Table 1. Summary of Studies on Cell Death Pathways upon FPM Exposure.
| cell death process | cell death signaling | cell types | ref |
|---|---|---|---|
| apoptosis | bax/bcl-2/caspase-3 | Mouse primary cortical neuron cells | (89) |
| miR-194-3p/caspase-3/DAPK1 | HBEpiCs | (90) | |
| JNK/P53 pathway | Rat cardiomyocyte | (91) | |
| TNF-α signaling pathway | A549 cells | (43) | |
| MAPK 2/STAT1 signaling pathway | A550 cells | (43) | |
| COX-2/PGES/PGE2 inflammatory pathway | MAEC | (45) | |
| p53-bax-caspase pathway | HUVECs | (46) | |
| β2AR/PI3K/Akt pathway | AC16 | (47) | |
| Nrf2 pathway | hESCs | (51) | |
| RIPK1 apoptotic signaling pathway | GC-2spd cells | (52) | |
| ROS-JNK/ERK apoptotic pathway | Rat embryonic cells | (53) | |
| autophagy | AMPK signaling pathway | A549 | (57) |
| NOS2 signaling pathway | BEAS-2B | (93) | |
| ATR-CHEK1/CHK1 inflammatory response axis | BEAS-2B | (60) | |
| ROS-AKT-mTOR axis | HUVECs | (61) | |
| NF-κB/p38/Akt signaling pathways | MAECs | (62) | |
| MyD88-mediated inflammatory pathway | Mouse liver cells | (63) | |
| pyroptosis | ROS-NLRP3-caspase-1 pathway | RAW 264.7 | (69) |
| NLRP3-caspase-1 pathway | Mouse cardiomyocytes | (94) | |
| NLRP3-caspase-1 pathway | BV-2 and HMC-3 microglial cell | (71) | |
| NLRP3-caspase-1 pathway | Mouse brain tissue | (95) | |
| NLRP3-caspase-1 pathway | Mouse microglia | (96) | |
| ferroptosis | iron overload/redox imbalance | EA.hy 926 and HUVECs | (76) |
| ERK/CREB pathway | Neuro-2a (N2A) and SH-SY5Y cells | (78) | |
| necroptosis | ATP synthesis | BEAS-2B | (79) |
Major Toxic Components of Fine Particulate Matter
Due to the emission sources and generation patterns, FPM might exhibit significantly different components. Typically, the cytotoxicity of FPM is attributable to its constituent harmful substances (e.g., heavy metals, polycyclic aromatic hydrocarbons (PAHs), and pathogens) (Figure 1).16,17 These components are deemed to be the determining causes of a range of detrimental effects.
Figure 1.
Proposed primary components that are responsible for the cytotoxicity of various FPM, including PAHs, pathogens, and heavy metals.
The heavy metals on FPM originate from a diverse variety of generation sources, such as fuel combustion, geogenic sources, metallurgy, and resuspended dust, leading to various profiles and amounts.18 For example, in the FPM samples collected from Beijing, China during 2013–2019, Pb, Mn, Cd, Sb, Sr, Ni, and other heavy metals were detected,19 and in the FPM samples extracted in Los Angeles between 2005 and 2018, Al, Ba, Cd, Co, Cr, Cu, Pb, Zn etc. were detected.20 Similar heavy metals were also observed on FPM in other countries such as Japan and Poland.21,22 Notably, some heavy metals on the FPM are known to be highly carcinogenic, such as As, Pb, Cd, and Co, and thus, needed to be carefully incorporated into the health risk assessment of FPM.
Another type of toxic components on FPM is PAHs. Contrary to heavy metals, PAHs might be generated through incomplete combustion processes from biomass burning, coal burning, and vehicle emissions.23 Typical species of PAHs on FPM were variable, including but not limited to acenaphthylene, anthracene, acenaphthene, chrysene, fluorene, fluoranthene, pyrene, phenanthrene, naphthalene, benzo[a]anthracene, benzo[b]fluoroanthene, benzo[k]fluoroanthene, benzo[a]pyrene, benzo[ghi]perylene, dibenzo[a,h]anthracene, and indeno[123-cd]pyrene.24 Mounting studies have pointed out that long-term exposure to PAHs loading on FPM may impair lung function and increase the risk of lung cancer.25 For example, loading with benzo[a]pyrene (B[a]P), a well-known human carcinogen categorized by the International Agency for Research on Cancer (IARC),26 the fine biochar particles would incur severe phospholipid membrane disruption in macrophages and impair the innate immune defenses.27
Apart from the heavy metals and PAHs, pathogen (e.g., fungi, bacteria, and viruses) loadings on the FPM have gained attention. Based on the epidemiological studies showing the positive relationship between FPM pollution and pathogen transmission, many studies have documented the hitchhiking of pathogens on the FPM and the possible health risks.28−30 The FPM-induced damage to the respiratory system would cause inflammation of cells, thereby increasing susceptibility to free pathogens through various mechanisms, such as reactive oxygen species (ROS) generation.31,32 In addition, the coexposure of FPM and pathogens could significantly increase both the transmission of and infection by pathogens. For instance, Liu et al. demonstrated that the coexposure of FPM and Pseudomonas aeruginosa would increase the transmission of P. aeruginosa, which induces structural damage in the upper respiratory tract associated with systematic inflammatory reactions.33
Meanwhile, the attached copollutants, namely, the components, are all responsible for the toxicity of FPM. The different components involve a range of physical and biochemical mechanisms. For example, size is a critical property in determining the cytotoxicity. In addition, the biotransformation of particles in vivo is a critical factor as it considerably alters the physiochemical properties and related cytotoxicity of FPM. Therefore, regarding the overall adverse health outcomes of FPM, increasing efforts should be invested to shed light on the contributions and roles of the complex components and in vivo fate of FPM in causing overall cytotoxicity.
Major Cell Death Pathways Induced by Fine Particulate Matter
Apoptosis
As a major cell death pathway, apoptosis plays an important role in the biological maintenance of homeostasis and has been widely studied.34 Aberrant apoptosis of cells would give rise to cardiovascular disease, immune disease, neurodegenerative diseases and osteoporosis.35−37 Apoptosis could be visualized through multiple characteristics, including shrinkage of the cytoplasm, condensation of nuclei, detachment from the surrounding tissue, and convolution of outlines.7 Two apoptotic pathways have been proposed: intrinsic and extrinsic pathways.38 The intrinsic pathway is controlled by the B-cell lymphoma 2 (bcl-2) family proteins, while the extrinsic pathway involves the members of the tumor necrosis factor (TNF) and resultant deadly signaling cascade, which will occur upon FPM exposure.39
Typically, the apoptotic pathway induced by FPM exposure is regulated by a set of well-defined signaling pathways, and caspase-3 molecule can be regarded as one important marker.7 Generally, FPM exposure could induce apoptosis of in various cell types (Figure 2). For example, Chen et al. found that the collected FPM samples could alter the protein expression of apoptosis-related markers (i.e., BCL2-Associated X (bax) and B-cell lymphoma-2 (bcl-2)) and subsequently activate caspase-3, leading to apoptosis of neurons.40 It has been also uncovered that exposure of real FPM could induce activation of caspase-3 by downregulating miR-194-3p and upregulating death associated protein kinase 1 (DAPK1) expression in human bronchial epithelial cells.41 In addition to the in vitro studies above, Wang et al. found that FPM exposure promoted inflammation and oxidative stress in hyperlipidemic rats, by triggering caspase-3 in cardiomyocytes via the JNK/P53 pathway.42
Figure 2.
Illustration of the mechanisms of apoptosis induced by FPM. Apoptosis induced by FPM can be induced by inflammation and ROS generation. FPM-induced inflammation further activates the bax/bcl-2/caspase-3, TNF-α, and miR-194-3p/caspase-3/DAPK1 signaling pathways and etc. FPM-induced ROS generation triggers pathways, such as the Nrf2 and RIPK1 apoptotic signaling pathways.
Although the outcome (namely, caspase-3 activation) has been clarified, several pathways have been implicated for the FPM exposure (Table 1). One important pathway is mitogen-activated protein kinase (MAPK) induced by inflammation and ROS generation.34 For instance, many studies have found that FPM could sensitize lung epithelial cells to apoptosis via nuclear factor kappa-B (NF-κB), TNF-α, and signal transducer and activator of transcription 1 (STAT1) signaling pathways, leading to the inflammation in the respiratory system.43 This pathway has also been validated after silica nanoparticle (one typical model FPM) exposure to the lung epithelial cells, that the cells would be primed to undergo apoptosis.44 Not limited to the epithelial cells, numerous pathways, such as cyclooxygenase-2 (COX-2)/prostaglandin E synthase (PGES)/prostaglandin E2 (PGE2) inflammatory pathway and p53-bax-caspase pathway, and β2-adrenergic receptor (β2AR)/ phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, were verified in the endothelial cells and cardiomyocytes, corresponding to the cardiology disorders, e.g., plaque progression and/or acute coronary syndromes.45−47 Similar pathways are also reportedly activated in endothelial cells upon exposure to other nanoparticles, such as titanium oxide (TiO2), zinc oxide (ZnO), and copper oxide (CuO), inducing apoptosis.48−50
Remarkably, FPM induced similar apoptosis pathways with metal-based nanoparticles. Since many metal species are found on FPM, the cytotoxicity might be attributed to the metal ion release via cell membrane damage and ROS generation. However, unlike the liquid matrix in vivo, other pathways, including nuclear factor erythroid 2-related factor (Nrf2) pathway, receptor-interacting protein kinase 1 (RIPK1) apoptotic signaling pathway and ROS- c-Jun N-terminal kinases (JNK)/extracellular signal-regulated kinases (ERK) apoptotic pathway, were revealed in various epidermal cells, in response to the dermal exposure of FPM.51−53 For comparison, exposure of nano-ZnO would also induce apoptosis in human keratinocytes through the Nrf2 pathway.54 Thus, it could be deduced that the metal ion release may occur after endocytosis or within the lysosomes.
Since unexpected apoptosis links to structural damages and disorders, even diseases, clarifying FPM-induced cell death pathways may help understand its cytotoxic mechanisms. Thereby, the different signaling pathways occur in various cell types, and these differences appear to be largely dependent on the biotransformation of FPM both in vivo and in vitro.
Autophagy
Autophagy is defined as a crucial pathway for cells to retain cellular equilibrium and respond to external stress, as a process where the cellular components or even whole organelles are sequestered into lysosomes for degradation. The lysosomes then digest these substrates and create new cellular structures.8 In general, autophagy is triggered when cells are stressed and is characterized by isolated membranes, autophagic vesicles, and autophagic lysosomes.8 In addition, autophagy has also been reported to be involved in both the adaptive and the innate immune system.8 Although autophagy is a normal process during the cell cycle, the reactions of autophagy may cause metabolic stress, cell death, and even cancer and neurodegenerative diseases.55
Normally, autophagy involves numerous proteins, which can be used as markers, e.g., autophagy related 5 (Atg5), autophagy related 12 (Atg12), Beclin1, and microtubule-associated protein 1 light chain 3 (MAP1LC3) (Figure 3).8 For example, it has been reported that metal-based nanoparticles could mediate autophagy by upregulating these proteins.56 Similar to apoptosis, autophagy could be induced by ROS generation upon FPM exposure in some model cell lines, via adenosine 5′-monophosphate-activated protein kinase (AMPK) signaling and nitric oxide synthase 2 (NOS2) signaling pathways.57,58 These signaling pathways have been also assessed in cytotoxicity studies of nanoparticles, as reflected by the induction of autophagy via inhibition of the mammalian target of rapamycin (mTOR) pathway.59
Figure 3.
Illustration of the mechanisms of autophagy induced by FPM. Autophagy induced by FPM can be induced via ROS generation through diverse signaling, e.g., NF-κB, NOS2 and ART pathways.
In addition to ROS generation, the FPM-induced inflammatory reactions could promote cellular autophagy through ATR-CHEK1/CHK1 inflammatory response axis, ROS-AKT-mTOR axis, NF-κB/p38/Akt signaling pathways and MyD88-mediated inflammatory pathway.60−63 Accordingly, it has been found that silica nanoparticles could induce autophagy in bronchial epithelial cells by inhibiting PI3K/AKT/mTOR expression,64 and ZnO nanoparticles would lead to autophagy in macrophages via the PI3K/mTOR/AKT cascade.49
Apoptotic and autophagic pathways can intercross at various levels. In fact, both cell death pathways could be induced via FPM-induced ROS generation; however, apoptosis is always the preferred pathway. Apoptosis and autophagy are strongly correlated as they share similar stimuli associated with different downstream signaling pathways. In some points of view, autophagy blocks the trigger of apoptosis, and apoptosis-associated caspase activation reversely inhibits the autophagic process. The dialogue between autophagy and apoptosis will significantly affects both cell death pathways as well as immune reactions.65
Pyroptosis
Pyroptosis, also referred to as cellular inflammatory necrosis, is another programmed cell death pathway.66 It is characterized by constant cell growth until the cell membrane ruptures, expelling the contents and inducing severe inflammatory responses. Recently, pyroptosis has received great attention owing to its association with innate immunity and disease, such as neurological disorders, infections and atherosclerosis.66
Cellular pyroptosis has been demonstrated to rely on the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3), NLR family CARD domain containing 4 (NLRC4), and absent in melanoma 2 (AIM2) inflammasomes, and caspase-1 signaling.67 Therefore, most studies on FPM-induced pyroptosis are centered on these pathways (Figure 4). For example. Jia et al. found that FPM could disrupt the alveolar–capillary barrier by upregulating oxidative stress levels, which further induced cellular pyroptosis via caspase-1 signaling pathways.68 Ren et al. reported that FPM could upregulate the ROS-NLRP3-mediated pathway and cause pyroptosis.69 Compared to the previous studies showing that ROS generated by ZnO nanoparticles would activate NF-κB-mediated NLRP3 inflammasomes in A549 cells,70 the pyroptosis induced by FPM appeared to be attributed to the metal ion release. In addition, the activation of NLRP3 and the consequent cellular pyroptosis following FPM exposure have been observed both in vitro (such as BV-2 and HMC-3 microglial cell lines) and in vivo.71−73
Figure 4.
Illustration of the mechanisms of pyroptosis, ferroptosis, and necroptosis induced by FPM. The three cell death patterns are mainly induced by FPM through ROS generation.
Pyroptosis is regarded as an alternative form of apoptosis, which is a proinflammatory state coupled to inflammatory cytokine release in the surrounding tissues. Caspase-1 is a major marker of pyroptosis, which is often accompanied by apoptosis. Thus, it is difficult to differentiate pyroptosis and apoptosis within a single cell, however, pyroptosis is more likely to occur when cells contain high amounts of gasdermin D (GSDMD) protein. Notably, pyroptosis is often observed in the central nervous system and has been correlated with brain disorders. Considering the possible translocation of FPM into the brain, the pyroptosis induced by FPM and the mechanisms involved in the central nervous system should be further explored.
Ferroptosis and Necroptosis
Ferroptosis is a recently defined regulated programmed cell death.74 During ferroptosis, cellular cysteine transport proteins are blocked and glutathione peroxidase (GPX4) is inhibited, resulting in the depletion of intracellular glutathione (GSH).74 Consequently, lipid peroxidation is intensified to extent of cell death, which is also caused by GPX4 enzyme suppression.74 The susceptibility to ferroptosis is closely correlated with the metabolism of amino acids, iron, and polyunsaturated fatty acids, as well as the production of GSH, phospholipids, NADPH, and coenzyme Q10.75
To date, the correlation between FPM and ferroptosis has been rarely reported (Figure 4). For example, Wang and Tang found that FPM could induce ferroptosis in human endothelial cells via inducing iron overload and redox imbalance.76 Remarkably, many studies unveiled that heavy metals, e.g., As, Pb and Cd, could induce imbalances in redox systems, thereby causing ferroptosis.77 In an another study, FPM exposure would lead to ferroptosis in neuronal cells by inhibiting the extracellular regulated protein kinases (ERK)/cAMP-response element binding protein (CREB) pathway.78
Necroptosis is a form of regulated cell death that critically relies on RIPK3 and mixed lineage kinase domain-like (MLKL).11 Necroptosis occurs when cells are exposed by extreme threatens, such as high-dose of toxicants (Figure 4). Duan et al. found that high-dose FPM would induce BEAS-2B cell necroptosis by downregulating the enzymes responsible for adenosine triphosphate (ATP) synthesis.79 Similar to ferroptosis, necroptosis could be induced by heavy metals, however, studies regarding FPM-induced necroptosis are scarcely known.80,81
Collectively, both ferroptosis and necroptosis can be triggered by ROS stimuli, which might be generated by copollutants or the particle itself. However, these pathways induced by FPM warrant further study for better understanding of cytotoxicity mechanisms. For instance, it has been proposed that there is crosstalk between ferroptosis and apoptosis upon polystyrene microplastics exposure; however, the complex relationships between the two pathways need to be elucidated.82
Conclusions and Perspectives
As air pollution is becoming a serious problem globally, the health risks posed by FPM are gradually coming into focus. Epidemiological and toxicological studies have shown that FPM is associated with various diseases and disorders, however, the molecular mechanisms underlying these associations remain unclear. One reason for this is that the cell death pathways (i.e., autophagy, apoptosis, pyroptosis, ferroptosis, and necroptosis) are complex, which indicates an intertwined crosstalk and correlation with each other. For example, it has been demonstrated that FPM would induce apoptosis through different pathways, such as the NF-κB/STAT1 signaling pathway. In another occasion, FPM would also trigger cell autophagy through the NF-κB pathway. Although the two pathways involve NF-κB, the downstream pathways and outcomes are starkly different.
Although the findings of the studies regarding the cytotoxicity of engineered nanoparticles with controllable physiochemical properties have helped understand the underlying molecular mechanisms of FPM, the complex components of FPM, namely, heavy metals, PAHs, and even pathogens, would lead to more complicated cytotoxicity profiles. Apparently, the different pathways summarized here are largely attributable to the characteristics of collected FPM. For example, Cd-induced cytotoxicity is often considered to be because of ROS generation, and whether Cd loading on FPM contributes to inducing cell death needs to be further studied. Some essential trace elements, such as Fe, should be also considered in inducing inflammatory reactions.83 It seems that ROS generation is a major trigger for different cell death pathways, especially programmed cell death. Many stresses, such as exotic invaders (namely fine particles), pathogens, heavy metals and PAHs, which disrupt the cellular homeostasis, essentially enhance the production of ROS (Figure 5). Thus, whether the heavy metals and PAHs attached to or released from FPM should be clarified, that is, understanding the bioaccessibility of the copollutants on FPM.
Figure 5.
Proposed schematic of cell death pathways caused by ROS generation upon FPM exposure. Overview of regulated cell death pathways due to ROS, highlighting FPM as a stimulus with different exposure doses.
Another important factor determined the cytotoxicity pathways is the cell type. According to previous studies, inhaled particles could pass the air–blood barrier and then translocate to other organs,84e.g., heart,85 brain, and even knee joint.82,86,87 Thus, the target cells of FPM vary in diverse studies and the reported mechanisms are different. For example, the FPM could sensitize lung epithelial cells to apoptosis via the NF-κB pathway and induce apoptosis on endothelial cells through the COX-2/PGES/PGE2 pathway. In addition, the cytotoxicity profiles of FPM highly depend on the uptake of different cells. For instance, macrophages would undergo autophagy after endocytosis of carbon-based FPM, while epithelial cells would be affected by the pollutants loading on FPM without apparent endocytosis of particles.27,88 Collectively, the whole picture in the cytotoxicity profiles of FPM should be further teased out.
Therefore, the current knowledge gaps on the cell death pathway caused by FPM should be filled by future studies. The following points are a few recommendations for future studies.
-
(i)
Although recent studies have comprehensively delineated certain cell death pathways, such as apoptosis and autophagy, novel FPM-induced cell death pathways (e.g., ferroptosis and cuproptosis) warrant further exploration. In addition, owing to the complex relationships between these cell death pathways, a more specific and detailed network and crosstalk among the different pathways should be pictured after FPM exposure.
-
(ii)
It should be noted that the pathways caused by FPM appeared differently in various organs or tissues. For instance, the death pathways induced by FPM are different in various cells, such as epithelial and endothelial cells (Table 1). Considering the translocation of inhaled FPM to secondary organs (i.e., direct toxicity) and secretion of cytokines upon FPM exposure (i.e., indirect toxicity), the different cell death patterns and signaling pathways on various organs should be differentiated clearly.
-
(iii)
Owing to the “Trojan horse” effects of FPM, some trace elements, such as Fe, Cu, and Mn, could be transferred into cells, inducing cell metabolism disorders. For example, a large amount of Fe in cells would induce change of redox potential and further ferroptosis. Thus, the influences on cell metabolism and the resulting cytotoxicity of FPM should be clarified in future studies.
-
(iv)
Mounting data have evidenced the implications of airborne particles in pathogen (e.g., fungi, bacteria, and viruses) transmission and epidemic spreading. The roles of pathogens on FPM should be further investigated. For example, whether the pathogens loaded on FPM would increase their accessibility and invasion is hardly understood so far. Thus, the cell death pathways caused by complex FPM and associated pathogens need to be elucidated.
Acknowledgments
This project was supported by the Ministry of Science and Technology of the People’s Republic of China (grant number: 2021YFE0101500) and the National Natural Science Foundation of China (Grant numbers: 22276211, 22021003 and 22150006).
Author Contributions
CRediT: Yue Wu writing-original draft (supporting).
The authors declare no competing financial interest.
References
- Huang Q.; Hu D.; Wang X.; Chen Y.; Wu Y.; Pan L.; Li H.; Zhang J.; Deng F.; Guo X.; et al. The modification of indoor PM2.5 exposure to chronic obstructive pulmonary disease in Chinese elderly people: A meet-in-metabolite analysis. Environ. Int. 2018, 121, 1243–1252. 10.1016/j.envint.2018.10.046. [DOI] [PubMed] [Google Scholar]
- McGuinn L. A.; Schneider A.; McGarrah R. W.; Ward-Caviness C.; Neas L. M.; Di Q.; Schwartz J.; Hauser E. R.; Kraus W. E.; Cascio W. E.; Diaz-Sanchez D.; Devlin R. B. Association of long-term PM(2.5) exposure with traditional and novel lipid measures related to cardiovascular disease risk. Environ. Int. 2019, 122, 193–200. 10.1016/j.envint.2018.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei Y.; Cao X. N.; Tang X. L.; Shen L. J.; Lin T.; He D. W.; Wu S. D.; Wei G. H. Urban fine particulate matter (PM2.5) exposure destroys blood–testis barrier (BTB) integrity through excessive ROS-mediated autophagy. Toxicology mechanisms methods 2018, 28, 302–319. 10.1080/15376516.2017.1410743. [DOI] [PubMed] [Google Scholar]
- Wang B.-R.; Shi J.-Q.; Ge N.-N.; Ou Z.; Tian Y.-Y.; Jiang T.; Zhou J.-S.; Xu J.; Zhang Y.-D. PM2.5 exposure aggravates oligomeric amyloid beta-induced neuronal injury and promotes NLRP3 inflammasome activation in an in vitro model of Alzheimer’s disease. Journal of neuroinflammation 2018, 15, 1–12. 10.1186/s12974-018-1178-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masalaite A.; Holzinger R.; Ceburnis D.; Remeikis V.; Ulevičius V.; Röckmann T.; Dusek U. Sources and atmospheric processing of size segregated aerosol particles revealed by stable carbon isotope ratios and chemical speciation. Environ. Pollut. 2018, 240, 286–296. 10.1016/j.envpol.2018.04.073. [DOI] [PubMed] [Google Scholar]
- Wang W.; Lin Y.; Yang H.; Ling W.; Liu L.; Zhang W.; Lu D.; Liu Q.; Jiang G. Internal exposure and distribution of airborne fine particles in the human body: Methodology, current understandings, and research Needs. Environ. Sci. Technol. 2022, 56, 6857–6869. 10.1021/acs.est.1c07051. [DOI] [PubMed] [Google Scholar]
- Elmore S. Apoptosis: a review of programmed cell death. Toxicologic pathology 2007, 35, 495–516. 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mizushima N. Autophagy: process and function. Genes development 2007, 21, 2861–2873. 10.1101/gad.1599207. [DOI] [PubMed] [Google Scholar]
- Bergsbaken T.; Fink S. L.; Cookson B. T. Pyroptosis: host cell death and inflammation. Nature Reviews Microbiology 2009, 7, 99–109. 10.1038/nrmicro2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dixon S. J.; Lemberg K. M.; Lamprecht M. R.; Skouta R.; Zaitsev E. M.; Gleason C. E.; Patel D. N.; Bauer A. J.; Cantley A. M.; Yang W. S.; et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. 10.1016/j.cell.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pasparakis M.; Vandenabeele P. Necroptosis and its role in inflammation. Nature 2015, 517, 311–320. 10.1038/nature14191. [DOI] [PubMed] [Google Scholar]
- Tsvetkov P.; Coy S.; Petrova B.; Dreishpoon M.; Verma A.; Abdusamad M.; Rossen J.; Joesch-Cohen L.; Humeidi R.; Spangler R. D.; Eaton J. K.; Frenkel E.; Kocak M.; Corsello S. M.; Lutsenko S.; Kanarek N.; Santagata S.; Golub T. R. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science 2022, 375, 1254–1261. 10.1126/science.abf0529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andón F. T.; Fadeel B. Programmed cell death: molecular mechanisms and implications for safety assessment of nanomaterials. Accounts of chemical research 2013, 46, 733–742. 10.1021/ar300020b. [DOI] [PubMed] [Google Scholar]
- De Stefano D.; Carnuccio R.; Maiuri M. C. Nanomaterials toxicity and cell death modalities. Journal of Drug Delivery 2012, 2012, 167896. 10.1155/2012/167896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia T.; Zhu Y.; Mu L.; Zhang Z. F.; Liu S. Pulmonary diseases induced by ambient ultrafine and engineered nanoparticles in twenty-first century. National Science Review 2016, 3, 416–429. 10.1093/nsr/nww064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia Y. Y.; Wang Q.; Liu T. Toxicity Research of PM2.5 Compositions In Vitro. International Journal of Environmental Research and Public Health 2017, 14, 232. 10.3390/ijerph14030232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shang Y.; Zhu T.; Lenz A. G.; Frankenberger B.; Tian F.; Chen C.; Stoeger T. Reduced in vitro toxicity of fine particulate matter collected during the 2008 Summer Olympic Games in Beijing: the roles of chemical and biological components. Toxicology in vitro: an international journal published in association with BIBRA 2013, 27, 2084–93. 10.1016/j.tiv.2013.08.004. [DOI] [PubMed] [Google Scholar]
- Dias G. M.; Edwards G. C. Differentiating natural and anthropogenic sources of metals to the environment. Human and Ecological Risk Assessment 2003, 9, 699–721. 10.1080/713610005. [DOI] [Google Scholar]
- Li F.; Yan J.; Wei Y.; Zeng J.; Wang X.; Chen X.; Zhang C.; Li W.; Chen M.; Lü G. PM2.5-bound heavy metals from the major cities in China: Spatiotemporal distribution, fuzzy exposure assessment and health risk management. Journal of Cleaner Production 2021, 286, 124967. 10.1016/j.jclepro.2020.124967. [DOI] [Google Scholar]
- Farahani V. J.; Soleimanian E.; Pirhadi M.; Sioutas C. Long-term trends in concentrations and sources of PM2.5–bound metals and elements in central Los Angeles. Atmos. Environ. 2021, 253, 118361. 10.1016/j.atmosenv.2021.118361. [DOI] [Google Scholar]
- Zhang X.; Eto Y.; Aikawa M. Risk assessment and management of PM(2.5)-bound heavy metals in the urban area of Kitakyushu, Japan. Science of The Total Environment 2021, 795, 148748. 10.1016/j.scitotenv.2021.148748. [DOI] [PubMed] [Google Scholar]
- Sielski J.; Kaziród-Wolski K.; Jóźwiak M. A.; Jóźwiak M. The influence of air pollution by PM2.5, PM10 and associated heavy metals on the parameters of out-of-hospital cardiac arrest. Science of The Total Environment 2021, 788, 147541. 10.1016/j.scitotenv.2021.147541. [DOI] [PubMed] [Google Scholar]
- Lin Y.; Ma Y.; Qiu X.; Li R.; Fang Y.; Wang J.; Zhu Y.; Hu D. Sources, transformation, and health implications of PAHs and their nitrated, hydroxylated, and oxygenated derivatives in PM2.5 in Beijing. Journal of Geophysical Research: Atmospheres 2015, 120, 7219–7228. 10.1002/2015JD023628. [DOI] [Google Scholar]
- Hassanvand M. S.; Naddafi K.; Faridi S.; Nabizadeh R.; Sowlat M. H.; Momeniha F.; Gholampour A.; Arhami M.; Kashani H.; Zare A.; Niazi S.; Rastkari N.; Nazmara S.; Ghani M.; Yunesian M. Characterization of PAHs and metals in indoor/outdoor PM10/PM2.5/PM1 in a retirement home and a school dormitory. Science of The Total Environment 2015, 527–528, 100–110. 10.1016/j.scitotenv.2015.05.001. [DOI] [PubMed] [Google Scholar]
- Boström C. E.; Gerde P.; Hanberg A.; Jernström B.; Johansson C.; Kyrklund T.; Rannug A.; Törnqvist M.; Victorin K.; Westerholm R. Cancer risk assessment, indicators, and guidelines for polycyclic aromatic hydrocarbons in the ambient air. Environ. Health Perspect. 2002, 110, 451–488. 10.1289/ehp.110-1241197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WHO . Agents classified by the IARC Monographs.; International Agency for Research on Cancer, 2012; Vols. 1–106. [Google Scholar]
- Ma J.; Liu X.; Yang Y.; Qiu J.; Dong Z.; Ren Q.; Zuo Y. Y.; Xia T.; Chen W.; Liu S. Binding of benzo[a]pyrene alters the bioreactivity of fine biochar particles toward macrophages leading to deregulated macrophagic defense and autophagy. ACS Nano 2021, 15, 9717–9731. 10.1021/acsnano.1c00324. [DOI] [PubMed] [Google Scholar]
- Toepfer I.; Favet J.; Schulte A.; Schmölling M.; Butte W.; Triplett E. W.; Broughton W. J.; Gorbushina A. A. Pathogens as potential hitchhikers on intercontinental dust. Aerobiologia 2012, 28, 221–231. 10.1007/s10453-011-9230-2. [DOI] [Google Scholar]
- Zhang X.; Li Z.; Hu J.; Yan L.; He Y.; Li X.; Wang M.; Sun X.; Xu H. The biological and chemical contents of atmospheric particulate matter and implication of its role in the transmission of bacterial pathogenesis. Environmental Microbiology 2021, 23, 5481–5486. 10.1111/1462-2920.15679. [DOI] [PubMed] [Google Scholar]
- Pivato A.; Amoruso I.; Formenton G.; Di Maria F.; Bonato T.; Vanin S.; Marion A.; Baldovin T. Evaluating the presence of SARS-CoV-2 RNA in the particulate matters during the peak of COVID-19 in Padua, northern Italy. Science of The Total Environment 2021, 784, 147129. 10.1016/j.scitotenv.2021.147129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mushtaq N.; Ezzati M.; Hall L.; Dickson I.; Kirwan M.; Png K. M.; Mudway I. S.; Grigg J. Adhesion of Streptococcus pneumoniae to human airway epithelial cells exposed to urban particulate matter. Journal of allergy and clinical immunology 2011, 127, 1236–1242.e2. 10.1016/j.jaci.2010.11.039. [DOI] [PubMed] [Google Scholar]
- Chen X.; Liu J.; Zhou J.; Wang J.; Chen C.; Song Y.; Pan J. Urban particulate matter (PM) suppresses airway antibacterial defence. Respiratory research 2018, 19, 5. 10.1186/s12931-017-0700-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J.; Chen X.; Dou M.; He H.; Ju M.; Ji S.; Zhou J.; Chen C.; Zhang D.; Miao C.; Song Y. Particulate matter disrupts airway epithelial barrier via oxidative stress to promote Pseudomonas aeruginosa infection. Journal of thoracic disease 2019, 11, 2617–2627. 10.21037/jtd.2019.05.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uzman A. Molecular biology of the cell (4th ed.): Alberts, B., Johnson, A., Lewis, J., Raff, M., Roberts, K., and Walter, P. Biochemistry and Molecular Biology Education 2003, 31, 212–214. 10.1002/bmb.2003.494031049999. [DOI] [Google Scholar]
- Mollazadeh S.; Fazly Bazzaz B. S.; Kerachian M. A. Role of apoptosis in pathogenesis and treatment of bone-related diseases. Journal of orthopaedic surgery and research 2015, 10, 15. 10.1186/s13018-015-0152-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahoney J. A.; Rosen A. Apoptosis and autoimmunity. Current Opinion in Immunology 2005, 17, 583–588. 10.1016/j.coi.2005.09.018. [DOI] [PubMed] [Google Scholar]
- Mattson M. P. Apoptosis in neurodegenerative disorders. Nat. Rev. Mol. Cell Biol. 2000, 1, 120–130. 10.1038/35040009. [DOI] [PubMed] [Google Scholar]
- Aggarwal B. B. Apoptosis and nuclear factor-κB: a tale of association and dissociation. Biochemical pharmacology 2000, 60, 1033–1039. 10.1016/S0006-2952(00)00393-2. [DOI] [PubMed] [Google Scholar]
- Dagher Z.; Garcon G.; Billet S.; Gosset P.; Ledoux F.; Courcot D.; Aboukais A.; Shirali P. Activation of different pathways of apoptosis by air pollution particulate matter (PM2.5) in human epithelial lung cells (L132) in culture. Toxicology 2006, 225, 12–24. 10.1016/j.tox.2006.04.038. [DOI] [PubMed] [Google Scholar]
- Chen M.; Li B.; Sang N. Particulate matter (PM(2.5)) exposure season-dependently induces neuronal apoptosis and synaptic injuries. Journal of environmental sciences (China) 2017, 54, 336–345. 10.1016/j.jes.2016.10.013. [DOI] [PubMed] [Google Scholar]
- Zhou T.; Zhong Y.; Hu Y.; Sun C.; Wang Y.; Wang G. PM(2.5) downregulates miR-194–3p and accelerates apoptosis in cigarette-inflamed bronchial epithelium by targeting death-associated protein kinase 1. International journal of chronic obstructive pulmonary disease 2018, 13, 2339–2349. 10.2147/COPD.S168629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q.; Gan X.; Li F.; Chen Y.; Fu W.; Zhu X.; Xu D.; Long M.; Xu D. PM(2.5) Exposure Induces More Serious Apoptosis of Cardiomyocytes Mediated by Caspase3 through JNK/ P53 Pathway in Hyperlipidemic Rats. International journal of biological sciences 2019, 15, 24–33. 10.7150/ijbs.28633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dou C.; Zhang J.; Qi C. Cooking oil fume-derived PM(2.5) induces apoptosis in A549 cells and MAPK/NF-κB/STAT1 pathway activation. Environmental science and pollution research international 2018, 25, 9940–9948. 10.1007/s11356-018-1262-5. [DOI] [PubMed] [Google Scholar]
- Skuland T.; Ovrevik J.; Lag M.; Schwarze P.; Refsnes M. Silica nanoparticles induce cytokine responses in lung epithelial cells through activation of a p38/TACE/TGF-alpha/EGFR-pathway and NF-kappaBeta signalling. Toxicol. Appl. Pharmacol. 2014, 279, 76–86. 10.1016/j.taap.2014.05.006. [DOI] [PubMed] [Google Scholar]
- Yin J.; Xia W.; Li Y.; Guo C.; Zhang Y.; Huang S.; Jia Z.; Zhang A. COX-2 mediates PM2.5-induced apoptosis and inflammation in vascular endothelial cells. American journal of translational research 2017, 9, 3967–3976. [PMC free article] [PubMed] [Google Scholar]
- Wang W.; Deng Z.; Feng Y.; Liao F.; Zhou F.; Feng S.; Wang X. PM(2.5) induced apoptosis in endothelial cell through the activation of the p53-bax-caspase pathway. Chemosphere 2017, 177, 135–143. 10.1016/j.chemosphere.2017.02.144. [DOI] [PubMed] [Google Scholar]
- Yang X.; Zhao T.; Feng L.; Shi Y.; Jiang J.; Liang S.; Sun B.; Xu Q.; Duan J.; Sun Z. PM2. 5-induced ADRB2 hypermethylation contributed to cardiac dysfunction through cardiomyocytes apoptosis via PI3K/Akt pathway. Environ. Int. 2019, 127, 601–614. 10.1016/j.envint.2019.03.057. [DOI] [PubMed] [Google Scholar]
- Gholinejad Z.; Ansari M. H. K.; Rasmi Y. Titanium dioxide nanoparticles induce endothelial cell apoptosis via cell membrane oxidative damage and p38, PI3K/Akt, NF-κB signaling pathways modulation. Journal of Trace Elements in Medicine Biology 2019, 54, 27–35. 10.1016/j.jtemb.2019.03.008. [DOI] [PubMed] [Google Scholar]
- Roy R.; Singh S. K.; Chauhan L. K. S.; Das M.; Tripathi A.; Dwivedi P. D. Zinc oxide nanoparticles induce apoptosis by enhancement of autophagy via PI3K/Akt/mTOR inhibition. Toxicol. Lett. 2014, 227, 29–40. 10.1016/j.toxlet.2014.02.024. [DOI] [PubMed] [Google Scholar]
- Kundu J.; Kim D. H.; Chae I. G.; Lee J. K.; Lee S.; Jeong C. H.; Chun K. S. Silicon dioxide nanoparticles induce COX-2 expression through activation of STAT3 signaling pathway in HaCaT cells. Toxicology in Vitro 2018, 52, 235–242. 10.1016/j.tiv.2018.06.008. [DOI] [PubMed] [Google Scholar]
- Jin L.; Ni J.; Tao Y.; Weng X.; Zhu Y.; Yan J.; Hu B. N-acetylcysteine attenuates PM(2.5)-induced apoptosis by ROS-mediated Nrf2 pathway in human embryonic stem cells. Science of The Total Environment 2019, 666, 713–720. 10.1016/j.scitotenv.2019.02.307. [DOI] [PubMed] [Google Scholar]
- Zhang J.; Liu J.; Ren L.; Wei J.; Duan J.; Zhang L.; Zhou X.; Sun Z. PM2.5 induces male reproductive toxicity via mitochondrial dysfunction, DNA damage and RIPK1 mediated apoptotic signaling pathway. Science of The Total Environment 2018, 634, 1435–1444. 10.1016/j.scitotenv.2018.03.383. [DOI] [PubMed] [Google Scholar]
- Yuan X.; Wang Y.; Li L.; Zhou W.; Tian D.; Lu C.; Yu S.; Zhao J.; Peng S. PM(2.5) induces embryonic growth retardation: Potential involvement of ROS-MAPKs-apoptosis and G0/G1 arrest pathways. Environmental toxicology 2016, 31, 2028–2044. 10.1002/tox.22203. [DOI] [PubMed] [Google Scholar]
- Yin Y.; Peng H.; Shao J.; Zhang J.; Li Y.; Pi J.; Guo J. NRF2 deficiency sensitizes human keratinocytes to zinc oxide nanoparticles-induced autophagy and cytotoxicity. Environmental Toxicology and Pharmacology 2021, 87, 103721. 10.1016/j.etap.2021.103721. [DOI] [PubMed] [Google Scholar]
- Saha S.; Panigrahi D. P.; Patil S.; Bhutia S. K. Autophagy in health and disease: A comprehensive review. Biomedicine Pharmacotherapy 2018, 104, 485–495. 10.1016/j.biopha.2018.05.007. [DOI] [PubMed] [Google Scholar]
- Shang Y.; Wang Q.; Li J.; Liu H.; Zhao Q.; Huang X.; Dong H.; Chen W.; Gui R.; Nie X. Zirconia nanoparticles induce HeLa cell death through mitochondrial apoptosis and autophagy pathways mediated by ROS. Frontiers in chemistry 2021, 9, 522708. 10.3389/fchem.2021.522708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y.; Lin Z.; Huang H.; He H.; Yang L.; Chen T.; Yang T.; Ren N.; Jiang Y.; Xu W.; Kamp D. W.; Liu T.; Liu G. AMPK is required for PM2.5-induced autophagy in human lung epithelial A549 cells. International journal of clinical and experimental medicine 2015, 8, 58–72. [PMC free article] [PubMed] [Google Scholar]
- Zhu X. M.; Wang Q.; Xing W. W.; Long M. H.; Fu W. L.; Xia W. R.; Jin C.; Guo N.; Xu D. Q.; Xu D. G. PM2.5 induces autophagy-mediated cell death via NOS2 signaling in human bronchial epithelium cells. International journal of biological sciences 2018, 14, 557–564. 10.7150/ijbs.24546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia L.; Hao S. L.; Yang W. X. Nanoparticles induce autophagy via mTOR pathway inhibition and reactive oxygen species generation. Nanomedicine (Lond) 2020, 15, 1419–1435. 10.2217/nnm-2019-0387. [DOI] [PubMed] [Google Scholar]
- Xu X.; Wang H.; Liu S.; Xing C.; Liu Y.; Aodengqimuge; Zhou W.; Yuan X.; Ma Y.; Hu M.; Hu Y.; Zou S.; Gu Y.; Peng S.; Yuan S.; Li W.; Ma Y.; Song L. TP53-dependent autophagy links the ATR-CHEK1 axis activation to proinflammatory VEGFA production in human bronchial epithelial cells exposed to fine particulate matter (PM2.5). Autophagy 2016, 12, 1832–1848. 10.1080/15548627.2016.1204496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding R.; Zhang C.; Zhu X.; Cheng H.; Zhu F.; Xu Y.; Liu Y.; Wen L.; Cao J. ROS-AKT-mTOR axis mediates autophagy of human umbilical vein endothelial cells induced by cooking oil fumes-derived fine particulate matters in vitro. Free Radical Biol. Med. 2017, 113, 452–460. 10.1016/j.freeradbiomed.2017.10.386. [DOI] [PubMed] [Google Scholar]
- Xia W. R.; Fu W.; Wang Q.; Zhu X.; Xing W. W.; Wang M.; Xu D. Q.; Xu D. G. Autophagy induced FHL2 upregulation promotes IL-6 production by activating the NF-κB pathway in mouse aortic endothelial cells after exposure to PM2.5. International Journal of Molecular Sciences 2017, 18, 1484. 10.3390/ijms18071484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu Y.; Zheng Z.; Kim H.; Yang Z.; Zhang G.; Shi X.; Sun F.; Peng C.; Ding Y.; Wang A.; Chen L. C.; Rajagopalan S.; Sun Q.; Zhang K. Inhalation exposure to PM(2.5) counteracts hepatic steatosis in mice fed high-fat diet by stimulating hepatic autophagy. Sci. Rep. 2017, 7, 16286. 10.1038/s41598-017-16490-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bao X.; Shi M. Toxic effect of silica nanoparticles on bronchial epithelial cells. Materials Express 2022, 12, 255–262. 10.1166/mex.2022.2160. [DOI] [Google Scholar]
- Wang Y.; Tang M. PM2.5 induces autophagy and apoptosis through endoplasmic reticulum stress in human endothelial cells. Science of The Total Environment 2020, 710, 136397. 10.1016/j.scitotenv.2019.136397. [DOI] [PubMed] [Google Scholar]
- Yu P.; Zhang X.; Liu N.; Tang L.; Peng C.; Chen X. Pyroptosis: mechanisms and diseases. Signal Transduction and Targeted Therapy 2021, 6, 128. 10.1038/s41392-021-00507-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miao E. A.; Rajan J. V.; Aderem A. Caspase-1-induced pyroptotic cell death. Immunological Reviews 2011, 243, 206–214. 10.1111/j.1600-065X.2011.01044.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia R.; Wei M.; Zhang X.; Du R.; Sun W.; Wang L.; Song L. Pyroptosis participates in PM2.5-induced air–blood barrier dysfunction. Environmental Science and Pollution Research 2022, 29, 60987–60997. 10.1007/s11356-022-20098-0. [DOI] [PubMed] [Google Scholar]
- Ren F.; Xu J.; Zhang J.; Xu X.; Huang L.; Sun W.; Li R.; Li F. PM2.5 induced lung injury through upregulating ROS-dependent NLRP3 Inflammasome-Mediated Pyroptosis. Immunobiology 2022, 227, 152207. 10.1016/j.imbio.2022.152207. [DOI] [PubMed] [Google Scholar]
- Liang X.; Zhang D.; Liu W.; Yan Y.; Zhou F.; Wu W.; Yan Z. Reactive oxygen species trigger NF-κB-mediated NLRP3 inflammasome activation induced by zinc oxide nanoparticles in A549 cells. Toxicology and Industrial Health 2017, 33, 737–745. 10.1177/0748233717712409. [DOI] [PubMed] [Google Scholar]
- Gao L.; Qin J. X.; Shi J. Q.; Jiang T.; Wang F.; Xie C.; Gao Q.; Zhi N.; Dong Q.; Guan Y. T. Fine particulate matter exposure aggravates ischemic injury via NLRP3 inflammasome activation and pyroptosis. CNS Neuroscience & Therapeutics 2022, 28, 1045–1058. 10.1111/cns.13837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu C.; Zhang H.; Cui S.; Han B.; Zhou L.; Zhang N.; Su X.; Niu Y.; Chen W.; Chen R.; Zhang R.; Zheng Y. Ambient PM2.5 caused depressive-like responses through Nrf2/NLRP3 signaling pathway modulating inflammation. Journal of hazardous materials 2019, 369, 180–190. 10.1016/j.jhazmat.2019.02.026. [DOI] [PubMed] [Google Scholar]
- Wang B. R.; Shi J. Q.; Ge N. N.; Ou Z.; Tian Y. Y.; Jiang T.; Zhou J. S.; Xu J.; Zhang Y. D. PM2.5 exposure aggravates oligomeric amyloid beta-induced neuronal injury and promotes NLRP3 inflammasome activation in an in vitro model of Alzheimer’s disease. Journal of Neuroinflammation 2018, 15, 132. 10.1186/s12974-018-1178-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie Y.; Hou W.; Song X.; Yu Y.; Huang J.; Sun X.; Kang R.; Tang D. Ferroptosis: process and function. Cell Death Differ. 2016, 23, 369–379. 10.1038/cdd.2015.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stockwell B. R.; Friedmann Angeli J. P.; Bayir H.; Bush A. I.; Conrad M.; Dixon S. J.; Fulda S.; Gascón S.; Hatzios S. K.; Kagan V. E.; Noel K.; Jiang X.; Linkermann A.; Murphy M. E.; Overholtzer M.; Oyagi A.; Pagnussat G. C.; Park J.; Ran Q.; Rosenfeld C. S.; Salnikow K.; Tang D.; Torti F. M.; Torti S. V.; Toyokuni S.; Woerpel K. A.; Zhang D. D. Ferroptosis: A Regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017, 171, 273–285. 10.1016/j.cell.2017.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y.; Tang M. PM2.5 induces ferroptosis in human endothelial cells through iron overload and redox imbalance. Environ. Pollut. 2019, 254, 112937. 10.1016/j.envpol.2019.07.105. [DOI] [PubMed] [Google Scholar]
- Sharma B.; Singh S.; Siddiqi N. J. Biomedical implications of heavy metals induced imbalances in redox systems. BioMed. Research International 2014, 2014, 640754. 10.1155/2014/640754. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Xiong Q.; Tian X.; Xu C.; Ma B.; Liu W.; Sun B.; Ru Q.; Shu X. PM2.5 exposure-induced ferroptosis in neuronal cells via inhibiting ERK/CREB pathway. Environmental toxicology 2022, 37, 2201–2213. 10.1002/tox.23586. [DOI] [PubMed] [Google Scholar]
- Duan S.; Zhang M.; Sun Y.; Fang Z.; Wang H.; Li S.; Peng Y.; Li J.; Li J.; Tian J.; Yin H.; Yao S.; Zhang L. Mechanism of PM2.5-induced human bronchial epithelial cell toxicity in central China. Journal of hazardous materials 2020, 396, 122747. 10.1016/j.jhazmat.2020.122747. [DOI] [PubMed] [Google Scholar]
- Zhang J.; Hao X.; Xu S. Selenium prevents lead-induced necroptosis by restoring antioxidant functions and blocking MAPK/NF-κB pathway in chicken lymphocytes. Biological Trace Element Research 2020, 198, 644–653. 10.1007/s12011-020-02094-y. [DOI] [PubMed] [Google Scholar]
- Zhang S.; Che L.; He C.; Huang J.; Guo N.; Shi J.; Lin Y.; Lin Z. Drp1 and RB interaction to mediate mitochondria-dependent necroptosis induced by cadmium in hepatocytes. Cell Death Disease 2019, 10, 523. 10.1038/s41419-019-1730-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin K.; Wang D.; Zhao H.; Wang Y.; Zhang Y.; Liu Y.; Li B.; Xing M. Polystyrene microplastics up-regulates liver glutamine and glutamate synthesis and promotes autophagy-dependent ferroptosis and apoptosis in the cerebellum through the liver-brain axis. Environ. Pollut. 2022, 307, 119449. 10.1016/j.envpol.2022.119449. [DOI] [PubMed] [Google Scholar]
- Aust A. E.; Ball J. C.; Hu A. A.; Lighty J. S.; Smith K. R.; Straccia A. M.; Veranth J. M.; Young W. C. Particle characteristics responsible for effects on human lung epithelial cells. Res. Rep. Health Eff Inst 2002, 1–65, 67–76. [PubMed] [Google Scholar]
- Qi Y.; Chen Y.; Xia T.; Lynch I.; Liu S. Extra-pulmonary translocation of exogenous ambient nanoparticles in the human body. ACS Nano 2023, 17, 12. 10.1021/acsnano.2c09299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calderón-Garcidueñas L.; González-Maciel A.; Mukherjee P. S.; Reynoso-Robles R.; Pérez-Guillé B.; Gayosso-Chávez C.; Torres-Jardón R.; Cross J. V.; Ahmed I. A.; Karloukovski V. V.; et al. Combustion-and friction-derived magnetic air pollution nanoparticles in human hearts. Environmental Research 2019, 176, 108567. 10.1016/j.envres.2019.108567. [DOI] [PubMed] [Google Scholar]
- Qi Y.; Wei S.; Xin T.; Huang C.; Pu Y.; Ma J.; Zhang C.; Liu Y.; Lynch I.; Liu S. Passage of exogeneous fine particles from the lung into the brain in humans and animals. Proc. Natl. Acad. Sci. U. S. A. 2022, 119, e2117083119. 10.1073/pnas.2117083119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qi Y.; Wei S.; Chen Y.; Pu Y.; Liu S.; Liu Y. Intrusion of inhaled exotic ultrafine particles into the knee joint in humans and animals: A risk to the joint and surrounding tissues. Nano Today 2022, 43, 101426. 10.1016/j.nantod.2022.101426. [DOI] [Google Scholar]
- Liu X.; Ma J.; Ji R.; Wang S.; Zhang Q.; Zhang C.; Liu S.; Chen W. Biochar fine particles enhance uptake of benzo(a)pyrene to macrophages and epithelial cells via different mechanisms. Environmental Science & Technology Letters 2021, 8, 218–223. 10.1021/acs.estlett.0c00900. [DOI] [Google Scholar]
- Chen M.; Li B.; Sang N. Particulate matter (PM(2.5)) exposure season-dependently induces neuronal apoptosis and synaptic injuries. Environ. Sci. (China) 2017, 54, 336–345. 10.1016/j.jes.2016.10.013. [DOI] [PubMed] [Google Scholar]
- Zhou T.; Zhong Y.; Hu Y.; Sun C.; Wang Y.; Wang G. PM(2.5) downregulates miR-194–3p and accelerates apoptosis in cigarette-inflamed bronchial epithelium by targeting death-associated protein kinase 1. International journal of chronic obstructive pulmonary disease 2018, 13, 2339–2349. 10.2147/COPD.S168629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q.; Gan X.; Li F.; Chen Y.; Fu W.; Zhu X.; Xu D.; Long M.; Xu D. PM(2.5) exposure induces more serious apoptosis of cardiomyocytes mediated by caspase3 through JNK/ P53 pathway in hyperlipidemic rats. International journal of biological sciences 2019, 15, 24–33. 10.7150/ijbs.28633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu X. M.; Wang Q.; Xing W. W.; Long M. H.; Fu W. L.; Xia W. R.; Jin C.; Guo N.; Xu D. Q.; Xu D. G. PM2.5 induces autophagy-mediated cell death via NOS2 signaling in human bronchial epithelium cells. International journal of biological sciences 2018, 14, 557–564. 10.7150/ijbs.24546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan S.; Wang N.; Huang L.; Zhao Y.; Shao H.; Jin Y.; Zhang R.; Li C.; Wu W.; Wang J.; Feng F. NLRP3 inflammasome activation is associated with PM(2.5) -induced cardiac functional and pathological injury in mice. Environmental toxicology 2019, 34, 1246–1254. 10.1002/tox.22825. [DOI] [PubMed] [Google Scholar]
- Chu C.; Zhang H.; Cui S.; Han B.; Zhou L.; Zhang N.; Su X.; Niu Y.; Chen W.; Chen R.; Zhang R.; Zheng Y. Ambient PM2.5 caused depressive-like responses through Nrf2/NLRP3 signaling pathway modulating inflammation. Journal of hazardous materials 2019, 369, 180–190. 10.1016/j.jhazmat.2019.02.026. [DOI] [PubMed] [Google Scholar]
- Wang B. R.; Shi J. Q.; Ge N. N.; Ou Z.; Tian Y. Y.; Jiang T.; Zhou J. S.; Xu J.; Zhang Y. D. PM2.5 exposure aggravates oligomeric amyloid beta-induced neuronal injury and promotes NLRP3 inflammasome activation in an in vitro model of Alzheimer’s disease. Journal of neuroinflammation 2018, 15, 132. 10.1186/s12974-018-1178-5. [DOI] [PMC free article] [PubMed] [Google Scholar]





