Fine particulate matter‑induced cardiac developmental toxicity (Review)
- Authors:
- Published online on: October 29, 2024 https://doi.org/10.3892/etm.2024.12756
- Article Number: 6
-
Copyright: © Meng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
Environmental fine particulate matter of diameter ≤2.5 µm (PM2.5) possesses small volume, large surface area, and is able to absorb harmful substances. It is a mixture consisting of various particles including organic, inorganic, metal and trace elements, and has become an important risk factor threatening human health (1,2). There is evidence to suggest that the toxicity of PM2.5 varies depending on its components (3-5). The organic compounds in PM2.5, including polycyclic aromatic hydrocarbons (PAHs), have attracted widespread attention due to their association with developmental toxicity (6). Moreover, PM2.5 can easily enter the circulatory system and cross the placental barrier, thereby exerting negative impacts on fetal development (7). Congenital heart defect (CHD), the most common congenital defect in humans, accounts for ~1% of all live births (8). As the first organ to form, the heart is extremely sensitive to environmental stress during embryonic development (9). The exact mechanisms underlying CHD have yet to be fully elucidated, although they are currently considered to arise as a consequence of the combination between genetic and environmental factors (10). Gestational exposure to PM2.5 has been reported to be closely correlated with the occurrence of CHD in offspring (11-14). PM2.5 is one of the major air pollutants in the world, posing a huge threat to human health. It is estimated that air pollution caused 9 million premature deaths in 2015, accounting for 16% of the global death toll, with 1.2 million deaths induced by PM2.5. In fact, environmental pollution has become the leading cause of reversible death and disability resulting from cardiovascular diseases or cancers. Animal studies have also demonstrated that exposure to PM2.5 can significantly increase the incidence of cardiac abnormalities in mice, chickens and zebrafish (15-17). Only 10-25% of the total cases of CHD have been demonstrated to be caused solely by genetic factors, with the majority of the cases being associated with external factors (18). Recent investigations by Yan et al (19) have summarized five primary mechanisms through which PM2.5 affects adverse birth outcomes: Transcriptional and translational regulation, oxidative stress (OS) and inflammatory responses, and epigenetic regulation. While the present study review provides valuable insights into the broader developmental toxicity of PM2.5, it offers a limited focus on the specific cardiac effects. Feng et al (20) systematically reviewed the molecular and pathophysiological mechanisms by which PM2.5 impacts the cardiovascular system, encompassing metabolic activation, OS, genetic toxicity, inflammation, Ca2+ dysregulation, autophagy interference and apoptosis induction. Additionally, Liang et al (21) proposed an adverse outcome pathway framework to elucidate the relationship between PM2.5-induced molecular events and adverse cardiac outcomes, suggesting that excessive reactive oxygen species (ROS) generation and activation of aromatic hydrocarbon receptors (AhR) are critical initiating events (21). These events lead to OS, endoplasmic reticulum stress (ERS), DNA damage, inflammation and activation of the Wnt/β-catenin pathway, ultimately resulting in apoptosis and impaired cardiomyocyte differentiation.
Despite significant advances in understanding PM2.5's cardiac developmental toxicity, there remains a notable deficiency in research focusing on the specific components of PM2.5 that drive these detrimental effects. The mechanisms through which gestational exposure to PM2.5 induces cardiac developmental toxicity require further elucidation, and our current understanding remains limited. Therefore, it is essential to explore strategies for preventing and controlling PM2.5 exposure and to investigate the underlying mechanisms of its cardiac developmental toxicity. The present review aimed to highlight the need for further research on the specific components of PM2.5 that drive cardiac developmental toxicity. These mechanisms were detailed and the sources and components of PM2.5 were linked with their corresponding pathways of action, enhancing the understanding of PM2.5-induced cardiac developmental toxicity. Finally, mitigation strategies to reduce health risks associated with PM2.5 exposure were discussed and future perspectives on these strategies were outlined.
2. Cardiac developmental toxicity of PM2.5
Previous epidemiological studies on the association between PM2.5 exposure and CHD have yielded inconsistent conclusions (12,22-26), probably due to the heterogeneities of the studies concerned. Existing evidence suggests that the association between PM2.5 and CHD is mainly focused on pregnant women exposed to PM2.5 between the second and seventh week of pregnancy, a critical period for cardiac development (27). Furthermore, pro-gestational exposure to PM2.5 is also detrimental for pregnant women and infants (28). It is worth noting that maternal exposure to PM2.5 increases the risk of CHD in offspring, with the most susceptible time windows being 7-12 weeks before pregnancy and 3-9 weeks after pregnancy, demonstrating the especially adverse effects of PM2.5 exposure on the risk of developing CHD with respect to cardiac development during these two critical periods (29). Several studies that have explored the association between gestational PM2.5 exposure and CHD subtypes are presented in Table I.
3. Potential mechanisms underlying the cardiac developmental toxicity of PM2.5
Interference with genes associated with cardiac development
As transcription factors, GATA4 and NKX2.5 perform crucial roles in fetal cardiac development. Gestational PM2.5 exposure may increase the risk of GATA4 and NKX2.5 mutations, directly causing fetal cardiac abnormalities. Wu et al (30) found that gestational PM2.5 exposure leads to cardiac hypertrophy with elevated mRNA levels of GATA4 in offspring mice. Moreover, the important regulatory role of GATA4 in signaling pathways involved in cardiac development has been confirmed (31,32). It has been revealed to regulate the expression of key downstream genes involved in cardiac cell proliferation, development and hypertrophy, including ANP, CARP, a-MHC and β-MHC (32).
As a key factor in myocardial formation, the downregulation of GATA4 leads to an increase in the risk of cardiac structural abnormalities and cardiovascular malformations in the fetus. Inhibition of GATA4 in the early stage of cardiac development has been revealed to be associated with myocardial hypoplasia and CHD, whereas its inactivation in the late stage of cardiac development leads to decreased cardiac function (32). GATA4 is involved in normal cardiac development, functional gene expression, and the pathological processes of cardiac hypertrophy. It has been recognized as a key effector mediating cardiac gene transcription in response to hypertrophic stimuli. In addition, during myocardial hypertrophy, GATA4 serves as a molecular ‘bridge’ connecting multiple nuclear factors, including myocyte enhancer factor 2C (Mef2c), Nkx2.5 and AP1 (33,34).
Dysfunction of genes associated with heart function
In addition to the importance of the concentration of PM2.5, specific chemical components therein may exert more critical and important roles in the negative effects on health (35). Although heavy metals and PAHs only account for a small proportion of the PM2.5 mass, their potential toxicity should not be underestimated. PM2.5 exposure elicits stronger effects on the expression of cardiac genes than it does on genes in the lung, especially those genes associated with collagen, laminin and calcium (Ca2+) signaling (36). PM2.5 exposure leads to elevated levels of several genes associated with collagen deposition, including collagen type I, α1, Col3α1 and transforming growth factor β1 (TGFβ1) (37). Cardiac fibrosis is associated with an imbalance between the generation and degradation of extracellular matrix, resulting in the accumulation of scar tissue; consequently, PM2.5-induced cardiac fibrosis reduce the compliance of the extracellular matrix, impairing the cardiac capability to contract and relax normally. Qi et al (38) reported that PM2.5 and its water-soluble components associated with transportation induce cardiomyocytes dysfunction through ERS and autophagy. In this regard, heavy metals and PAHs in PM2.5 may be the primary influencing factors (38). In addition, the impact of PM2.5 exposure on cardiac function has been revealed to be seasonal, as levels of locally sourced elements in PM2.5 demonstrate significant seasonal changes (39). In summer, the emissions of iron (Fe), lead (Pb) and zinc (Zn) from steel plants, as well as Fe, titanium (Ti), chromium (Cr), manganese (Mn) and arsenic (As) from automobiles, have been revealed to be significantly associated with a reduction of the standard deviation of the mean to mean intervals (36). By contrast, in winter, elements such as barium (Ba), Zn, As and rubidium (Rb) were revealed to be correlated with an increased heart rate (39). Moreover, an association between an increased heart rate, albeit with decreased heart rate variability, was observed with nickel (Ni) and element carbon (40,41).
Ca2+ ions act as an important mediator in maintaining the normal contraction and relaxation of the heart, and numerous studies have identified calmodulin as an important target for cardiac dysfunction. After uterine exposure to PM2.5, significant changes in the levels of Ca2+-regulatory proteins, including sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2A (SERCA2A), sodium (Na+)/Ca2+ exchanger (NCX) and the voltage-gated Ca2+ channel (Cav1.2), were observed in fetal mice hearts on day 14 following pregnancy (42). Newborn mice exposed to PM2.5 during pregnancy experience cardiac dysfunction due to changes in Ca2+-processing proteins that are associated with downregulated levels of NCX and CaV1.2 in the heart. Furthermore, in a study on heart failure in rabbits, inhibition of NCX led to a reduction in the burden of premature ventricular beats, although there was no resultant inhibition of secondary Ca2+ elevation. As a compensatory mechanism, the levels of Serca2A and phosphorylated phospholamban were found to increase to cope with higher intracellular Ca2+ concentrations. The cardiac action potential performs a crucial role in cardiac synchronization, as it is regulated by cardiac ion channels such as voltage-gated Na+ and potassium (K+) channels (43). Park et al (44) found that exposure to particulate matters induced the abnormal expression (downregulation) of fetal heart ion channel-associated genes, including scn5lab, kcnq1 and kcnq1.
OS
OS, caused by free radicals, serves as a key factor leading to cellular and tissue oxidative damage, as well as being a major driver of aging and various diseases. ROS are the most important free radical that cause oxidative damage to the body. OS is the most common mechanism underlying PM2.5-induced damage (45,46). Transition metals [Fe, Cu and Mn] and organic compounds from PM2.5 are able to induce the production of ROS and reactive nitrogen species (RNS), and the ability of PM2.5 to induce ROS has been significantly correlated with the concentrations of PAHs and specific transition metals therein (47). PM2.5 may induce OS in target cells through a variety of pathways. First, PM2.5 contains persistent free radicals that are found in the environment, especially combustion-derived particles (48). Secondly, numerous organic compounds from PM2.5 can be metabolized into reactive electrophilic metabolites, which thereby induce the further generation of ROS (49). Thirdly, transition metals can induce ROS through the Fenton reaction (50). Finally, OS may also be caused by the PM2.5-mediated activation of inflammatory cells, which are able to produce both ROS and RNS (51). On the other hand, PM2.5 may also decrease the cellular antioxidant capacity through downregulating the expression of antioxidant enzymes, such as superoxide dismutase and glutathione metabolizing enzymes (52). ROS react with biomolecules such as proteins and DNA, resulting in various adverse effects on cells, including the disruption of their structure and function, which ultimately leads to damage to target cells and tissues. Two pathways are mainly involved in the pathogenic mechanisms underlying: One is gene damage resulting from genetic mutations, and the other is damage that is caused to the cell membrane, which results in changes in its permeability through lipid peroxidation, leading to physiological changes such as inflammation.
The embryonic development of both humans and zebrafish is abnormally sensitive to OS induced by ROS, and excessive ROS production is considered one of the factors contributing to CHD (53,54). Ren et al (55) demonstrated that extractable organic matter (EOM) from PM2.5 is able to induce ROS production, thereby increasing the levels of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway-associated genes [namely, SOD2, glutathione S-transferase (GST)P1/2, catalase (CAT) and heme oxygenase-1 (HO-1)], with the Nrf2 signaling pathway being the major pathway that is activated by OS. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), an AhR agonist, was revealed to cause an upregulation of the protein levels and activity of Nrf2 in mice (56). The presence of multiple AhR-binding elements located in the promoter and first intron of Nrf2a and Nrf2b suggests that AhR exerts a regulatory role with respect to their transcription (57). According to other research results, OS, in turn, may inhibit the activity of AhR (58). Elbekai and El-Kadi (59) reported that the ROS scavenger N-acetylcysteine (NAC) could ameliorate the inhibitory effects of chromium on AhR activity in human liver cell line (59). NAC treatment led to an increase in the activity of cytochrome P450, family 1, subfamily A, polypeptide 1 (Cyp1a1), whereas the inhibitory effects of AhR inhibitor, CH223191, were alleviated (60). However, Ren et al (55) found that NAC did not reduce EOM-induced AhR activity, suggesting that the effects of OS on the AhR signaling pathway may be species- or cell type-specific (55). Zebrafish possess two Nrf2 genes (Nrf2a and Nrf2b), whose downstream genes (SOD2, GSTP1/2, CAT and HO-1) exert a range of antioxidant effects, and this may represent a negative feedback mechanism to circumvent EOM-induced excessive ROS (57).
AhR are activated by PAHs from PM2.5, which consequently upregulates the levels of CYP metabolic enzymes and induces ROS via superoxide/hydrogen peroxide (61-63). Vertebrate embryos are highly susceptible to OS due to their limited antioxidant capacity (54). Ren et al (55) demonstrated that CH223191 and NAC are able to markedly alleviate PM2.5-induced zebrafish embryonic cardiac abnormalities. Furthermore, the two compounds were also revealed to reduce EOM-induced ROS generation, DNA damage and cell apoptosis, ameliorating the resultant changes in the mRNA expression levels of genes associated with cardiac development (NKX2.5 and SOX9B), OS (NRF2A, NRF2B, GSTP1, GSTP 2, SOD2, HO-1 and CAT) and apoptosis (p53 and Bax). These results confirmed that AhR mediates EOM-induced OS, leading to DNA damage and cell apoptosis, thereby promoting the cardiac developmental toxicity of PM2.5 (55). The most significant OS response induced by PM2.5 exposure is excessive oxidative phosphorylation in myocardial cells, which ultimately leads to mitochondrial damage and myocardial cell death (64). It is noteworthy that such adverse effects may often be significantly alleviated by antioxidants (65), demonstrating the potential of antioxidants in either preventing or mitigating OS damage caused by PM2.5 exposure.
Inflammation
Inflammation is an adaptive response for the body that both enables the clearance of harmful stimuli and heals damaged tissues. However, persistent or chronic inflammation may be detrimental to the body (66). As an important mechanism that is associated with PM2.5 toxicity, the inflammatory response may impose the negative effects of PM2.5 on the cardiovascular, pulmonary and nervous systems (67). The PAHs, metals, water-soluble ions as well as various bioactive substances (such as endotoxins) that are contained in PM2.5 may cause inflammation, a process that is associated with the polarization of pro-inflammatory macrophages (68). PM2.5 has been revealed to cause an increase in the levels of ROS in macrophages, and is recognized by the Toll-like receptors TLR4 and TLR2, leading to the induction or exacerbation of acute inflammation and thereby promoting M1 polarization of macrophages (69). This process may also involve the activation of Notch signaling due to a decreased level of the microRNA, miR-34a-5p (70). In addition, exposure to PAHs has been revealed to upregulate the levels of inducible nitric oxide synthase (iNOS), NLR family pyrin domain containing 3 (NLRP3) and tissue protease B in macrophages, demonstrating that pyroptosis provides the basis for the pro-inflammatory polarization of macrophages induced by exposure to PAHs (71). Myocardial macrophages are able to eliminate the defective mitochondria that are released by cardiomyocytes, thereby maintaining cardiac mitochondrial homeostasis. However, in the absence of membrane-bound bone marrow epithelial reproductive receptor tyrosine kinase (MerTK), myocardial macrophages lose the ability to capture and eliminate defective mitochondria, leading to dysfunctional cardiac metabolism and left ventricular dysfunction, suggesting that MerTK fulfills a crucial role in supporting cardiac homeostasis (72). Pro-inflammatory polarization of myocardial macrophages promotes the lysis of MerTK, which affects the ability of myocardial macrophages to participate in cardiac repair, consequently leading to cardiac homeostasis imbalance, myocardial injury and decreased cardiac function.
The important pro-inflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6 are involved in the pathogenesis of heart failure, cardiac hypertrophy and fibrosis (73,74). It has been revealed that exposure to PM2.5 in the uterus induces the expression of pro-inflammatory cytokines in the hearts of the offspring mice, leading to cardiac inflammation (30,42). Li et al (75) through studying the cardiac inflammatory response, demonstrated that the levels of TNF-α and IL-1β were significantly increased in offspring mice subjected to uterine PM2.5 exposure. Long-term exposure to PM2.5 was revealed to cause a marked upregulation of the levels of intercellular adhesion molecule-1 (ICAM-1) and C-reactive protein in rat myocardial tissues, leading to ultrastructural changes in myocardial cells and inflammatory cell influx (76). Among the signaling molecules that regulate the inflammatory response, nuclear factor-κB (NF-κB) is the major signaling molecule that is involved in the production of cytokines, chemokines and growth factors, regulating the expression of immune and inflammatory response-associated genes (77). The NF-κB signaling pathway, involved in tissue damage, has been reported to have a role in systemic inflammation induced by PM2.5 (78). Inflammation fulfills crucial roles in systemic myocardial hypertrophy and cardiotoxicity induced by particulate matter (79). Jiao et al (80) found that the PM2.5-mediated induction of inflammation is dependent on the activation of the key transcription factor NF-κB, which enhances the expression of the downstream factors, TNF-α and IL-1β. Exposure to PM2.5 in vivo has been revealed to activate the NF-κB signaling pathway, leading to inflammatory responses in target tissues and organs (81). Interesting, the activation of NF-κB, with the subsequent inflammatory response that is caused by exposure to PM2.5, may be suppressed by antioxidants, suggesting the involvement of ROS and/or RNS in PM2.5-mediated NF-κB activation (52). Considering that NF-κB also triggers the generation of ROS and nitric oxide (NO), this may form a positive feedback loop that amplifies downstream responses upon PM2.5 exposure (82). An increased level of OS resulting from exposure to PM2.5, in turn, mediates the activation of downstream inflammatory signaling pathways, including the mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK)/p53, Nrf2/NLRP3, TLR/MyD88 and extracellular signal-regulated kinase (ERK)/AKT pathways (83). PM2.5 has been revealed to increase the protein level of cleaved IL-1β, a key downstream factor for NLRP3 inflammasome activation, further confirming that PM2.5 can activate the NLRP3 inflammasome in myocardial tissue; NLRP3 inflammasome activation, in itself, has a potential role in mediating the pathological damage resulting from PM2.5 exposure in the mouse heart (84). The augmented levels of ROS triggered by exposure to PM2.5 may activate the MAPK and NF-κB pathways, thereby increasing the synthesis of inflammatory proteins and leading to changes in membrane permeability and mitochondrial dysfunction (85). It is worth noting that mitochondrial DNA (mtDNA) lacks the ability to repair DNA, making it more susceptible to oxidative damage compared with nuclear DNA. Mitochondrial dysfunction and subsequent cell death can trigger inflammation in various types of tissues (86). Mitochondrial dysfunction makes a key contribution to the PM2.5-mediated inflammatory response (87). mtDNA and n-formyl peptides that are released from dysfunctional mitochondria both trigger inflammation. PM2.5 exposure has also been revealed to increase the expression and release of adhesion molecules, including E-selectin, P-selectin and ICAM-1, leading to monocyte/macrophage adhesion (88), whereas, on the other hand, diminishing the levels of circulating endothelial progenitor cells that are involved in postnatal endothelial repair and regeneration (89), thereby exacerbating the inflammatory response. Inflammatory factors such as cyclooxygenase-2 (COX-2) are able to inhibit the activity of Ca2+ pumps in the endoplasmic reticulum, thereby inducing ERS through upregulating iNOS expression (90), suggesting that inflammation induced by PM2.5 can trigger ERS. Furthermore, PM2.5 has been revealed to activate the unfolded protein response (UPR), which provides an additional mechanism for triggering ERS (91). UPR signaling both stimulates the expression of inflammatory cytokines and induces the activation of NF-κB (92), suggesting that UPR signaling makes an important contribution towards PM2.5-induced ERS in the inflammatory process, and that this serves as an inflammatory factor both as a cause and as a consequence of ERS (93). Ca2+ leakage from the endoplasmic reticulum directly drives the production of mitochondrial ROS (mtROS), affecting downstream signaling pathways and rendering cells more susceptible to autophagy (94). It is now well documented that inflammation, ERS and autophagy are closely interlinked, and that these processes can interact with each other. Taken together, these aforementioned findings suggest that the cardiac developmental toxicity that is caused by PM2.5 is associated with inflammation, ERS and autophagy.
Mitochondrial impairment
The biogenesis and functional improvement of mitochondria are crucial processes for enabling the differentiation and maturation of the heart (95). Previously, investigations of the molecular mechanisms associated with mitochondria underlying the toxic effects of environmental pollution have been mainly focused on the mitochondrial permeability transition pore (mPTP), mitochondrial dynamics, mtDNA function and the mitochondrial respiratory chain system, along with mitochondrial damage-associated signaling pathways. PM2.5 was found to induce mitochondrial impairment in exposed individuals (96), and mitochondrial dysfunction has been revealed to mediate the cardiovascular damage caused by PM2.5 to a certain extent (97). Enhancing the production of cardiac energy may be achieved through growing the mitochondria count (98), and swelling, disrupted crista and mitochondrial vacuolization represent the primary manifestations for cardiac mitochondrial pathological changes (99). Acute exposure to PM can lead to significant mitochondrial dysfunction, accompanied by decreased cardiac oxygen consumption, succinate dehydrogenase activity and mitochondrial membrane potential, as well as impaired oxidative phosphorylation (100). These findings suggested that mitochondrial damage caused by PM2.5 exposure may have a bearing on mitochondrial dysfunction.
Inflammatory response and OS, fulfilling important roles in PM2.5-induced cardiac injury, can produce a large number of free radicals that are closely associated with mitochondrial damage (101). Proteins or complexes modulating cell apoptosis are only able to function via cytochrome c after entering the mitochondrial membrane (102). Therefore, mitochondria exert a crucial role in the cardiac toxicity that is mediated by PM2.5. In order to exert their own function, mitochondria must undergo continuous fission and fusion, abnormalities of which may induce diseases (103). OPA1 mitochondrial dynamin-like GTPase (OPA1) along with Mfp-1 and -2 jointly regulate mitochondrial fusion, with the large GTPase, dynamin-related protein 1 (Drp1) and the mitochondrial outer membrane protein adaptor, fission 1 (Fis1) mediating mitochondrial fission (104). The normal expression of fusion/fission genes is a prerequisite for the normal function of mitochondria, otherwise mitochondrial dysfunction may occur (105). Wang et al (16) identified elevated levels of OPA1, Mfn1, Drp1 and Fis1 in offspring rats with the dosage of gestational PM2.5 exposure, and surmised that the dysregulated mitochondrial fusion/fission genes resulting from gestation PM2.5 exposure in these rats would exert detrimental effects on mitochondrial damage, subsequently leading to an induction of cardiac developmental toxicity in the offspring.
OS, an imbalance of Ca2+ homeostasis, and inflammation are all closely associated with mitochondrial dysfunction in various heart diseases (106). First, mitochondria are the main source of ROS production (107). Excessive ROS has been revealed to induce lipid peroxidation, thereby leading to mitochondrial permeability, decreased membrane potential and mitochondrial swelling (108). In addition, mitochondrial superoxide reacts with RNS such as NO to form peroxynitrite, further damaging mitochondrial structure and function (109); therefore, mitochondria are sensitive targets for PM2.5 and OS (110). Rodriguez-Enriquez et al (111) reported that ROS, Ca2+ overload, decreased mitochondrial membrane potential and excessive mitochondrial permeability are key triggers of mitochondrial swelling and outer membrane rupture. A strong correlation was found to exist between mitochondrial dysfunction and the severity of inflammation (112). It is well established that peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) is the main mediator for mitochondrial biogenesis and function in mammals (113), and environmental chemicals have been revealed to induce mitochondrial damage via inhibiting PGC-1α (114). Chen et al (115) reported that AhR activated by PM2.5 directly inhibits sirtuin 1, thereby both reducing the levels of PGC-1α and increasing its level of acetylation, which has the effect of diminishing its activity. Damaged PGC-1α subsequently induces mitochondrial dysfunction, ultimately leading to cardiac developmental defects in zebrafish juveniles (115). In addition, this research group also found that EOM derived from PM2.5 induces the overexpression of CYP1A1 via activating AhR, leading to the generation of mtROS. The increase in mtROS levels subsequently exacerbates the opening of the mPTP, which, in turn, promotes the accumulation of mtROS. Opening of the mPTP promotes the release of pro-apoptotic substances, thereby triggering the intrinsic apoptotic pathway and leading to cardiac defects (116). Further investigations are required, however, regarding the possible involvement of other associated mechanisms of mitochondrial dysfunction elicited by PM2.5, such as cytochrome c release, mtDNA changes and cell apoptosis, along with mitochondrial genomic variations in PM2.5-induced cardiac developmental toxicity.
Epigenetic modification
Epigenetic modifications, including DNA methylation (DNAm), histone modifications and RNA-mediated processes, are sensitive to environmental stress and are considered to serve as a ‘bridge’ between environmental and genetic factors by certain researchers (117,118). Epigenetic modifications fulfill an important role in cardiac development and the occurrence of various diseases, with DNAm as the primary form, which can be inherited and reversed. To date, however, little is known regarding the underlying molecular mechanisms through which PM2.5 triggers the epigenetic changes that lead to cardiac developmental toxicity.
1 N6-Methyladenosine (M6A) RNA methylation
M6A RNA methylation, as the most common form of RNA modification, accounts for ~60% of the total number of RNA modifications. M6A RNA methylation, a dynamic and reversible process, occurs under the regulation of methyltransferases (including METTL3 and METTL14), demethylases (such as FTO and ALKBH5,), and binding proteins (including YTHDF1/2/3 and ythdc2/2) (119). M6A RNA methylation regulates gene expression through affecting mRNA stability, selective splicing, nuclear output and protein translation (120,121). M6A RNA methylation has been reported to be involved in excessive cellular ROS production and apoptosis (122-124). A crucial role of m6A modification in heart development has been demonstrated (124,125), and PM2.5 has been revealed to induce changes in m6A RNA methylation in rats and mice (126,127). Ji et al (128) found that EOM from PM2.5 caused a significant inhibition of m6A RNA methylation levels in zebrafish juvenile hearts mediated via the AhR, although this inhibitory effect was restored by supplementation with betaine (the predominant methyl donor in the carbon metabolism cycle). Betaine can also mitigate EOM-induced ROS generation, cell apoptosis and cardiac defects, suggesting that EOM inhibits m6A RNA methylation by interfering with mettl14/mettl3 expression, leading to cardiac defects (128). These findings validated the hypothesis that m6A modification fulfills an important role in cardiac developmental toxicity induced by PM2.5 exposure, although the antioxidant activity of betaine should not be overlooked. On the other hand, other studies have revealed that exposure to PM2.5 leads to an upregulation of the levels of Mettl3 and total m6A methylation in mice lung tissues (127,129). The differences noted in the expression levels of m6A methyltransferase may be due to the differential responses of these genes to PM2.5 exposure in embryonic/larval and adult tissues (130); another possibility is that changes in the level of m6A RNA methylation induced by PM2.5 exposure may be due to species specificity.
Supplementing the diet with AhR inhibitor, CH223191, has been reported to successfully circumvent the occurrence of EOM-induced cardiac defects in zebrafish juveniles (17,55). Either adding betaine or overexpressing mett13/14 was revealed to ameliorate the effects of EOM-induced intracellular and mtROS, as well as reducing the level of apoptosis in zebrafish juvenile cardiomyocytes. Therefore, changes that occur in the level of m6A RNA methylation may be an important underlying cause of EOM-induced cardiac abnormalities. M6A modification has been revealed to regulate OS and cell apoptosis via regulating the expression of m6A-modified genes (131,132). Cao et al (133) reported that exposure to PM2.5 increases ROS generation and apoptosis in rat cardiomyocytes, leading to cardiac injury. In addition, EOM from PM2.5 led to OS-mediated cell apoptosis in zebrafish juvenile hearts (55). Collectively, these studies have demonstrated that gestational exposure to PM2.5 may cause OS and cell apoptosis through altering m6A modification, thereby causing cardiac developmental toxicity.
DNAm. DNAm, one of the most extensively studied epigenetic modifications, performs a crucial role in cardiac development. Abnormal DNAm has been revealed to be associated with the pathogenesis of CHD (134), and it has been revealed that PM2.5 causes abnormal changes in DNAm (135); therefore, it is possible that DNAm may be associated with the cardiac developmental toxicity that is induced by PM2.5 exposure. As a major environmental sensor, AhR is able to bind and be activated by various environmental pollutants, including PAHs (136,137). Following activation, AhR is translocated from the cytoplasm to the nucleus, where it regulates the transcription of target genes by directly binding to exogenous xenobiotic response elements in the promoter regions (138). AhR activated by TCDD was revealed to regulate the expression of DNA methyltransferase in zebrafish juveniles (139). Jiang et al (140) also found that EOM from PM2.5 was able to activate AhR, resulting in abnormal DNAm in the heart of zebrafish juveniles. Regarding the specific process of DNAm, PM2.5 exposure caused an upregulation of the levels of DNA methyltransferase 1 (DNMT1) in the lungs of mice; however, the level of DNMT1 was found to be downregulated in zebrafish embryos (140,141). Trace elements such as As, Pb, cadmium (Cd) and mercury carried by PM2.5 particles are capable of penetrating the placenta (142), and this phenomenon has been revealed to cause alterations in placental DNAm (143,144). Supplementing the diet with folate was revealed to alleviate EOM-induced DNAm changes, thereby protecting zebrafish embryos against the cardiac developmental toxicity of PM2.5 (140); this probably occurred since folate can act as a methyl donor to affect the expression of DNAm-associated genes (145).
Exposure to air pollutants may alter epigenetic modifications such as DNAm, which, in turn, may affect inflammation, disease development and the risk of deterioration. Exposure to several air pollutants associated with transportation, including PM2.5, black carbon, ozone, nitrogen oxides and PAHs, leads to a decrease in DNAm. This may be due to both the reduced expression of methionine adenosyl-transferase 1A and single carbon metabolism efficiency mediated by oxidative species, resulting in a scarcity of the methyl donor of S-adenosylmethionine that is required for establishing and maintaining DNAm (146). Goodson et al (147) found that in utero exposure to diesel exhaust (DE) induced a decreased level of DNAm in the first exon of GM6307, suggesting that DE can affect the developing heart by altering epigenetic patterns. The mechanism(s) through which PM2.5 exposure leads to DNAm changes, however, have yet to be fully elucidated. It has been revealed that exposure to DE increases the production of ROS (148), which, in turn, interact with DNA, thereby oxidizing methyl-cytosine to hydroxymethyl-cytosine (HMC). HMC has been revealed to prevent the binding of methyl binding proteins (MBPs) to methylated cytosine (149), which prevents normal chromatin silencing from occurring at these sites. In addition, 8-oxoguanine produced by guanine oxidative damage was also found to inhibit the binding of MBPs, thereby hindering the silencing of chromatin regions (149).
PM2.5 has also been revealed to disrupt DNAm profiles (150), probably resulting in an exacerbation of the oxidative and inflammatory responses following PM2.5 exposure. PM2.5 inhalation exerts acute effects on DNAm in the promoter regions of genes that are associated with mitochondrial function and oxidative metabolism (151). Although mitochondria possess their own genetic material that differs from nuclear DNA, the majority of mitochondrial proteins are encoded by the nuclear genome. Exposure to PM2.5 has been revealed to cause a marked alteration in the DNAm of nuclear genes in the mitochondrial pathway, suggesting that mitochondria form the primary target of PM2.5. DNAm is a modifiable biochemical process, and supplementing the diet with B vitamins to ensure that methylation takes place has become an attractive means of drug intervention to counteract the loss of DNAm of inflammatory genes caused by PM2.5 (152). In addition, supplementing B vitamins may also minimize DNA hypermethylation to a great extent.
Histone acetylation modification. Histone acetylation modification is an important topic for epigenetic research. Unlike DNAm, the effects of histone modifications on gene expression may vary, depending on specific chemical modifications (153). Abnormal histone modifications associated with exposure to various environmental chemicals may lead to a large number of diseases, including cardiovascular diseases. For example, the histone modifications H3K9me2 and H3K9ac were revealed to be associated with As exposure, increasing the risk of several cardiovascular diseases (154). In addition, Zhang et al (155) demonstrated an association between the H3K36me3 modification and exposure to PAHs and DNA damage, suggesting that the involvement of specific histone modifications in PAHs results in an induction of DNA damage responses. The processes of histone acetylation and deacetylation are considered to provide an important regulatory mechanism for mediating cardiovascular development and myocardial injury. Histone deacetylation has been demonstrated to participate in the regulation of gene transcription under stress or pathological conditions (156). Histone acetylation has an important role in myocardial hypertrophy events that are induced by PM2.5 exposure. Significantly increased protein levels of acetylated H3K9 were observed in the hearts of mice exposed to PM2.5, which led to an upregulation of hypertrophic transcription factors (75). In summary, the imbalances between histone methylation and demethylation, as well as between acetylation and deacetylation, that are caused by PM2.5 exposure are considered to increase the likelihood of cardiac dysplasia and cardiovascular system-associated diseases.
Among the histone acetyltransferases (HATs), p300 is closely associated with the transcriptional regulation of cardiac development (157). SIRT3, the third type of histone deacetylase, is able to inhibit the OS response and promote the tricarboxylic acid cycle, which has the effect of enhancing myocardial ATP energy supply and contraction, as well as regulating the energy metabolism balance (158). Knockout of SIRT3 was revealed to lead to myocardial mitochondrial dysfunction and cardiac dysfunction (159). Furthermore, the abnormal expression of HATs and HDAC led to imbalanced histone acetylation modifications, giving rise to cardiac developmental disorders (160). Exposure to PM2.5 in the uterus is known to lead to cardiac hypertrophy in adulthood. P300/CREB binding protein mediated histone acetylation modification may exert an important role in the upregulation of thickening transcription factors, such as GATA binding protein 4 (GATA4) and Mef2c. To date, the mechanism(s) of PM2.5-induced histone modification are poorly understood. Environmental chemicals may directly alter histone methyltransferases or demethylases. For example, Ni exposure has been revealed to inhibit the activity of lysine-specific demethylase 3A by binding and substituting Fe2+ ions, thereby increasing H3K9me2 modification (161).
ERS
The endoplasmic reticulum performs a crucial role in terms of protein synthesis and folding, and post-translational modifications. The disruption of endoplasmic reticulum function may lead to accumulated unfolded or misfolded proteins in the lumen, which activates the UPR, a complex intracellular signaling pathway aimed at restoring protein balance. The endoplasmic reticulum is closely associated with normal development and homeostasis of the internal environment, and it has a crucial role in cardiac development and function (162). Zhu et al (163) found that Cd exposure increased ERS in myocardial tissue and primary cardiomyocytes, which was manifested in elevated levels of stress-associated genes. Impaired cardiac contractility and prolonged diastolic duration have been revealed to be common pathological features of the ERS-stimulated heart (164). Previous studies have also suggested that PM2.5 is capable to induce ERS (165,166); however, the mechanism(s) underlying PM2.5-induced ERS, and its role in cardiac development, has yet to be elucidated. EOM from PM2.5 was revealed to induce AhR-mediated ROS production in zebrafish embryonic hearts (17,55,167). In addition, OS induces ERS through disrupting the normal processes of protein folding/transport and altering Ca2+ homeostasis (168-170). On the other hand, ERS was also demonstrated to increase the content of ROS, and to induce OS (171). Early-stage embryos are highly susceptible to oxidative damage, and excessive ROS is considered one of the causative agents for CHD (53,54); therefore, PM2.5 may induce ERS through AhR-mediated ROS overproduction, thereby inducing cardiac developmental toxicity via oxidative damage.
Cardiac development is a coordinated process depending on the subtle balance among cell proliferation, apoptosis and differentiation. It is well established that long-term or severe ERS can lead to cell apoptosis, with C/EBP homologous protein (CHOP) being recognized as one of the most important mediators. The expression of CHOP may be upregulated through activating all three ERS sensors, namely: Activating transcription factor 6, protein kinase RNA like endoplasmic reticulum kinase and inositol requiring enzyme 1α. As a transcription factor, CHOP induces cell apoptosis through downregulating members of the antiapoptotic Bcl protein family and increasing the level of endoplasmic reticulum oxidoreductin 1α. EOM was reported to induce apoptosis of zebrafish embryonic cardiomyocytes, although the increased level of apoptosis was attenuated via inhibiting AhR activity or ROS production (55,172); furthermore, ERS was found to have a key role in this process.
Autophagy
Autophagy is crucial for heart development. Numerous autophagic defects are known to be associated with cardiovascular diseases, including atherosclerosis and cardiomyopathy (172). Autophagy has an important role in the process of cardiac remodeling, including the morphogenesis of cardiac tissues and their eventual differentiation into cardiomyocytes. Atg5-deficient mice were demonstrated to have abnormal heart valves and separated ventricular (173). In a zebrafish cardiac development model, knocking down the core autophagy genes resulted in various defects, including cardiac blood-flow defects and atrial enlargement, among other defects (174). In addition, knocking down these autophagy genes resulted in profound changes in the levels of developmental genes, including certain key transcription factors that are necessary for cardiac development. The knockdown of these genes also led to the accumulation of dead cells in the developing heart, demonstrating the necessity of autophagic clearance of dead cells for normal cardiac remodeling. Cardiac-specific Atg5 deficiency in adult mice was revealed to lead to mitochondrial aggregation and ventricular dilation, demonstrating the vital role that autophagy has in cardiac cell development and homeostasis (175).
As a common heavy metal adsorbed on PM2.5, Cd has been revealed to induce autophagy through a variety of mechanisms, including the ROS-dependent signaling pathway. Cd can disrupt the electron transport chain in mitochondria, especially via binding to the Q0 site of cytochrome b on complex III, leading to an accumulation of semi-ubiquinone. As an unstable molecule that easily transfers electrons to molecular oxygen, semi-ubiquinone results in the formation of superoxide and OS (176). Secondly, there is the ERS pathway: Ca2+ is an important signaling molecule for ERS-induced autophagy (177). ERS leads to the release of Ca2+ from the endoplasmic reticulum into the cytoplasm (178), thereby activating various kinases that are involved in the autophagy signaling pathway, including mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (177). Ca2+ on the outer surface of the endoplasmic reticulum membrane is closely associated with the initiation of autophagosome formation (179). Thirdly, there is the mTOR pathway: Cd activates the AKT/mTOR pathway, thus initiating autophagy, which thereby induces various diseases (180). Finally, there are the Beclin-1 and Bcl-2 family pathways: In the presence of Cd, the increased release of Ca2+ in the endoplasmic reticulum leads to a separation of Beclin-1 from Bcl-2, which thereby activates cellular autophagy (181).
As another cellular protective mechanism for UPR, autophagy contributes to the degradation of the accumulated unfolded or misfolded proteins in the endoplasmic reticulum, thereby restoring endoplasmic reticulum homeostasis and further improving the overall cell survival rate (182). However, autophagy is also a ‘double-edged sword’ since excessive autophagy may promote cell death through excessive self-digestion and the degradation of essential cellular components (183), thereby bringing about embryonic developmental toxicity (184). Autophagy exerts important roles in the processes of cellular and tissue balance, specialization, tissue differentiation and organogenesis (185); in addition, inflammation, ERS and autophagy are closely associated, and these processes have been revealed to interact with each other (186).
Apoptosis
As the principal means of cell death, apoptosis has an important role in maintaining cellular homeostasis. Abnormal apoptosis can give rise to various diseases, including cardiovascular diseases. Apoptosis in mammalian cells is mainly triggered through two pathways: The endogenous (intrinsic) pathway initiated by mitochondria, and the exogenous (extrinsic) pathway initiated by death receptors. The former is controlled by the Bcl-2 family of proteins, whereas the latter involves members of the TNF family of proteins, with the resultant signaling cascade (187). Apoptosis induced by exposure to PM2.5 has a participatory role in a series of signaling pathways, including the MAPK (133) and PI3K/Akt (188) pathways, with caspase-3 serving as a vital biomarker in this process (189). Yang et al (190) demonstrated that the mitochondria-mediated apoptosis pathway has a key role in the PM2.5-induced toxicity of AC16 cardiomyocytes, leading to cardiac dysfunction. The mitochondrial pathway mainly activates caspase-9 by releasing cytochrome c into the cytoplasm, triggering downstream cascade reactions, and ultimately activating downstream caspase-3(191). However, in zebrafish, 2,3-bromofluoranthene derived from PAHs is able to induce apoptosis of vascular endothelial cells and cardiac toxicity through both pathways simultaneously (192). In addition, dysregulation of cellular Ca2+ homeostasis may also lead to cardiomyocyte apoptosis. Ca2+ is one of the most important signal-transduction systems in cells, and a low intracellular Ca2+ concentration is a prerequisite for normal cellular function. After PM2.5 has entered the circulatory system, it leads to an increase in the intracellular Ca2+ concentration, and an overload of Ca2+ will lead to DNA degradation, free radical production and protein kinase activation, ultimately leading to cell apoptosis (193).
Apoptosis can be induced by DNA damage and is crucial for normal cardiac development (194,195). Previous studies have suggested that excessive production of ROS during early embryonic development in zebrafish may lead to DNA damage and cell apoptosis (196-198). Ren et al (55) found that the levels of 8-hydroxydeoxyguanosine (8-OHdG) and cH2AX were raised in the embryonic hearts of zebrafish exposed to EOM, although these increases were significantly reduced in the presence of the ROS scavenger, NAC. This further demonstrated that NAC is able to attenuate EOM-induced apoptosis in zebrafish embryonic cardiomyocyte. Bcl2 binding component 3 (BBC3), a member of the Bcl-2 family, is an important participant in apoptosis (199). BBC3 is localized at the mitochondria under apoptotic stimulations, leading to mitochondria-mediated intrinsic cell apoptosis (200). Traf4a, a zebrafish homolog for human TNF receptor-associated factor 4, is also involved in the regulation of apoptosis (201,202). TRAF4 is also essential for development and can regulate ROS generation by stabilizing NADPH oxidase complexes (203). Knocking down BBC3 or TRAF4 leads to the termination of EOM-induced excessive ROS production and apoptosis in zebrafish embryonic hearts, suggesting that both genes are required for this process. Therefore, overexpression of these two genes may exacerbate cardiac abnormalities in zebrafish juveniles induced by EOM derived from PM2.5.
However, it is necessary to further investigate whether other forms of cell death besides apoptosis and autophagic death, such as ferroptosis, may be associated with the cardiac developmental toxicity induced by PM2.5 exposure, since PM2.5 exposure results in excessive amount of ROS, and severely damaged mitochondria release large amount of Fe, thereby inducing ferroptosis.
AhR signaling
AhR, an essential ligand-activated transcription factor for the cytochrome P450 pathway, controls the expression of genes such as CYP1A1, CYP1B1 and CYP1A2 in the cytochrome P450 family (204). AhR can be activated by numerous environmental pollutants, including PM2.5 (55). Following activation, AhR is dissociated from binding its ligands and enters the nucleus, forming a dimer with AhR nuclear transport protein, subsequently binding with enhancers to form heterologous reaction elements that are involved in the regulation of the expression of cytochrome P450 family genes. Employing the P19 cell line as an in vitro model, Chen et al (167) found that exposure to EOM derived from PM2.5 for 2 days led to an inhibition of cardiac differentiation for the next 14 days, demonstrating the persistent adverse effects of PM2.5 on cardiac development. Mechanistically, AhR mediates the inhibitory effects of EOM on P19 cell cardiac differentiation, probably through dysregulation of cell proliferation, altering the normal processes of Wnt signaling, and inducing breaks of DNA double strands.
It has been revealed that EOM derived from PM2.5 activates the AhR signaling pathway, leading to cardiac abnormalities in zebrafish embryos (17,205). AhR performs an essential role in the cardiac development of fish, mammals, and other organisms. Activation of AhR can impair the cardiac differentiation of human embryonic stem cells (ESCs) (206-208). Considering that PAHs [such as BaK and benzo(a)pyrene] in EOM are strong AhR agonists, and that AhR signaling is activated following exposure to EOM, it may be inferred that AhR mediates EOM-induced cardiac developmental toxicity. The AhR repressor (AhRR) forms a negative feedback loop with AhR (209). Two types of AhRR analogs (Ahrra and Ahrrb) exist in zebrafish, and knocking down Ahrrb (but not Ahrra) was revealed to enhance the inducive effects of the AhR agonist TCDD on the CYP1 superfamily genes (210). Therefore, the AhR inhibitor, CH223191, may inhibit the AhR signaling pathway by inducing Ahrrb expression. In EOM-treated zebrafish, the mRNA levels of the most important AhR subtype, Ahr2, were found to remain unchanged, suggesting that the EOM activation of AhR may be based on conformational changes, rather than on mRNA level changes (211).
As a typical type of PAHs, exposure to TCDD impairs the cardiac differentiation of ESCs, and this impairment is mainly mediated by AhR. The generation of cardiomyocytes was most significantly inhibited in the case of human ESCs (and not mouse ESCs) exposed to TCDD during the ESC stage. By contrast, in the absence of TCDD, AhR is significantly inhibited in mouse ESCs, which decreases the expression of numerous pluripotent genes (212). In addition, ESC cardiac differentiation was found to be suppressed by TCDD exposure during embryonic formation via disrupting activin, bone morphogenetic protein and the Wnt signaling pathway, and through altering the expression of homologous cassette transcription factors (213-216). These differences suggest that human and mouse ESCs exhibit different susceptibility to TCDD toxicity, possibly due to species-specific differential expression patterns of AhR and its cofactors (217). Furthermore, AHR may regulate differential target genes in different species or cells (218,219). Therefore, similarly to TCDD, PM2.5 can inhibit the activation of mesodermal genes through AhR binding, interfering with the differentiation and development of normal cells, and thereby inhibiting mesodermal differentiation. Jiang et al (220) also found that PM2.5 can activate the PI3K/akt2/mammalian target of rapamycin complex 1 signaling pathway through AhR/ROS induced PTEN inhibition, leading to activation of the mitochondria-mediated intrinsic apoptotic pathway and Wnt signaling inhibition, resulting in heart defects in zebrafish juveniles. It has been documented that folate supplementation during pregnancy helps to resist PM2.5-induced cardiac developmental toxicity via targeting the AhR and Wnt/β catenin signaling pathways (205). This provides theoretical support for alleviating PM2.5-induced cardiac developmental toxicity.
Wnt signaling
The Wnt/β-catenin signaling pathway has an essential role in the cardiac development of vertebrates (221), and its activation may induce cardiac specification in the early developmental stages, although this may be suppressed later (221). As a core transcription factor of the typical Wnt signaling pathway, β-catenin is able to regulate the expression of key genes in cardiac development (222). Chen et al (167) found that the mRNA and protein levels of β-catenin were downregulated in cells exposed to EOM derived from PM2.5, suggesting a role of Wnt signaling in EOM-exerted cardiotoxic effects. It is well established that crosstalk exists between the AhR and Wnt/β-catenin signaling pathways (223,224), since activated AhR can antagonize β-catenin in colon cancer cells and zebrafish embryos (224). The Wnt/b-catenin signaling pathway has also been revealed to be crucial for embryonic cardiac development. In the absence of Wnt, cytoplasmic β-catenin is phosphorylated and degraded by a disruption complex composed of adenomatosis polysaccharide coli, Axin and glycogen synthase kinase-3β. Upon Wnt stimulation, cytoplasmic β-catenin is translocated into the nucleus, where it activates the transcription of genes essential for cardiac specification, such as Nkx2.5 and Sox9 (224-226). CH223191 and the Wnt/β-catenin activator, CHIR, were found to rescue the most of the EOM-induced cardiac defects, suggesting the involvement of the AhR and Wnt/β-catenin signaling pathways in cardiac developmental toxicity resulting from PM2.5 exposure, and therefore the feasibility of employing AhR or Wnt/β-catenin antagonists to prevent the cardiac developmental toxicity from occurring (17). The typical Wnt/β-catenin signaling pathway regulates multiple steps in cardiac differentiation (167). The activation of Wnt signaling is crucial for both the formation of the mesoderm in the early stage of development as well as the morphogenesis of cardiac valve formation in later ones (227). Previous studies have demonstrated the inhibition of Wnt signaling by EOM in the embryonic heart of zebrafish (17,205), and both treatment with the ERS inhibitor 4-phenylbutyric acid and CHOP knockdown significantly attenuated these inhibitory effects, probably by means of either affecting β-catenin expression or inhibiting T cell factor (228).
DNA damage
Heavy metals and PAHs derived from PM2.5 can either individually or synergistically disrupt the double-helix structure of DNA, leading to DNA damage. Valavanidis et al (229) identified a positive correlation between DNA reactivity and the concentration of total PAHs and transition metals. The expression and methylation of 8-oxoguanine glycosylase (OGG1) were found to be associated with the ability of PAHs to induce oxidative DNA damage (230). Epigenetic changes fulfill an important role in the regulation of PAH-induced DNA damage. H3K79 di-methylation (H3K79me2) is essential for DNA damage repair, and Zhang et al (231) found that exposure to PAHs reduced its overall level, revealing that it was probably serving as a marker for cellular homeostasis disruption. H3K79me2-deficient cells are more susceptible to benzopyrene-induced DNA damage than are normal cells. Improper methylation of H3K79me2 can lead to low efficiency in DNA damage repair. Therefore, after long-term exposure to PAHs, abnormal H3K79me2 may lead to genomic instability and accumulation of DNA mutations, thereby causing DNA damage. Zhao et al (232) found that PM2.5 and PAHs cause significant activation of the DNA damage-susceptibility gene GADD153, resulting in a reduction of the expression of the DNA-repair genes, human MutT homolog 1 (MTH1) and X-ray repair cross complementing 1, and this inhibitory effect exceeds the clearance effect of OGG1 on damaged DNA, thereby increasing the risk of cardiac DNA damage.
OS, ionizing radiation and chemical reagents are all capable of causing DNA damage (233). Components of water-soluble PM2.5 extracts are more likely to induce DNA oxidative damage compared with organic compounds (234). OS-induced DNA damage has been revealed to be a key mechanism of action in urban PM2.5 pollution (235). Excessive ROS generated by pollutants induces OS, thereby mediating DNA damage in the mouse heart (236). The organic components and transition metals (including Fe, Cu, Ni and Zn) in PM2.5 can directly generate ROS (237), which either directly causes DNA deamination and base oxidation, or indirectly induces base alkylation through lipid peroxidation (238). Therefore, PM2.5-induced OS can lead to DNA damage. DNA damage is usually associated with cell apoptosis (239). Both ROS- or RNS-mediated DNA damage and redox-mediated inhibition of DNA-damage response proteins may lead to changes in DNA structure, thereby activating DNA-repair signaling. The latter can regulate the activities of certain apoptosis factors, further demonstrating the close association between DNA damage and cell apoptosis (240). In human or animal cells, heavy metals derived from PM2.5 can cause various types of DNA damage, including chain breakage, diminishing the activity of endonuclease III, and damaging guanine glycosidase-sensitive sites (49). Interestingly, antioxidants and ROS scavengers can significantly block the DNA damage resulting from PM2.5 exposure.
DNA damage can also cause cell-cycle arrest and induce apoptosis, and this may extensively disrupt the potential of progenitor cells, thereby impairing cardiac development (241). There is a large body of evidence suggesting that environmental pollutants, including PM2.5, may attack DNA by means of OS (45). Excessive ROS production during zebrafish embryonic development has been revealed to lead to DNA damage and apoptosis (196). Elevated levels of 8-OHdG and γ-h2ax were observed in the zebrafish embryonic heart, although these were significantly circumvented by treatment with the ROS scavenge, NAC. However, NAC could not completely reverse the DNA-damage signaling processes induced by PM2.5, suggesting that OS is only partly responsible for causing the DNA damage (242). The cardiac DNA damage caused by PM2.5 is most likely to be involved in multiple signaling pathways and synergistic effects of multiple molecules, and further in-depth exploration in this regard is required.
Ca2+ homeostasis disorder
Ca2+ is essential for cardiac automation, electrical conduction, excitation transcription coupling and maintenance of vascular tone. It is not only necessary for cardiovascular contraction and relaxation, but it also serves a crucial role as a second messenger in signal-transduction pathways (243). A Ca2+ imbalance can lead to various types of cardiomyopathy, and Ca2+ homeostasis is often considered a key factor in heart disease. Therefore, accurate Ca2+ signaling is crucial for maintaining cardiac function. Exposure to PM2.5 preferentially affects the expression of Ca2+ signaling-associated genes in human pluripotent stem cell-derived cardiomyocytes, thereby increasing the likelihood of arrhythmia (244). PM2.5-mediated OS and Ca2+ influx in endothelial cells cause cellular damage, ultimately leading to cell death (245). PM2.5 exposure may also increase the concentration of intracellular free Ca2+ ions by altering the expression of Ca2+ channel-associated proteins in the mouse heart (246). Ca2+ also stimulates the assembly of contractile cytoskeleton structures in developing cells. The endoplasmic reticulum is known to have Ca2+ channels which play a crucial role in Ca2+ regulation (247). An increase in OS will lead to oxidative inactivation of endoplasmic reticulum Ca2+-ATPase, thereby increasing cytoplasmic Ca2+ levels (248). Numerous details of the mechanism underlying PM2.5-mediated intracellular Ca2+ imbalance in OS have been elucidated (246).
Cd can induce lipid abnormalities through ERS and Ca2+ imbalance (249). In addition, Cd disrupts Ca2+ homeostasis, affects vital genes associated with Ca2+ channels, and leads to excessive Ca2+ in the cytoplasm. Following exposure to Cd, the IrxA cluster, Mefs and Tbxs family transcription factors were found to be downregulated, suggesting the impairment of cardiac transcription, abnormal expression of cardiac markers (TnnT2, TnnC1, Gata4, Gata6 and Nkx2-5), and the inhibition of cardiomyocyte maturation and differentiation (163).
4. Conclusion
Given the enormous environmental harm caused by PM2.5, society in its entirety can take certain actions to combat the damaging effects, including governmental policies, urban planning, technology and raising social ecological awareness. The strategies for regulating PM2.5 include reducing dust emissions during construction, transportation and other processes (including through dust removal and filtration), controlling vehicle and industrial emissions (through promoting hybrid or electric vehicles), and transitioning from traditional energy sources to renewable energy sources (such as hydro, solar, geothermal, wind and nuclear energy). One of the most important aspects is to reduce fossil fuel-associated PM2.5, as fossil fuel combustion occupies a central role in the health impacts associated with PM2.5. PM2.5 air pollution derived from fossil fuel combustion has been linked to over 10 million premature deaths (250). These ubiquitous emissions from fossil fuel combustion are one of the biggest contributors to the adverse effects of PM2.5 on health. The particles from fossil fuel combustion contain abundant transition metals [such as Ni, vanadium (V), Fe and Cu] that readily participate in redox reactions that generate OS. At the same time, sulfur (S), which is PM2.5-adsorbent, increases the bioavailability of the transition metals, greatly enhancing the possibility of fossil fuel-associated PM2.5 being a causative agent of OS and endangering overall health (251). In 2019, environmental air pollution caused ~7 million deaths worldwide (252). With the intensification of climate change, environmental air pollution is worsening. Since 1990, the number of deaths caused by environmental air pollution has increased by 51%, and continues to rise (253). Climate change poses a serious threat to human health, with the primary driving factor determined to be the sharp increase in greenhouse gas emissions that are caused by extensive fossil fuel combustion. A total of ~85% of air particulate pollution is caused by fuel combustion, and almost all air pollution is associated with sulfur oxides and nitrogen oxides (254). Therefore, reducing the use of fossil fuels, and the dependence on fossil fuels, is of great significance for optimizing the health benefits of mitigating climate change.
With the intensification of global air pollution, exposure to PM2.5 has been found to correlate closely with the incidence of CHD. The reported molecular mechanisms underlying the PM2.5-exposure-induced cardiac developmental toxicity mainly include: interference with genes related to cardiac development, dysfunction of genes associated with heart function, OS, inflammation, mitochondrial impairment, epigenetic modification, ERS, autophagy, apoptosis, AhR signaling, Wnt signaling, DNA damage and disorders of Ca2+ homeostasis (Fig. 1). Due to the complexity, diversity and unclear toxicity attributed to PM2.5, continuing investigations on cardiac development toxicity derived from PM2.5 exposure inevitably face challenges, and it is necessary to further elucidate the mechanisms underlying PM2.5-induced CHD in our future studies.
As a key factor in cardiac development and homeostasis, AhR signaling fulfills a key role in cardiac development. The interruption of AhR function during development can lead to potential cardiac developmental toxicity, and this comprises a number of dysregulated signaling pathways that participate in cardiac development, function and metabolism. At the same time, AhR is also a target for environmental factors that may disrupt the homeostasis of AhR, laying the foundation for CHD. Due to the widespread presence of AhR antagonists and methyl donors such as flavonoids, curcumin and betaine, that are found in daily foods, these may serve as favorable candidates for addressing the abnormal activation of AhR and changes in m6A RNA methylation derived from PM2.5 exposure. Moreover, phytochemicals are able to alleviate the adverse effects of PM2.5 exposure on human health, predominantly via inhibiting OS, ERS and Fe deposition, which subsequently alleviates inflammatory reactions, along with regulating autophagy. Taken together, these findings provide a potential approach for therapeutic intervention with regard to the cardiac developmental toxicity caused by PM2.5 exposure.
Although mitochondria are the most important source of ROS, increasing evidence has suggested that other organelles as the potential sources of ROS, such as the endoplasmic reticulum, peroxisomes and cell membranes, are also important. Therefore, understanding the sources of ROS is of great significance for targeted therapy, dosage selection and pollution control. Considering the high energy consumption of the heart and the role of mitochondria as the body's energy production factory, mitochondria may be a promising therapeutic target for the prevention and treatment of CHD. Given the deepening understanding of mitochondrial biology, the widespread application of large-scale experimental animal models, and the rapid development of new scientific and technological advancements, mitochondrial medicine may become a realistic therapeutic option in the near future.
The molecular mechanisms underlying the toxicity of PM2.5 exposure to cardiac development are markedly more complex than were previously considered, and there are numerous remaining issues to be addressed; for example, how interactions between various cells (such as crosstalk among endothelial, smooth muscle and immune cells) may have a role in this process, and what key factors and signaling pathways are involved in the cardiac developmental toxicity that is induced by PM2.5 exposure. It remains unclear how these signaling pathways interact with each other, and how the concentration and duration of PM2.5 exposure affect them. These issues retain their significance, and merit further in-depth research. In addition, the chemical composition of PM2.5 is complex, with different sources and toxic effects, and the components of PM2.5 vary differentially across different regions and seasons. Therefore, it is difficult to form systematic research conclusions. In addition, further investigations are needed to determine whether the toxic effects of PM2.5 on cardiac development are mediated solely with PM2.5 functioning as a carrier, or whether PM2.5 interacts with the toxic substances it carries. Therefore, the interactions among different components of PM2.5 and their combined effects with other air pollutants should be also explored. Evidently, it is necessary to perform additional studies on the spatiotemporal distribution characteristics and physicochemical properties of PM2.5, and analyze its effects on cardiac development at different stages, locations and levels, including an assessment of the various physicochemical components.
In summary, the present review elaborates the potential molecular mechanisms underlying the cardiac developmental toxicity induced by gestational PM2.5 (including some of its specific components) exposure, and the complexity presents numerous challenges and opportunities for future investigations. Understanding the interplay of various signaling pathways during this process, alongside the concentration and duration of PM2.5 exposure, will be crucial for advancing our knowledge in this field. In addition, exploring the individual and synergistical cardiac developmental toxicity effects induced by differential PM2.5 components will be vital for developing effective intervention measures and regulatory strategies. In order to deepen our understanding of the cardiac developmental toxicity induced by PM2.5, future researches should focus on longitudinal studies evaluating the long-term effects of early exposure on the cardiac outcomes. This will provide insights into potential interventions to mitigate these effects. Addressing these multifaceted challenges will provide supports for public health policies to reduce exposure to PM2.5 and improve population health outcomes. Ultimately, a comprehensive understanding of PM2.5's toxicological effects will contribute to the scientific community and empower policymakers to implement effective strategies safeguarding public health, particularly among vulnerable populations.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
The data generated in the present study are included in the figures and/or tables of this article.
Authors' contributions
XM, WD and ZS conceived and designed the study. XM and WD collected the literature. XM, WD and ZS analyzed the literature, and drafted and reviewed the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Dominici F, Greenstone M and Sunstein CR: Science and regulation. Particulate matter matters. Science. 344:257–259. 2014.PubMed/NCBI View Article : Google Scholar | |
Thurston GD, Ahn J, Cromar KR, Shao Y, Reynolds HR, Jerrett M, Lim CC, Shanley R, Park Y and Hayes RB: Ambient particulate matter air pollution exposure and mortality in the NIH-AARP diet and health cohort. Environ Health Perspect. 124:484–490. 2016.PubMed/NCBI View Article : Google Scholar | |
Huang Q, Chi Y, Deng J, Liu Y, Lu Y, Chen J and Dong S: Fine particulate matter 2.5 exerted its toxicological effect by regulating a new layer, long non-coding RNA. Sci Rep. 7(9392)2017.PubMed/NCBI View Article : Google Scholar | |
Long JF, Waldman WJ, Kristovich R, Williams M, Knight D and Dutta PK: Comparison of ultrastructural cytotoxic effects of carbon and carbon/iron particulates on human monocyte-derived macrophages. Environ Health Perspect. 113:170–174. 2005.PubMed/NCBI View Article : Google Scholar | |
Samek L, Furman L, Mikrut M, Regiel-Futyra A, Macyk W, Stochel G and van Eldik R: Chemical composition of submicron and fine particulate matter collected in Krakow, Poland. Consequences for the APARIC project. Chemosphere. 187:430–439. 2017.PubMed/NCBI View Article : Google Scholar | |
Mesquita SR, van Drooge BL, Reche C, Guimarães L, Grimalt JO, Barata C and Piña B: Toxic assessment of urban atmospheric particle-bound PAHs: Relevance of composition and particle size in Barcelona (Spain). Environ Pollut. 184:555–562. 2014.PubMed/NCBI View Article : Google Scholar | |
Wang L, Luo D, Liu X, Zhu J, Wang F, Li B and Li L: Effects of PM2.5 exposure on reproductive system and its mechanisms. Chemosphere. 264(128436)2021.PubMed/NCBI View Article : Google Scholar | |
Hoffman JIE, Kaplan S and Liberthson RR: Prevalence of congenital heart disease. Am Heart J. 147:425–439. 2004.PubMed/NCBI View Article : Google Scholar | |
Olson EN: Gene regulatory networks in the evolution and development of the heart. Science. 313:1922–1927. 2006.PubMed/NCBI View Article : Google Scholar | |
Li M, Li J, Wei C, Lu Q, Tang X, Erickson SW, MacLeod SL and Hobbs CA: A three-way interaction among maternal and fetal variants contributing to congenital heart defects. Ann Hum Genet. 80:20–31. 2016.PubMed/NCBI View Article : Google Scholar | |
Hu CY, Huang K, Fang Y, Yang XJ, Ding K, Jiang W, Hua XG, Huang DY, Jiang ZX and Zhang XJ: Maternal air pollution exposure and congenital heart defects in offspring: A systematic review and meta-analysis. Chemosphere. 253(126668)2020.PubMed/NCBI View Article : Google Scholar | |
Huang CC, Chen BY, Pan SC, Ho YL and Guo YL: Prenatal exposure to PM2.5 and congenital heart diseases in Taiwan. Sci Total Environ. 655:880–886. 2019.PubMed/NCBI View Article : Google Scholar | |
Li D, Xu W, Qiu Y, Pan F, Lou H, Li J, Jin Y, Wu T, Pan L, An J, et al: Maternal air pollution exposure and neonatal congenital heart disease: A multi-city cross-sectional study in eastern China. Int J Hyg Environ Health. 240(113898)2022.PubMed/NCBI View Article : Google Scholar | |
Zhang Q, Sun S, Sui X, Ding L, Yang M, Li C, Zhang C, Zhang X, Hao J, Xu Y, et al: Associations between weekly air pollution exposure and congenital heart disease. Sci Total Environ. 757(143821)2021.PubMed/NCBI View Article : Google Scholar | |
Jiang Q, Zhang C, Chen S, Shi L, Li DC, Lv N, Cui L, Chen Y and Zheng Y: Particulate matter 2.5 induced developmental cardiotoxicity in chicken embryo and hatchling. Front Pharmacol. 11(841)2020.PubMed/NCBI View Article : Google Scholar | |
Wang H, Peng X, Cao F, Wang Y, Shi H, Lin S, Zhong W and Sun J: Cardiotoxicity and mechanism of particulate matter 2.5 (PM2.5) exposure in offspring rats during pregnancy. Med Sci Monit. 23:3890–3896. 2017.PubMed/NCBI View Article : Google Scholar | |
Zhang H, Yao Y, Chen Y, Yue C, Chen J, Tong J, Jiang Y and Chen T: Crosstalk between AhR and wnt/β-catenin signal pathways in the cardiac developmental toxicity of PM2.5 in zebrafish embryos. Toxicology. 355-356:31–38. 2016.PubMed/NCBI View Article : Google Scholar | |
Lage K, Greenway SC, Rosenfeld JA, Wakimoto H, Gorham JM, Segrè AV, Roberts AE, Smoot LB, Pu WT, Pereira AC, et al: Genetic and environmental risk factors in congenital heart disease functionally converge in protein networks driving heart development. Proc Natl Acad Sci USA. 109:14035–14040. 2012.PubMed/NCBI View Article : Google Scholar | |
Yan R, Ma D, Liu Y, Wang R, Fan L, Yan Q, Chen C, Wang W, Ren Z, Ku T, et al: Developmental toxicity of fine particulate matter: Multifaceted exploration from epidemiological and laboratory perspectives. Toxics. 12(274)2024.PubMed/NCBI View Article : Google Scholar | |
Feng S, Huang F, Zhang Y, Feng Y, Zhang Y, Cao Y and Wang X: The pathophysiological and molecular mechanisms of atmospheric PM2.5 affecting cardiovascular health: A review. Ecotoxicol Environ Saf. 249(114444)2023.PubMed/NCBI View Article : Google Scholar | |
Liang C, Ding R, Sun Q, Liu S, Sun Z and Duan J: An overview of adverse outcome pathway links between PM2.5 exposure and cardiac developmental toxicity. Environ Health. 2:105–113. 2024.PubMed/NCBI View Article : Google Scholar | |
Agay-Shay K, Friger M, Linn S, Peled A, Amitai Y and Peretz C: Air pollution and congenital heart defects. Environ Res. 124:28–34. 2013.PubMed/NCBI View Article : Google Scholar | |
Girguis MS, Strickland MJ, Hu X, Liu Y, Bartell SM and Vieira VM: Maternal exposure to traffic-related air pollution and birth defects in Massachusetts. Environ Res. 146:1–9. 2016.PubMed/NCBI View Article : Google Scholar | |
Lavigne E, Lima I, Hatzopoulou M, Van Ryswyk K, Decou ML, Luo W, van Donkelaar A, Martin RV, Chen H, Stieb DM, et al: Spatial variations in ambient ultrafine particle concentrations and risk of congenital heart defects. Environ Int. 130(104953)2019.PubMed/NCBI View Article : Google Scholar | |
Schembari A, Nieuwenhuijsen MJ, Salvador J, de Nazelle A, Cirach M, Dadvand P, Beelen R, Hoek G, Basagaña X and Vrijheid M: Traffic-related air pollution and congenital anomalies in Barcelona. Environ Health Perspect. 122:317–323. 2014.PubMed/NCBI View Article : Google Scholar | |
Zhang B, Liang S, Zhao J, Qian Z, Bassig BA, Yang R, Zhang Y, Hu K, Xu S, Zheng T and Yang S: Maternal exposure to air pollutant PM2.5 and PM10 during pregnancy and risk of congenital heart defects. J Expo Sci Environ Epidemiol. 26:422–427. 2016.PubMed/NCBI View Article : Google Scholar | |
Jenkins KJ, Correa A, Feinstein JA, Botto L, Britt AE, Daniels SR, Elixson M, Warnes CA and Webb CL: American Heart Association Council on Cardiovascular Disease in the Young. Noninherited risk factors and congenital cardiovascular defects: Current knowledge: A scientific statement from the American heart association council on cardiovascular disease in the Young: Endorsed by the American academy of pediatrics. Circulation. 115:2995–3014. 2007.PubMed/NCBI View Article : Google Scholar | |
Lassi ZS, Imam AM, Dean SV and Bhutta ZA: Preconception care: Caffeine, smoking, alcohol, drugs and other environmental chemical/radiation exposure. Reprod Health. 11 (Suppl 3)(S6)2014.PubMed/NCBI View Article : Google Scholar | |
Chang YC, Lin YT, Jung CR, Chen KW and Hwang BF: Maternal exposure to fine particulate matter and congenital heart defects during preconception and pregnancy period: A cohort-based case-control study in the Taiwan maternal and child health database. Environ Res. 231(116154)2023.PubMed/NCBI View Article : Google Scholar | |
Wu X, Pan B, Liu L, Zhao W, Zhu J, Huang X and Tian J: In utero exposure to PM2.5 during gestation caused adult cardiac hypertrophy through histone acetylation modification. J Cell Biochem. 120:4375–4384. 2019.PubMed/NCBI View Article : Google Scholar | |
Watt AJ, Battle MA, Li J and Duncan SA: GATA4 is essential for formation of the proepicardium and regulates cardiogenesis. Proc Natl Acad Sci USA. 101:12573–12578. 2004.PubMed/NCBI View Article : Google Scholar | |
Jiang SY, Xu M and Zhang YY: Role of GATA-4 in cardiac development and remodeling. Sheng Li Ke Xue Jin Zhan. 39:302–306. 2008.PubMed/NCBI(In Chinese). | |
Akazawa H and Komuro I: Roles of cardiac transcription factors in cardiac hypertrophy. Circ Res. 92:1079–1088. 2003.PubMed/NCBI View Article : Google Scholar | |
Morimoto T, Hasegawa K, Wada H, Kakita T, Kaburagi S, Yanazume T and Sasayama S: Calcineurin-GATA4 pathway is involved in beta-adrenergic agonist-responsive endothelin-1 transcription in cardiac myocytes. J Biol Chem. 276:34983–34989. 2001.PubMed/NCBI View Article : Google Scholar | |
Yang Y, Ruan Z, Wang X, Yang Y, Mason TG, Lin H and Tian L: Short-term and long-term exposures to fine particulate matter constituents and health: A systematic review and meta-analysis. Environ Pollut. 247:874–882. 2019.PubMed/NCBI View Article : Google Scholar | |
Sancini G, Farina F, Battaglia C, Cifola I, Mangano E, Mantecca P, Camatini M and Palestini P: Health risk assessment for air pollutants: alterations in lung and cardiac gene expression in mice exposed to Milano winter fine particulate matter (PM2.5). PLoS One. 9(e109685)2014.PubMed/NCBI View Article : Google Scholar | |
Qin G, Xia J, Zhang Y, Guo L, Chen R and Sang N: Ambient fine particulate matter exposure induces reversible cardiac dysfunction and fibrosis in juvenile and older female mice. Part Fibre Toxicol. 15(27)2018.PubMed/NCBI View Article : Google Scholar | |
Qi Z, Song Y, Ding Q, Liao X, Li R, Liu G, Tsang S and Cai Z: Water soluble and insoluble components of PM2.5 and their functional cardiotoxicities on neonatal rat cardiomyocytes in vitro. Ecotoxicol Environ Saf. 168:378–387. 2019.PubMed/NCBI View Article : Google Scholar | |
Shaffer F and Ginsberg JP: An overview of heart rate variability metrics and norms. Front Public Health. 5(258)2017.PubMed/NCBI View Article : Google Scholar | |
Wagner JG, Kamal AS, Morishita M, Dvonch JT, Harkema JR and Rohr AC: PM2.5-induced cardiovascular dysregulation in rats is associated with elemental carbon and temperature-resolved carbon subfractions. Part Fibre Toxicol. 11(25)2014.PubMed/NCBI View Article : Google Scholar | |
Chen R, Qiao L, Li H, Zhao Y, Zhang Y, Xu W, Wang C, Wang H, Zhao Z, Xu X, et al: Fine particulate matter constituents, nitric oxide synthase DNA methylation and exhaled nitric oxide. Environ Sci Technol. 49:11859–11865. 2015.PubMed/NCBI View Article : Google Scholar | |
Tanwar V, Adelstein JM, Grimmer JA, Youtz DJ, Sugar BP and Wold LE: PM2.5 exposure in utero contributes to neonatal cardiac dysfunction in mice. Environ Pollut. 230:116–124. 2017.PubMed/NCBI View Article : Google Scholar | |
Grant AO: Cardiac ion channels. Circ Arrhythm Electrophysiol. 2:185–194. 2009.PubMed/NCBI View Article : Google Scholar | |
Park KH, Choi YJ, Min WK, Lee SH, Kim J, Jeong SH, Lee JH, Choi BM and Kim S: Particulate matter induces arrhythmia-like cardiotoxicity in zebrafish embryos by altering the expression levels of cardiac development- and ion channel-related genes. Ecotoxicol Environ Saf. 263(115201)2023.PubMed/NCBI View Article : Google Scholar | |
Gualtieri M, Longhin E, Mattioli M, Mantecca P, Tinaglia V, Mangano E, Proverbio MC, Bestetti G, Camatini M and Battaglia C: Gene expression profiling of A549 cells exposed to Milan PM2.5. Toxicol Lett. 209:136–145. 2012.PubMed/NCBI View Article : Google Scholar | |
Kouassi KS, Billet S, Garçon G, Verdin A, Diouf A, Cazier F, Djaman J, Courcot D and Shirali P: Oxidative damage induced in A549 cells by physically and chemically characterized air particulate matter (PM2.5) collected in Abidjan, Côte d'Ivoire. J Appl Toxicol. 30:310–320. 2010.PubMed/NCBI View Article : Google Scholar | |
Briedé JJ, De Kok TMCM, Hogervorst JGF, Moonen EJC, Op Den Camp CLB and Kleinjanst JCS: Development and application of an electron spin resonance spectrometry method for the determination of oxygen free radical formation by particulate matter. Environ Sci Technol. 39:8420–8426. 2005.PubMed/NCBI View Article : Google Scholar | |
Gehling W, Khachatryan L and Dellinger B: Hydroxyl radical generation from environmentally persistent free radicals (EPFRs) in PM2.5. Environ Sci Technol. 48:4266–4272. 2014.PubMed/NCBI View Article : Google Scholar | |
Longhin E, Holme JA, Gutzkow KB, Arlt VM, Kucab JE, Camatini M and Gualtieri M: Cell cycle alterations induced by urban PM2.5 in bronchial epithelial cells: characterization of the process and possible mechanisms involved. Part Fibre Toxicol. 10(63)2013.PubMed/NCBI View Article : Google Scholar | |
Huang Q, Zhang J, Peng S, Tian M, Chen J and Shen H: Effects of water soluble PM2.5 extracts exposure on human lung epithelial cells (A549): A proteomic study. J Appl Toxicol. 34:675–687. 2014.PubMed/NCBI View Article : Google Scholar | |
Kannan S, Misra DP, Dvonch JT and Krishnakumar A: Exposures to airborne particulate matter and adverse perinatal outcomes: A biologically plausible mechanistic framework for exploring potential effect modification by nutrition. Environ Health Perspect. 114:1636–1642. 2006.PubMed/NCBI View Article : Google Scholar | |
Feng S, Gao D, Liao F, Zhou F and Wang X: The health effects of ambient PM2.5 and potential mechanisms. Ecotoxicol Environ Saf. 128:67–74. 2016.PubMed/NCBI View Article : Google Scholar | |
Li SY, Sigmon VK, Babcock SA and Ren J: Advanced glycation endproduct induces ROS accumulation, apoptosis, MAP kinase activation and nuclear O-GlcNAcylation in human cardiac myocytes. Life Sci. 80:1051–1056. 2007.PubMed/NCBI View Article : Google Scholar | |
Moazzen H, Lu X, Ma NL, Velenosi TJ, Urquhart BL, Wisse LJ, Gittenberger-de Groot AC and Feng Q: N-Acetylcysteine prevents congenital heart defects induced by pregestational diabetes. Cardiovasc Diabetol. 13(46)2014.PubMed/NCBI View Article : Google Scholar | |
Ren F, Ji C, Huang Y, Aniagu S, Jiang Y and Chen T: AHR-mediated ROS production contributes to the cardiac developmental toxicity of PM2.5 in zebrafish embryos. Sci Total Environ. 719(135097)2020.PubMed/NCBI View Article : Google Scholar | |
Wang L, He X, Szklarz GD, Bi Y, Rojanasakul Y and Ma Q: The aryl hydrocarbon receptor interacts with nuclear factor erythroid 2-related factor 2 to mediate induction of NAD(P)H:quinoneoxidoreductase 1 by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Arch Biochem Biophys. 537:31–38. 2013.PubMed/NCBI View Article : Google Scholar | |
Rousseau ME, Sant KE, Borden LR, Franks DG, Hahn ME and Timme-Laragy AR: Regulation of Ahr signaling by Nrf2 during development: Effects of Nrf2a deficiency on PCB126 embryotoxicity in zebrafish (Danio rerio). Aquat Toxicol. 167:157–171. 2015.PubMed/NCBI View Article : Google Scholar | |
Dalton TP, Puga A and Shertzer HG: Induction of cellular oxidative stress by aryl hydrocarbon receptor activation. Chem Biol Interact. 141:77–95. 2002.PubMed/NCBI View Article : Google Scholar | |
Elbekai RH and El-Kadi AOS: The role of oxidative stress in the modulation of aryl hydrocarbon receptor-regulated genes by As3+, Cd2+, and Cr6+. Free Radic Biol Med. 39:1499–1511. 2005.PubMed/NCBI View Article : Google Scholar | |
Mohammadi-Bardbori A, Omidi M and Arabnezhad MR: Impact of CH223191-induced mitochondrial dysfunction on its Aryl hydrocarbon receptor agonistic and antagonistic activities. Chem Res Toxicol. 32:691–697. 2019.PubMed/NCBI View Article : Google Scholar | |
Kopf PG and Walker MK: 2,3,7,8-Tetrachlorodibenzo-p-dioxin increases reactive oxygen species production in human endothelial cells via induction of cytochrome P4501A1. Toxicol Appl Pharmacol. 245:91–99. 2010.PubMed/NCBI View Article : Google Scholar | |
Zangar RC, Davydov DR and Verma S: Mechanisms that regulate production of reactive oxygen species by cytochrome P450. Toxicol Appl Pharmacol. 199:316–331. 2004.PubMed/NCBI View Article : Google Scholar | |
Zhou B, Wang X, Li F, Wang Y, Yang L, Zhen X and Tan W: Mitochondrial activity and oxidative stress functions are influenced by the activation of AhR-induced CYP1A1 overexpression in cardiomyocytes. Mol Med Rep. 16:174–180. 2017.PubMed/NCBI View Article : Google Scholar | |
Pei Y, Jiang R, Zou Y, Wang Y, Zhang S, Wang G, Zhao J and Song W: Effects of fine particulate matter (PM2.5) on systemic oxidative stress and cardiac function in ApoE(-/-) mice. Int J Environ Res Public Health. 13(484)2016.PubMed/NCBI View Article : Google Scholar | |
Yang JL, Lu JY, Zhang MS, Qin G and Li CP: Involvement of heme oxygenase in PM2.5-toxicity in human umbilical vein endothelial cells. Zhonghua Xin Xue Guan Bing Za Zhi. 41:955–961. 2013.PubMed/NCBI(In Chinese). | |
Medzhitov R: Origin and physiological roles of inflammation. Nature. 454:428–435. 2008.PubMed/NCBI View Article : Google Scholar | |
Zhao J, Gao Z, Tian Z, Xie Y, Xin F, Jiang R, Kan H and Song W: The biological effects of individual-level PM(2.5) exposure on systemic immunity and inflammatory response in traffic policemen. Occup Environ Med. 70:426–431. 2013.PubMed/NCBI View Article : Google Scholar | |
Bekki K, Ito T, Yoshida Y, He C, Arashidani K, He M, Sun G, Zeng Y, Sone H, Kunugita N and Ichinose T: PM2.5 collected in China causes inflammatory and oxidative stress responses in macrophages through the multiple pathways. Environ Toxicol Pharmacol. 45:362–369. 2016.PubMed/NCBI View Article : Google Scholar | |
Shi Q, Zhao L, Xu C, Zhang L and Zhao H: High molecular weight hyaluronan suppresses macrophage M1 polarization and enhances IL-10 production in PM2.5-induced lung inflammation. Molecules. 24(1766)2019.PubMed/NCBI View Article : Google Scholar | |
Chen W, Liu Y, Chen J, Song Y, You M and Yang G: Long-term co-exposure DBP and BaP causes imbalance in liver macrophages polarization via activation of notch signaling regulated by miR-34a-5p in rats. Chem Biol Interact. 359(109919)2022.PubMed/NCBI View Article : Google Scholar | |
You M, Song Y, Chen J, Liu Y, Chen W, Cen Y, Zhao X, Tao Z and Yang G: Combined exposure to benzo(a)pyrene and dibutyl phthalate aggravates pro-inflammatory macrophage polarization in spleen via pyroptosis involving cathepsin B. Sci Total Environ. 881(163460)2023.PubMed/NCBI View Article : Google Scholar | |
Nicolás-Ávila JA, Lechuga-Vieco AV, Esteban-Martinez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, Rubio-Ponce A, Li JL, Balachander A, Quintana JA, et al: A network of macrophages supports mitochondrial homeostasis in the heart. Cell. 183:94–109.e23. 2020.PubMed/NCBI View Article : Google Scholar | |
Ueland T, Gullestad L, Nymo SH, Yndestad A, Aukrust P and Askevold ET: Inflammatory cytokines as biomarkers in heart failure. Clin Chim Acta. 443:71–77. 2015.PubMed/NCBI View Article : Google Scholar | |
Frati G, Schirone L, Chimenti I, Yee D, Biondi-Zoccai G, Volpe M and Sciarretta S: An overview of the inflammatory signalling mechanisms in the myocardium underlying the development of diabetic cardiomyopathy. Cardiovasc Res. 113:378–388. 2017.PubMed/NCBI View Article : Google Scholar | |
Li R, Zhao Y, Shi J, Zhao C, Xie P, Huang W, Yong T and Cai Z: Effects of PM2.5 exposure in utero on heart injury, histone acetylation and GATA4 expression in offspring mice. Chemosphere. 256(127133)2020.PubMed/NCBI View Article : Google Scholar | |
Ma XN, Li RQ, Xie JL, Li SH, Li JW and Yan XX: PM2.5-induced inflammation and myocardial cell injury in rats. Eur Rev Med Pharmacol Sci. 25:6670–6677. 2021.PubMed/NCBI View Article : Google Scholar | |
Fröde-Saleh TS and Calixto JB: Synergistic antiinflammatory effect of NF-kappaB inhibitors and steroidal or non steroidal antiinflammatory drugs in the pleural inflammation induced by carrageenan in mice. Inflamm Res. 49:330–337. 2000.PubMed/NCBI View Article : Google Scholar | |
Ryu YS, Kang KA, Piao MJ, Ahn MJ, Yi JM, Hyun YM, Kim SH, Ko MK, Park CO and Hyun JW: Particulate matter induces inflammatory cytokine production via activation of NFκB by TLR5-NOX4-ROS signaling in human skin keratinocyte and mouse skin. Redox Biol. 21(101080)2019.PubMed/NCBI View Article : Google Scholar | |
Li H, Shi Y, Wang X, Li P, Zhang S, Wu T, Yan Y, Zhan Y, Ren Y, Rong X, et al: Piceatannol alleviates inflammation and oxidative stress via modulation of the Nrf2/HO-1 and NF-κB pathways in diabetic cardiomyopathy. Chem Biol Interact. 310(108754)2019.PubMed/NCBI View Article : Google Scholar | |
Jiao Y, Wang S, Jiang L, Sun X, Li J, Liu X, Yao X, Zhang C, Wang N, Deng H and Yang G: 2-Undecanone protects against fine particles-induced heart inflammation via modulating Nrf2/HO-1 and NF-κB pathways. Environ Toxicol. 37:1642–1652. 2022.PubMed/NCBI View Article : Google Scholar | |
Zhang Y, Ji X, Ku T and Sang N: Inflammatory response and endothelial dysfunction in the hearts of mice co-exposed to SO2, NO2, and PM2.5. Environ Toxicol. 31:1996–2005. 2016.PubMed/NCBI View Article : Google Scholar | |
Chen M, Qin X, Qiu L, Chen S, Zhou H, Xu Y, Hu Z, Zhang Y, Cao Q and Ying Z: Concentrated ambient PM2.5-induced inflammation and endothelial dysfunction in a murine model of neural IKK2 deficiency. Environ Health Perspect. 126(027003)2018.PubMed/NCBI View Article : Google Scholar | |
Hu B, Tong B, Xiang Y, Li SR, Tan ZX, Xiang HX, Fu L, Wang H, Zhao H and Xu DX: Acute 1-NP exposure induces inflammatory responses through activating various inflammatory signaling pathways in mouse lungs and human A549 cells. Ecotoxicol Environ Saf. 189(109977)2020.PubMed/NCBI View Article : Google Scholar | |
Duan S, Wang N, Huang L, Zhao Y, Shao H, Jin Y, Zhang R, Li C, Wu W, Wang J and Feng F: NLRP3 inflammasome activation is associated with PM2.5-induced cardiac functional and pathological injury in mice. Environ Toxicol. 34:1246–1254. 2019.PubMed/NCBI View Article : Google Scholar | |
Bevan GH, Al-Kindi SG, Brook RD, Münzel T and Rajagopalan S: Ambient air pollution and atherosclerosis: Insights into dose, time, and mechanisms. Arterioscler Thromb Vasc Biol. 41:628–637. 2021.PubMed/NCBI View Article : Google Scholar | |
West AP: Mitochondrial dysfunction as a trigger of innate immune responses and inflammation. Toxicology. 391:54–63. 2017.PubMed/NCBI View Article : Google Scholar | |
Breda CNS, Davanzo GG, Basso PJ, Saraiva Câmara NO and Moraes-Vieira PMM: Mitochondria as central hub of the immune system. Redox Biol. 26(101255)2019.PubMed/NCBI View Article : Google Scholar | |
Wang G, Zhao J, Jiang R and Song W: Rat lung response to ozone and fine particulate matter (PM2.5) exposures. Environ Toxicol. 30:343–356. 2015.PubMed/NCBI View Article : Google Scholar | |
Niu J, Liberda EN, Qu S, Guo X, Li X, Zhang J, Meng J, Yan B, Li N, Zhong M, et al: The role of metal components in the cardiovascular effects of PM2.5. PLoS One. 8(e83782)2013.PubMed/NCBI View Article : Google Scholar | |
Cardozo AK, Ortis F, Storling J, Feng YM, Rasschaert J, Tonnesen M, Van Eylen F, Mandrup-Poulsen T, Herchuelz A and Eizirik DL: Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic beta-cells. Diabetes. 54:452–461. 2005.PubMed/NCBI View Article : Google Scholar | |
Baccarelli A, Wright R, Bollati V, Litonjua A, Zanobetti A, Tarantini L, Sparrow D, Vokonas P and Schwartz J: Ischemic heart disease and stroke in relation to blood DNA methylation. Epidemiology. 21:819–828. 2010.PubMed/NCBI View Article : Google Scholar | |
Hotamisligil GS: Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 140:900–917. 2010.PubMed/NCBI View Article : Google Scholar | |
Bettigole SE and Glimcher LH: Endoplasmic reticulum stress in immunity. Annu Rev Immunol. 33:107–138. 2015.PubMed/NCBI View Article : Google Scholar | |
Song S, Tan J, Miao Y, Li M and Zhang Q: Crosstalk of autophagy and apoptosis: Involvement of the dual role of autophagy under ER stress. J Cell Physiol. 232:2977–2984. 2017.PubMed/NCBI View Article : Google Scholar | |
Ding Q, Qi Y and Tsang SY: Mitochondrial biogenesis, mitochondrial dynamics, and mitophagy in the maturation of cardiomyocytes. Cells. 10(2463)2021.PubMed/NCBI View Article : Google Scholar | |
Hou L, Zhu ZZ, Zhang X, Nordio F, Bonzini M, Schwartz J, Hoxha M, Dioni L, Marinelli B, Pegoraro V, et al: Airborne particulate matter and mitochondrial damage: A cross-sectional study. Environ Health. 9(48)2010.PubMed/NCBI View Article : Google Scholar | |
Xia T, Kovochich M and Nel AE: Impairment of mitochondrial function by particulate matter (PM) and their toxic components: Implications for PM-induced cardiovascular and lung disease. Front Biosci. 12:1238–1246. 2007.PubMed/NCBI View Article : Google Scholar | |
Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM and Kelly DP: Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial biogenesis. J Clin Invest. 106:847–856. 2000.PubMed/NCBI View Article : Google Scholar | |
Meng Z and Liu Y: Cell morphological ultrastructural changes in various organs from mice exposed by inhalation to sulfur dioxide. Inhal Toxicol. 19:543–551. 2007.PubMed/NCBI View Article : Google Scholar | |
Marchini T, Magnani N, D'Annunzio V, Tasat D, Gelpi RJ, Alvarez S and Evelson P: Impaired cardiac mitochondrial function and contractile reserve following an acute exposure to environmental particulate matter. Biochim Biophys Acta. 1830:2545–2552. 2013.PubMed/NCBI View Article : Google Scholar | |
Wang G, Zhen L, Lü P, Jiang R and Song W: Effects of ozone and fine particulate matter (PM2.5) on rat cardiac autonomic nervous system and systemic inflammation. Wei Sheng Yan Jiu. 42:554–560. 2013.PubMed/NCBI(In Chinese). | |
Wang Q and Zhang L, Yuan X, Ou Y, Zhu X, Cheng Z, Zhang P, Wu X, Meng Y and Zhang L: The relationship between the Bcl-2/Bax proteins and the mitochondria-mediated apoptosis pathway in the differentiation of adipose-derived stromal cells into neurons. PLoS One. 11(e0163327)2016.PubMed/NCBI View Article : Google Scholar | |
Zorzano A, Liesa M, Sebastian D, Segales J and Palacin M: Mitochondrial fusion proteins: Dual regulators of morphology and metabolism. Semin Cell Dev Biol. 21:566–574. 2010.PubMed/NCBI View Article : Google Scholar | |
Yang XD, Shi Q, Sun J, Lv Y, Ma Y, Chen C, Xiao K, Zhou W and Dong XP: Aberrant alterations of mitochondrial factors Drp1 and Opa1 in the brains of scrapie experiment rodents. J Mol Neurosci. 61:368–378. 2017.PubMed/NCBI View Article : Google Scholar | |
Westermann B: Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol. 11:872–884. 2010.PubMed/NCBI View Article : Google Scholar | |
Ikeda Y, Sciarretta S, Nagarajan N, Rubattu S, Volpe M, Frati G and Sadoshima J: New insights into the role of mitochondrial dynamics and autophagy during oxidative stress and aging in the heart. Oxid Med Cell Longev. 2014(210934)2014.PubMed/NCBI View Article : Google Scholar | |
Soberanes S, Urich D, Baker CM, Burgess Z, Chiarella SE, Bell EL, Ghio AJ, De Vizcaya-Ruiz A, Liu J, Ridge KM, et al: Mitochondrial complex III-generated oxidants activate ASK1 and JNK to induce alveolar epithelial cell death following exposure to particulate matter air pollution. J Biol Chem. 284:2176–2186. 2009.PubMed/NCBI View Article : Google Scholar | |
Castilho RF, Meinicke AR, Almeida AM, Hermes-Lima M and Vercesi AE: Oxidative damage of mitochondria induced by Fe(II)citrate is potentiated by Ca2+ and includes lipid peroxidation and alterations in membrane proteins. Arch Biochem Biophys. 308:158–163. 1994.PubMed/NCBI View Article : Google Scholar | |
Packer MA, Porteous CM and Murphy MP: Superoxide production by mitochondria in the presence of nitric oxide forms peroxynitrite. Biochem Mol Biol Int. 40:527–534. 1996.PubMed/NCBI View Article : Google Scholar | |
Meyer JN, Leung MCK, Rooney JP, Sendoel A, Hengartner MO, Kisby GE and Bess AS: Mitochondria as a target of environmental toxicants. Toxicol Sci. 134:1–17. 2013.PubMed/NCBI View Article : Google Scholar | |
Rodriguez-Enriquez S, He L and Lemasters JJ: Role of mitochondrial permeability transition pores in mitochondrial autophagy. Int J Biochem Cell Biol. 36:2463–2472. 2004.PubMed/NCBI View Article : Google Scholar | |
Urrutia PJ, Mena NP and Núñez MT: The interplay between iron accumulation, mitochondrial dysfunction, and inflammation during the execution step of neurodegenerative disorders. Front Pharmacol. 5(38)2014.PubMed/NCBI View Article : Google Scholar | |
Liang H and Ward WF: PGC-1alpha: A key regulator of energy metabolism. Adv Physiol Educ. 30:145–151. 2006.PubMed/NCBI View Article : Google Scholar | |
Prakash C and Kumar V: Arsenic-induced mitochondrial oxidative damage is mediated by decreased PGC-1α expression and its downstream targets in rat brain. Chem Biol Interact. 256:228–235. 2016.PubMed/NCBI View Article : Google Scholar | |
Chen J, Zhang M, Aniagu S, Jiang Y and Chen T: PM2.5 induces cardiac defects via AHR-SIRT1-PGC-1α mediated mitochondrial damage. Environ Toxicol Pharmacol. 106(104393)2024.PubMed/NCBI View Article : Google Scholar | |
Chen J, Zhang M, Zou H, Aniagu S, Jiang Y and Chen T: PM2.5 induces mitochondrial dysfunction via AHR-mediated cyp1a1 overexpression during zebrafish heart development. Toxicology. 487(153466)2023.PubMed/NCBI View Article : Google Scholar | |
Cavalli G and Heard E: Advances in epigenetics link genetics to the environment and disease. Nature. 571:489–499. 2019.PubMed/NCBI View Article : Google Scholar | |
Marcho C, Oluwayiose OA and Pilsner JR: The preconception environment and sperm epigenetics. Andrology. 8:924–942. 2020.PubMed/NCBI View Article : Google Scholar | |
Sun T, Wu R and Ming L: The role of m6A RNA methylation in cancer. Biomed Pharmacother. 112(108613)2019.PubMed/NCBI View Article : Google Scholar | |
Liu Q and Gregory RI: RNAmod: An integrated system for the annotation of mRNA modifications. Nucleic Acids Res. 47:W548–W555. 2019.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Li Y, Toth JI, Petroski MD, Zhang Z and Zhao JC: N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol. 16:191–198. 2014.PubMed/NCBI View Article : Google Scholar | |
Liu L, Li H, Hu D, Wang Y, Shao W, Zhong J, Yang S, Liu J and Zhang J: Insights into N6-methyladenosine and programmed cell death in cancer. Mol Cancer. 21(32)2022.PubMed/NCBI View Article : Google Scholar | |
Tang F, Chen L, Gao H, Xiao D and Li X: m6A: An emerging role in programmed cell death. Front Cell Dev Biol. 10(817112)2022.PubMed/NCBI View Article : Google Scholar | |
Yang Y, Shen S, Cai Y, Zeng K, Liu K, Li S, Zeng L, Chen L, Tang J, Hu Z, et al: Dynamic patterns of N6-methyladenosine profiles of messenger RNA correlated with the cardiomyocyte regenerability during the early heart development in mice. Oxid Med Cell Longev. 2021(5537804)2021.PubMed/NCBI View Article : Google Scholar | |
Shen S, Liu K, Li S, Rampes S, Yang Y, Huang Y, Tang J, Xia Z, Ma D and Zhang L: N6-methyladenosine modulates long non-coding RNA in the developing mouse heart. Cell Death Discov. 8(329)2022.PubMed/NCBI View Article : Google Scholar | |
Guo X, Lin Y, Lin Y, Zhong Y, Yu H, Huang Y, Yang J, Cai Y, Liu F, Li Y, et al: PM2.5 induces pulmonary microvascular injury in COPD via METTL16-mediated m6A modification. Environ Pollut. 303(119115)2022.PubMed/NCBI View Article : Google Scholar | |
He X, Zhang L, Liu S, Wang J, Liu Y, Xiong A, Jiang M, Luo L, Ying X and Li G: Methyltransferase-like 3 leads to lung injury by up-regulation of interleukin 24 through N6-methyladenosine-dependent mRNA stability and translation efficiency in mice exposed to fine particulate matter 2.5. Environ Pollut. 308(119607)2022.PubMed/NCBI View Article : Google Scholar | |
Ji C, Tao Y, Li X, Wang J, Chen J, Aniagu S, Jiang Y and Chen T: AHR-mediated m6A RNA methylation contributes to PM2.5-induced cardiac malformations in zebrafish larvae. J Hazard Mater. 457(131749)2023.PubMed/NCBI View Article : Google Scholar | |
Ning J, Du H, Zhang Y, Liu Q, Jiang T, Pang Y, Tian X, Yan L, Niu Y and Zhang R: N6-Methyladenosine modification of CDH1 mRNA promotes PM2.5-induced pulmonary fibrosis via mediating epithelial mesenchymal transition. Toxicol Sci. 185:143–157. 2022.PubMed/NCBI View Article : Google Scholar | |
Batista PJ, Molinie B, Wang J, Qu K, Zhang J, Li L, Bouley DM, Lujan E, Haddad B, Daneshvar K, et al: m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 15:707–719. 2014.PubMed/NCBI View Article : Google Scholar | |
Zhao T, Sun D, Zhao M, Lai Y, Liu Y and Zhang Z: N6-methyladenosine mediates arsenite-induced human keratinocyte transformation by suppressing p53 activation. Environ Pollut. 259(113908)2020.PubMed/NCBI View Article : Google Scholar | |
Zhuang C, Zhuang C, Luo X, Huang X, Yao L, Li J, Li Y, Xiong T, Ye J, Zhang F and Gui Y: N6-methyladenosine demethylase FTO suppresses clear cell renal cell carcinoma through a novel FTO-PGC-1α signalling axis. J Cell Mol Med. 23:2163–2173. 2019.PubMed/NCBI View Article : Google Scholar | |
Cao J, Qin G, Shi R, Bai F, Yang G, Zhang M and Lv J: Overproduction of reactive oxygen species and activation of MAPKs are involved in apoptosis induced by PM2.5 in rat cardiac H9c2 cells. J Appl Toxicol. 36:609–617. 2016.PubMed/NCBI View Article : Google Scholar | |
Dong W, Matsumura F and Kullman SW: TCDD induced pericardial edema and relative COX-2 expression in medaka (Oryzias Latipes) embryos. Toxicol Sci. 118:213–223. 2010.PubMed/NCBI View Article : Google Scholar | |
Shi Y, Zhao T, Yang X, Sun B, Li Y, Duan J and Sun Z: PM2.5-induced alteration of DNA methylation and RNA-transcription are associated with inflammatory response and lung injury. Sci Total Environ. 650:908–921. 2019.PubMed/NCBI View Article : Google Scholar | |
Avilla MN, Malecki KMC, Hahn ME, Wilson RH and Bradfield CA: The Ah receptor: Adaptive metabolism, ligand diversity, and the xenokine model. Chem Res Toxicol. 33:860–879. 2020.PubMed/NCBI View Article : Google Scholar | |
Hahn ME, Karchner SI and Merson RR: Diversity as opportunity: Insights from 600 million years of AHR evolution. Curr Opin Toxicol. 2:58–71. 2017.PubMed/NCBI View Article : Google Scholar | |
Jeuken A, Keser BJG, Khan E, Brouwer A, Koeman J and Denison MS: Activation of the Ah receptor by extracts of dietary herbal supplements, vegetables, and fruits. J Agric Food Chem. 51:5478–5487. 2003.PubMed/NCBI View Article : Google Scholar | |
Aluru N, Kuo E, Helfrich LW, Karchner SI, Linney EA, Pais JE and Franks DG: Developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin alters DNA methyltransferase (dnmt) expression in zebrafish (Danio rerio). Toxicol Appl Pharmacol. 284:142–151. 2015.PubMed/NCBI View Article : Google Scholar | |
Jiang Y, Li J, Ren F, Ji C, Aniagu S and Chen T: PM2.5-induced extensive DNA methylation changes in the heart of zebrafish embryos and the protective effect of folic acid. Environ Pollut. 255(113331)2019.PubMed/NCBI View Article : Google Scholar | |
Soberanes S, Gonzalez A, Urich D, Chiarella SE, Radigan KA, Osornio-Vargas A, Joseph J, Kalyanaraman B, Ridge KM, Chandel NS, et al: Particulate matter air pollution induces hypermethylation of the p16 promoter via a mitochondrial ROS-JNK-DNMT1 pathway. Sci Rep. 2(275)2012.PubMed/NCBI View Article : Google Scholar | |
Al-Saleh I, Shinwari N, Mashhour A, Mohamed Gel D and Rabah A: Heavy metals (lead, cadmium and mercury) in maternal, cord blood and placenta of healthy women. Int J Hyg Environ Health. 214:79–101. 2011.PubMed/NCBI View Article : Google Scholar | |
Maccani JZJ, Koestler DC, Lester B, Houseman EA, Armstrong DA, Kelsey KT and Marsit CJ: Placental DNA methylation related to both infant toenail mercury and adverse neurobehavioral outcomes. Environ Health Perspect. 123:723–729. 2015.PubMed/NCBI View Article : Google Scholar | |
Mohanty AF, Farin FM, Bammler TK, MacDonald JW, Afsharinejad Z, Burbacher TM, Siscovick DS, Williams MA and Enquobahrie DA: Infant sex-specific placental cadmium and DNA methylation associations. Environ Res. 138:74–81. 2015.PubMed/NCBI View Article : Google Scholar | |
Li W, Liu H, Yu M, Zhang X, Zhang Y, Liu H, Wilson JX and Huang G: Folic acid alters methylation profile of JAK-STAT and long-term depression signaling pathways in Alzheimer's disease models. Mol Neurobiol. 53:6548–6556. 2016.PubMed/NCBI View Article : Google Scholar | |
Maghbooli Z, Hossein-Nezhad A, Adabi E, Asadollah-Pour E, Sadeghi M, Mohammad-Nabi S, Zakeri Rad L, Malek Hosseini AA, Radmehr M, Faghihi F, et al: Air pollution during pregnancy and placental adaptation in the levels of global DNA methylation. PLoS One. 13(e0199772)2018.PubMed/NCBI View Article : Google Scholar | |
Goodson JM, Weldy CS, MacDonald JW, Liu Y, Bammler TK, Chien WM and Chin MT: In utero exposure to diesel exhaust particulates is associated with an altered cardiac transcriptional response to transverse aortic constriction and altered DNA methylation. FASEB J. 31:4935–4945. 2017.PubMed/NCBI View Article : Google Scholar | |
Wauters A, Dreyfuss C, Pochet S, Hendrick P, Berkenboom G, van de Borne P and Argacha JF: Acute exposure to diesel exhaust impairs nitric oxide-mediated endothelial vasomotor function by increasing endothelial oxidative stress. Hypertension. 62:352–358. 2013.PubMed/NCBI View Article : Google Scholar | |
Valinluck V, Tsai HH, Rogstad DK, Burdzy A, Bird A and Sowers LC: Oxidative damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding domain (MBD) of methyl-CpG binding protein 2 (MeCP2). Nucleic Acids Res. 32:4100–4108. 2004.PubMed/NCBI View Article : Google Scholar | |
Panni T, Mehta AJ, Schwartz JD, Baccarelli AA, Just AC, Wolf K, Wahl S, Cyrys J, Kunze S, Strauch K, et al: Genome-wide analysis of DNA methylation and fine particulate matter air pollution in three study populations: KORA F3, KORA F4, and the normative aging study. Environ Health Perspect. 124:983–990. 2016.PubMed/NCBI View Article : Google Scholar | |
Wei S, Segura S, Vendrell J, Aviles FX, Lanoue E, Day R, Feng Y and Fricker LD: Identification and characterization of three members of the human metallocarboxypeptidase gene family. J Biol Chem. 277:14954–14964. 2002.PubMed/NCBI View Article : Google Scholar | |
Bellavia A, Urch B, Speck M, Brook RD, Scott JA, Albetti B, Behbod B, North M, Valeri L, Bertazzi PA, et al: DNA hypomethylation, ambient particulate matter, and increased blood pressure: Findings from controlled human exposure experiments. J Am Heart Assoc. 2(e000212)2013.PubMed/NCBI View Article : Google Scholar | |
Shahbazian MD and Grunstein M: Functions of site-specific histone acetylation and deacetylation. Annu Rev Biochem. 76:75–100. 2007.PubMed/NCBI View Article : Google Scholar | |
Chervona Y, Hall MN, Arita A, Wu F, Sun H, Tseng HC, Ali E, Uddin MN, Liu X, Zoroddu MA, et al: Associations between arsenic exposure and global posttranslational histone modifications among adults in Bangladesh. Cancer Epidemiol Biomarkers Prev. 21:2252–2260. 2012.PubMed/NCBI View Article : Google Scholar | |
Zhang Z, Chen L, Xing X, Li D, Gao C, He Z, Li J, Zhu X, Xiao X, Wang S, et al: Specific histone modifications were associated with the PAH-induced DNA damage response in coke oven workers. Toxicol Res (Camb). 5:1193–1201. 2016.PubMed/NCBI View Article : Google Scholar | |
Wang Z, Zhao YT and Zhao TC: Histone deacetylases in modulating cardiac disease and their clinical translational and therapeutic implications. Exp Biol Med (Maywood). 246:213–225. 2021.PubMed/NCBI View Article : Google Scholar | |
Sun H, Yang X, Zhu J, Lv T, Chen Y, Chen G, Zhong L, Li Y, Huang X, Huang G and Tian J: Inhibition of p300-HAT results in a reduced histone acetylation and down-regulation of gene expression in cardiac myocytes. Life Sci. 87:707–714. 2010.PubMed/NCBI View Article : Google Scholar | |
Hu DX, Liu XB, Song WC and Wang JA: Roles of SIRT3 in heart failure: From bench to bedside. J Zhejiang Univ Sci B. 17:821–830. 2016.PubMed/NCBI View Article : Google Scholar | |
Li Y, Ma Y, Song L, Yu L, Zhang L, Zhang Y, Xing Y, Yin Y and Ma H: SIRT3 deficiency exacerbates p53/Parkin-mediated mitophagy inhibition and promotes mitochondrial dysfunction: Implication for aged hearts. Int J Mol Med. 41:3517–3526. 2018.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Miao X, Liu Y, Li F, Liu Q, Sun J and Cai L: Dysregulation of histone acetyltransferases and deacetylases in cardiovascular diseases. Oxid Med Cell Longev. 2014(641979)2014.PubMed/NCBI View Article : Google Scholar | |
Chen H, Giri NC, Zhang R, Yamane K, Zhang Y, Maroney M and Costa M: Nickel ions inhibit histone demethylase JMJD1A and DNA repair enzyme ABH2 by replacing the ferrous iron in the catalytic centers. J Biol Chem. 285:7374–7383. 2010.PubMed/NCBI View Article : Google Scholar | |
Prins D and Michalak M: Endoplasmic reticulum proteins in cardiac development and dysfunction. Can J Physiol Pharmacol. 87:419–425. 2009.PubMed/NCBI View Article : Google Scholar | |
Zhu Y, Guan H, Zhu X, Cai J, Jiao X, Shan J, Li Y, Wu Q and Zhang Z: Astilbin antagonizes developmental cardiotoxicity after cadmium exposure in chicken embryos by inhibiting endoplasmic reticulum stress and maintaining calcium homeostasis. Ecotoxicol Environ Saf. 270(115847)2024.PubMed/NCBI View Article : Google Scholar | |
Minamino T and Kitakaze M: ER stress in cardiovascular disease. J Mol Cell Cardiol. 48:1105–1110. 2010.PubMed/NCBI View Article : Google Scholar | |
Li R, Zhang M, Wang Y, Yung KKL, Su R, Li Z, Zhao L, Dong C and Cai Z: Effects of sub-chronic exposure to atmospheric PM2.5 on fibrosis, inflammation, endoplasmic reticulum stress and apoptosis in the livers of rats. Toxicol Res (Camb). 7:271–282. 2018.PubMed/NCBI View Article : Google Scholar | |
Wang Y and Tang M: PM2.5 induces autophagy and apoptosis through endoplasmic reticulum stress in human endothelial cells. Sci Total Environ. 710(136397)2020.PubMed/NCBI View Article : Google Scholar | |
Chen T, Jin H, Wang H, Yao Y, Aniagu S, Tong J and Jiang Y: Aryl hydrocarbon receptor mediates the cardiac developmental toxicity of EOM from PM2.5 in P19 embryonic carcinoma cells. Chemosphere. 216:372–378. 2019.PubMed/NCBI View Article : Google Scholar | |
de la Harpe A, Beukes N and Frost CL: CBD activation of TRPV1 induces oxidative signaling and subsequent ER stress in breast cancer cell lines. Biotechnol Appl Biochem. 69:420–430. 2022.PubMed/NCBI View Article : Google Scholar | |
Malhotra JD and Kaufman RJ: Endoplasmic reticulum stress and oxidative stress: A vicious cycle or a double-edged sword? Antioxid Redox Signal. 9:2277–2293. 2007.PubMed/NCBI View Article : Google Scholar | |
Ozgur R, Uzilday B, Sekmen AH and Turkan I: The effects of induced production of reactive oxygen species in organelles on endoplasmic reticulum stress and on the unfolded protein response in arabidopsis. Ann Bot. 116:541–553. 2015.PubMed/NCBI View Article : Google Scholar | |
Burgos-Morón E, Abad-Jiménez Z, Marañón AM, Iannantuoni F, Escribano-López I, López-Domènech S, Salom C, Jover A, Mora V, Roldan I, et al: Relationship between oxidative stress, ER stress, and inflammation in type 2 diabetes: The battle continues. J Clin Med. 8(1385)2019.PubMed/NCBI View Article : Google Scholar | |
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, et al: The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med. 10(1088575)2023.PubMed/NCBI View Article : Google Scholar | |
Lavandero S, Chiong M, Rothermel BA and Hill JA: Autophagy in cardiovascular biology. J Clin Invest. 125:55–64. 2015.PubMed/NCBI View Article : Google Scholar | |
Lee E, Koo Y, Ng A, Wei Y, Luby-Phelps K, Juraszek A, Xavier RJ, Cleaver O, Levine B and Amatruda JF: Autophagy is essential for cardiac morphogenesis during vertebrate development. Autophagy. 10:572–587. 2014.PubMed/NCBI View Article : Google Scholar | |
Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, et al: The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 13:619–624. 2007.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Fang J, Leonard SS and Rao KM: Cadmium inhibits the electron transfer chain and induces reactive oxygen species. Free Radic Biol Med. 36:1434–1443. 2004.PubMed/NCBI View Article : Google Scholar | |
Høyer-Hansen M and Jäättelä M: Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ. 14:1576–1582. 2007.PubMed/NCBI View Article : Google Scholar | |
Biagioli M, Pifferi S, Ragghianti M, Bucci S, Rizzuto R and Pinton P: Endoplasmic reticulum stress and alteration in calcium homeostasis are involved in cadmium-induced apoptosis. Cell Calcium. 43:184–195. 2008.PubMed/NCBI View Article : Google Scholar | |
Zheng Q, Chen Y, Chen D, Zhao H, Feng Y, Meng Q, Zhao Y and Zhang H: Calcium transients on the ER surface trigger liquid-liquid phase separation of FIP200 to specify autophagosome initiation sites. Cell. 185:4082–4098.e22. 2022.PubMed/NCBI View Article : Google Scholar | |
Sun M, Jiang Z, Gu P, Guo B, Li J, Cheng S, Ba Q and Wang H: Cadmium promotes colorectal cancer metastasis through EGFR/Akt/mTOR signaling cascade and dynamics. Sci Total Environ. 899(165699)2023.PubMed/NCBI View Article : Google Scholar | |
Lian J, Wu X, He F, Karnak D, Tang W, Meng Y, Xiang D, Ji M, Lawrence TS and Xu L: A natural BH3 mimetic induces autophagy in apoptosis-resistant prostate cancer via modulating Bcl-2-Beclin1 interaction at endoplasmic reticulum. Cell Death Differ. 18:60–71. 2011.PubMed/NCBI View Article : Google Scholar | |
Plácido AI, Pereira CM, Duarte AI, Candeias E, Correia SC, Santos RX, Carvalho C, Cardoso S, Oliveira CR and Moreira PI: The role of endoplasmic reticulum in amyloid precursor protein processing and trafficking: Implications for Alzheimer's disease. Biochim Biophys Acta. 1842:1444–1453. 2014.PubMed/NCBI View Article : Google Scholar | |
Su R, Jin X, Lyu L, Tian J, Amin S and Li Z: The potential immunotoxicity of fine particulate matter based on SD rat spleen. Environ Sci Pollut Res Int. 26:23958–23966. 2019.PubMed/NCBI View Article : Google Scholar | |
Rubiolo JA, López-Alonso H, Martinez P, Millán A, Cagide E, Vieytes MR, Vega FV and Botana LM: Yessotoxin induces ER-stress followed by autophagic cell death in glioma cells mediated by mTOR and BNIP3. Cell Signal. 26:419–432. 2014.PubMed/NCBI | |
Carloni S, Favrais G, Saliba E, Albertini MC, Chalon S, Longini M, Gressens P, Buonocore G and Balduini W: Melatonin modulates neonatal brain inflammation through endoplasmic reticulum stress, autophagy, and miR-34a/silent information regulator 1 pathway. J Pineal Res. 61:370–380. 2016.PubMed/NCBI View Article : Google Scholar | |
Zhang S, Jiang C, Liu H, Guan Z, Zeng Q, Zhang C, Lei R, Xia T, Gao H, Yang L, et al: Fluoride-elicited developmental testicular toxicity in rats: Roles of endoplasmic reticulum stress and inflammatory response. Toxicol Appl Pharmacol. 271:206–215. 2013.PubMed/NCBI View Article : Google Scholar | |
Dagher Z, Garçon G, Billet S, Gosset P, Ledoux F, Courcot D, Aboukais A and Shirali P: Activation of different pathways of apoptosis by air pollution particulate matter (PM2.5) in human epithelial lung cells (L132) in culture. Toxicology. 225:12–24. 2006.PubMed/NCBI View Article : Google Scholar | |
Yang X, Zhao T, Feng L, Shi Y, Jiang J, Liang S, Sun B, Xu Q, Duan J and Sun Z: PM2.5-induced ADRB2 hypermethylation contributed to cardiac dysfunction through cardiomyocytes apoptosis via PI3K/Akt pathway. Environ Int. 127:601–614. 2019.PubMed/NCBI View Article : Google Scholar | |
Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007.PubMed/NCBI View Article : Google Scholar | |
Yang X, Feng L, Zhang Y, Hu H, Shi Y, Liang S, Zhao T, Fu Y, Duan J and Sun Z: Cytotoxicity induced by fine particulate matter (PM2.5) via mitochondria-mediated apoptosis pathway in human cardiomyocytes. Ecotoxicol Environ Saf. 161:198–207. 2018.PubMed/NCBI View Article : Google Scholar | |
Bock FJ and Tait SWG: Mitochondria as multifaceted regulators of cell death. Nat Rev Mol Cell Biol. 21:85–100. 2020.PubMed/NCBI View Article : Google Scholar | |
Su CH, Chen SP, Chen LY, Yang JJ, Lee YC, Lee SS, Chen HH, Ng YY and Kuan YH: 3-Bromofluoranthene-induced cardiotoxicity of zebrafish and apoptosis in the vascular endothelial cells via intrinsic and extrinsic caspase-dependent pathways. Ecotoxicol Environ Saf. 228(112962)2021.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print). | |
Gombedza FC, Shin S, Kanaras YL and Bandyopadhyay BC: Abrogation of store-operated Ca2+ entry protects against crystal-induced ER stress in human proximal tubular cells. Cell Death Discov. 5(124)2019.PubMed/NCBI View Article : Google Scholar | |
Dlamini Z, Tshidino SC and Hull R: Abnormalities in alternative splicing of apoptotic genes and cardiovascular diseases. Int J Mol Sci. 16:27171–27190. 2015.PubMed/NCBI View Article : Google Scholar | |
Yan L, Zhou Y, Yu S, Ji G, Wang L, Liu W and Gu A: 8-Oxoguanine DNA glycosylase 1 (ogg1) maintains the function of cardiac progenitor cells during heart formation in zebrafish. Exp Cell Res. 319:2954–2963. 2013.PubMed/NCBI View Article : Google Scholar | |
Zhao X, Ren X, Zhu R, Luo Z and Ren B: Zinc oxide nanoparticles induce oxidative DNA damage and ROS-triggered mitochondria-mediated apoptosis in zebrafish embryos. Aquat Toxicol. 180:56–70. 2016.PubMed/NCBI View Article : Google Scholar | |
Zhao X, Wang S, Wu Y, You H and Lv L: Acute ZnO nanoparticles exposure induces developmental toxicity, oxidative stress and DNA damage in embryo-larval zebrafish. Aquat Toxicol. 136-137:49–59. 2013.PubMed/NCBI View Article : Google Scholar | |
Zhu L, Dong X, Xie H, Wang J, Wang J, Su J and Yu C: DNA damage and effects on glutathione-S-transferase activity induced by atrazine exposure in zebrafish (Danio rerio). Environ Toxicol. 26:480–488. 2011.PubMed/NCBI View Article : Google Scholar | |
Liu H, Cheng Y, Yang J, Wang W, Fang S, Zhang W, Han B, Zhou Z, Yao H, Chao J and Liao H: BBC3 in macrophages promoted pulmonary fibrosis development through inducing autophagy during silicosis. Cell Death Dis. 8(e2657)2017.PubMed/NCBI View Article : Google Scholar | |
Huang DC and Strasser A: BH3-Only proteins-essential initiators of apoptotic cell death. Cell. 103:839–842. 2000.PubMed/NCBI View Article : Google Scholar | |
Kedinger V, Alpy F, Tomasetto C, Thisse C, Thisse B and Rio MC: Spatial and temporal distribution of the traf4 genes during zebrafish development. Gene Expr Patterns. 5:545–552. 2005.PubMed/NCBI View Article : Google Scholar | |
Sax JK and El-Deiry WS: Identification and characterization of the cytoplasmic protein TRAF4 as a p53-regulated proapoptotic gene. J Biol Chem. 278:36435–36444. 2003.PubMed/NCBI View Article : Google Scholar | |
Ruan X, Zhang R, Li R, Zhu H, Wang Z, Wang C, Cheng Z and Peng H: The research progress in physiological and pathological functions of TRAF4. Front Oncol. 12(842072)2022.PubMed/NCBI View Article : Google Scholar | |
Zhang J, Cui S, Shen L, Gao Y, Liu W, Zhang C and Zhuang S: Promotion of bladder cancer cell metastasis by 2-mercaptobenzothiazole via its activation of Aryl hydrocarbon receptor transcription: Molecular dynamics simulations, cell-based assays, and machine learning-driven prediction. Environ Sci Technol. 56:13254–13263. 2022.PubMed/NCBI View Article : Google Scholar | |
Yue C, Ji C, Zhang H, Zhang LW, Tong J, Jiang Y and Chen T: Protective effects of folic acid on PM2.5-induced cardiac developmental toxicity in zebrafish embryos by targeting AhR and Wnt/β-catenin signal pathways. Environ Toxicol. 32:2316–2322. 2017.PubMed/NCBI View Article : Google Scholar | |
Bello SM, Heideman W and Peterson RE: 2,3,7,8-Tetrachlorodibenzo-p-dioxin inhibits regression of the common cardinal vein in developing zebrafish. Toxicol Sci. 78:258–266. 2004.PubMed/NCBI View Article : Google Scholar | |
Fu H, Wang L, Wang J, Bennett BD, Li JL, Zhao B and Hu G: Dioxin and AHR impairs mesoderm gene expression and cardiac differentiation in human embryonic stem cells. Sci Total Environ. 651:1038–1046. 2019.PubMed/NCBI View Article : Google Scholar | |
Lund AK, Goens MB, Nuñez BA and Walker MK: Characterizing the role of endothelin-1 in the progression of cardiac hypertrophy in aryl hydrocarbon receptor (AhR) null mice. Toxicol Appl Pharmacol. 212:127–135. 2006.PubMed/NCBI View Article : Google Scholar | |
Evans BR, Karchner SI, Franks DG and Hahn ME: Duplicate aryl hydrocarbon receptor repressor genes (ahrr1 and ahrr2) in the zebrafish Danio rerio: Structure, function, evolution, and AHR-dependent regulation in vivo. Arch Biochem Biophys. 441:151–167. 2005.PubMed/NCBI View Article : Google Scholar | |
Jenny MJ, Karchner SI, Franks DG, Woodin BR, Stegeman JJ and Hahn ME: Distinct roles of two zebrafish AHR repressors (AHRRa and AHRRb) in embryonic development and regulating the response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci. 110:426–441. 2009.PubMed/NCBI View Article : Google Scholar | |
Jayasundara N, Van Tiem Garner L, Meyer JN, Erwin KN and Di Giulio RT: AHR2-mediated transcriptomic responses underlying the synergistic cardiac developmental toxicity of PAHs. Toxicol Sci. 143:469–481. 2015.PubMed/NCBI View Article : Google Scholar | |
Ko CI, Fan Y, de Gannes M, Wang Q, Xia Y and Puga A: Repression of the Aryl hydrocarbon receptor is required to maintain mitotic progression and prevent loss of pluripotency of embryonic stem cells. Stem Cells. 34:2825–2839. 2016.PubMed/NCBI View Article : Google Scholar | |
Jiang Y, Wang D, Zhang G, Wang G, Tong J and Chen T: Disruption of cardiogenesis in human embryonic stem cells exposed to trichloroethylene. Environ Toxicol. 31:1372–1380. 2016.PubMed/NCBI View Article : Google Scholar | |
Wang Q, Chen J, Ko CI, Fan Y, Carreira V, Chen Y, Xia Y, Medvedovic M and Puga A: Disruption of aryl hydrocarbon receptor homeostatic levels during embryonic stem cell differentiation alters expression of homeobox transcription factors that control cardiomyogenesis. Environ Health Perspect. 121:1334–1343. 2013.PubMed/NCBI View Article : Google Scholar | |
Carreira VS, Fan Y, Kurita H, Wang Q, Ko CI, Naticchioni M, Jiang M, Koch S, Zhang X, Biesiada J, et al: Disruption of Ah receptor signaling during mouse development leads to abnormal cardiac structure and function in the adult. PLoS One. 10(e0142440)2015.PubMed/NCBI View Article : Google Scholar | |
Wang Q, Kurita H, Carreira V, Ko CI, Fan Y, Zhang X, Biesiada J, Medvedovic M and Puga A: Ah receptor activation by dioxin disrupts activin, BMP, and WNT signals during the early differentiation of mouse embryonic stem cells and inhibits cardiomyocyte functions. Toxicol Sci. 149:346–357. 2016.PubMed/NCBI View Article : Google Scholar | |
Ginis I, Luo Y, Miura T, Thies S, Brandenberger R, Gerecht-Nir S, Amit M, Hoke A, Carpenter MK, Itskovitz-Eldor J and Rao MS: Differences between human and mouse embryonic stem cells. Dev Biol. 269:360–380. 2004.PubMed/NCBI View Article : Google Scholar | |
Dere E, Lee AW, Burgoon LD and Zacharewski TR: Differences in TCDD-elicited gene expression profiles in human HepG2, mouse Hepa1c1c7 and rat H4IIE hepatoma cells. BMC Genomics. 12(193)2011.PubMed/NCBI View Article : Google Scholar | |
Suzuki T and Nohara K: Regulatory factors involved in species-specific modulation of arylhydrocarbon receptor (AhR)-dependent gene expression in humans and mice. J Biochem. 142:443–452. 2007.PubMed/NCBI View Article : Google Scholar | |
Jiang Y, Zhao X, Chen J, Aniagu S and Chen T: PM2.5 induces cardiac malformations via PI3K/akt2/mTORC1 signaling pathway in zebrafish larvae. Environ Pollut. 323(121306)2023.PubMed/NCBI View Article : Google Scholar | |
Ozhan G and Weidinger G: Wnt/β-catenin signaling in heart regeneration. Cell Regen. 4(3)2015.PubMed/NCBI View Article : Google Scholar | |
Ueno S, Weidinger G, Osugi T, Kohn AD, Golob JL, Pabon L, Reinecke H, Moon RT and Murry CE: Biphasic role for Wnt/beta-catenin signaling in cardiac specification in zebrafish and embryonic stem cells. Proc Natl Acad Sci USA. 104:9685–9690. 2007.PubMed/NCBI View Article : Google Scholar | |
Schneider AJ, Branam AM and Peterson RE: Intersection of AHR and Wnt signaling in development, health, and disease. Int J Mol Sci. 15:17852–17885. 2014.PubMed/NCBI View Article : Google Scholar | |
Wincent E, Stegeman JJ and Jönsson ME: Combination effects of AHR agonists and Wnt/β-catenin modulators in zebrafish embryos: Implications for physiological and toxicological AHR functions. Toxicol Appl Pharmacol. 284:163–179. 2015.PubMed/NCBI View Article : Google Scholar | |
Blache P, van de Wetering M, Duluc I, Domon C, Berta P, Freund JN, Clevers H and Jay P: SOX9 is an intestine crypt transcription factor, is regulated by the Wnt pathway, and represses the CDX2 and MUC2 genes. J Cell Biol. 166:37–47. 2004.PubMed/NCBI View Article : Google Scholar | |
Liu Z, Li T, Liu Y, Jia Z, Li Y, Zhang C, Chen P, Ma K, Affara N and Zhou C: WNT signaling promotes Nkx2.5 expression and early cardiomyogenesis via downregulation of Hdac1. Biochim Biophys Acta. 1793:300–311. 2009.PubMed/NCBI View Article : Google Scholar | |
Lin X and Xu X: Distinct functions of Wnt/beta-catenin signaling in KV development and cardiac asymmetry. Development. 136:207–217. 2009.PubMed/NCBI View Article : Google Scholar | |
Chiu CS, Tsai CH, Hsieh MS, Tsai SC, Jan YJ, Lin WY, Lai DW, Wu SM, Hsing HY, Arbiser JL and Sheu ML: Exploiting Honokiol-induced ER stress CHOP activation inhibits the growth and metastasis of melanoma by suppressing the MITF and β-catenin pathways. Cancer Lett. 442:113–125. 2019.PubMed/NCBI View Article : Google Scholar | |
Valavanidis A, Fiotakis K and Vlachogianni T: Airborne particulate matter and human health: Toxicological assessment and importance of size and composition of particles for oxidative damage and carcinogenic mechanisms. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 26:339–362. 2008.PubMed/NCBI View Article : Google Scholar | |
Fu Y, Niu Y, Pan B, Liu Y, Zhang B, Li X, Yang A, Nie J, Wang R and Yang J: OGG1 methylation mediated the effects of cell cycle and oxidative DNA damage related to PAHs exposure in Chinese coke oven workers. Chemosphere. 224:48–57. 2019.PubMed/NCBI View Article : Google Scholar | |
Zhang Z, Xing X, Jiang S, Qiu C, Mo Z, Chen S, Chen L, Wang Q, Xiao Y, Dong G, et al: Global H3K79 di-methylation mediates DNA damage response to PAH exposure in Chinese coke oven workers. Environ Pollut. 268(115956)2021.PubMed/NCBI View Article : Google Scholar | |
Zhao L, Zhang L, Chen M, Dong C, Li R and Cai Z: Effects of ambient atmospheric PM2.5, 1-nitropyrene and 9-nitroanthracene on DNA damage and oxidative stress in hearts of rats. Cardiovasc Toxicol. 19:178–190. 2019.PubMed/NCBI View Article : Google Scholar | |
Wang W, Li Y, Liu X, Jin M, Du H, Liu Y, Huang P, Zhou X, Yuan L and Sun Z: Multinucleation and cell dysfunction induced by amorphous silica nanoparticles in an L-02 human hepatic cell line. Int J Nanomedicine. 8:3533–3541. 2013.PubMed/NCBI View Article : Google Scholar | |
Gutiérrez-Castillo ME, Roubicek DA, Cebrián-García ME, De Vizcaya-Ruíz A, Sordo-Cedeño M and Ostrosky-Wegman P: Effect of chemical composition on the induction of DNA damage by urban airborne particulate matter. Environ Mol Mutagen. 47:199–211. 2006.PubMed/NCBI View Article : Google Scholar | |
Risom L, Møller P and Loft S: Oxidative stress-induced DNA damage by particulate air pollution. Mutat Res. 592:119–137. 2005.PubMed/NCBI View Article : Google Scholar | |
Zhang P, Yi LH, Meng GY, Zhang HY, Sun HH and Cui LQ: Apelin-13 attenuates cisplatin-induced cardiotoxicity through inhibition of ROS-mediated DNA damage and regulation of MAPKs and AKT pathways. Free Radic Res. 51:449–459. 2017.PubMed/NCBI View Article : Google Scholar | |
Ayres JG, Borm P, Cassee FR, Castranova V, Donaldson K, Ghio A, Harrison RM, Hider R, Kelly F, Kooter IM, et al: Evaluating the toxicity of airborne particulate matter and nanoparticles by measuring oxidative stress potential-a workshop report and consensus statement. Inhal Toxicol. 20:75–99. 2008.PubMed/NCBI View Article : Google Scholar | |
Meira LB, Bugni JM, Green SL, Lee CW, Pang B, Borenshtein D, Rickman BH, Rogers AB, Moroski-Erkul CA, McFaline JL, et al: DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J Clin Invest. 118:2516–2525. 2008.PubMed/NCBI View Article : Google Scholar | |
Zhang X, Jiang Y and Yang J: p53-independent signaling pathway in DNA damage-induced cell apoptosis. Zhejiang Da Xue Xue Bao Yi Xue Ban. 42:217–223. 2013.PubMed/NCBI(In Chinese). | |
De Zio D, Cianfanelli V and Cecconi F: New insights into the link between DNA damage and apoptosis. Antioxid Redox Signal. 19:559–571. 2013.PubMed/NCBI View Article : Google Scholar | |
Lorda-Diez CI, Solis-Mancilla ME, Sanchez-Fernandez C, Garcia-Porrero JA, Hurle JM and Montero JA: Cell senescence, apoptosis and DNA damage cooperate in the remodeling processes accounting for heart morphogenesis. J Anat. 234:815–829. 2019.PubMed/NCBI View Article : Google Scholar | |
Huang Y, Tao Y, Cai C, Chen J, Ji C, Aniagu S, Jiang Y and Chen T: Using immunofluorescence to Detect PM2.5-induced DNA damage in zebrafish embryo hearts. J Vis Exp, 2021. | |
Cartwright EJ, Oceandy D, Austin C and Neyses L: Ca2+ signalling in cardiovascular disease: The role of the plasma membrane calcium pumps. Sci China Life Sci. 54:691–698. 2011.PubMed/NCBI View Article : Google Scholar | |
Cai C, Huang J, Lin Y, Miao W, Chen P, Chen X, Wang J and Chen M: Particulate matter 2.5 induced arrhythmogenesis mediated by TRPC3 in human induced pluripotent stem cell-derived cardiomyocytes. Arch Toxicol. 93:1009–1020. 2019.PubMed/NCBI View Article : Google Scholar | |
Wang Y, Wu T and Tang M: Ambient particulate matter triggers dysfunction of subcellular structures and endothelial cell apoptosis through disruption of redox equilibrium and calcium homeostasis. J Hazard Mater. 394(122439)2020.PubMed/NCBI View Article : Google Scholar | |
Dong L, Sun W, Li F, Shi M, Meng X, Wang C, Meng M, Tang W, Liu H, Wang L and Song L: The harmful effects of acute PM2.5 exposure to the heart and a novel preventive and therapeutic function of CEOs. Sci Rep. 9(3495)2019.PubMed/NCBI View Article : Google Scholar | |
Xu R, Cao JW, Xu TC, Liu TJ, Zhu MR and Guo MY: Selenium deficiency induced inflammation and apoptosis via NF-κB and MAPKs pathways in muscle of common carp (Cyprinus carpio L.). Fish Shellfish Immunol. 138(108847)2023.PubMed/NCBI View Article : Google Scholar | |
Nowak WN, Deng J, Ruan XZ and Xu Q: Reactive oxygen species generation and atherosclerosis. Arterioscler Thromb Vasc Biol. 37:e41–e52. 2017.PubMed/NCBI View Article : Google Scholar | |
Rajakumar S, Bhanupriya N, Ravi C and Nachiappan V: Endoplasmic reticulum stress and calcium imbalance are involved in cadmium-induced lipid aberrancy in Saccharomyces cerevisiae. Cell Stress Chaperones. 21:895–906. 2016.PubMed/NCBI View Article : Google Scholar | |
Vohra K, Vodonos A, Schwartz J, Marais EA, Sulprizio MP and Mickley LJ: Global mortality from outdoor fine particle pollution generated by fossil fuel combustion: Results from GEOS-chem. Environ Res. 195(110754)2021.PubMed/NCBI View Article : Google Scholar | |
Maciejczyk P, Chen LC and Thurston G: The role of fossil fuel combustion metals in PM air pollution health associations. Atmosphere. 12(1086)2021. | |
Fuller R, Landrigan PJ, Balakrishnan K, Bathan G, Bose-O'Reilly S, Brauer M, Caravanos J, Chiles T, Cohen A, Corra L, et al: Pollution and health: A progress update. Lancet Planet Health. 6:e535–e547. 2022.PubMed/NCBI View Article : Google Scholar | |
Landrigan PJ, Britt M, Fisher S, Holmes A, Kumar M, Mu J, Rizzo I, Sather A, Yousuf A and Kumar P: Assessing the human health benefits of climate mitigation, pollution prevention, and biodiversity preservation. Ann Glob Health. 90(1)2024.PubMed/NCBI View Article : Google Scholar | |
Landrigan PJ, Fuller R, Acosta NJR, Adeyi O, Arnold R, Basu NN, Baldé AB, Bertollini R, Bose-O'Reilly S, Boufford JI, et al: The lancet commission on pollution and health. Lancet. 391:462–512. 2018.PubMed/NCBI View Article : Google Scholar | |
Yang BY, Qu Y, Guo Y, Markevych I, Heinrich J, Bloom MS, Bai Z, Knibbs LC, Li S, Chen G, et al: Maternal exposure to ambient air pollution and congenital heart defects in China. Environ Int. 153(106548)2021.PubMed/NCBI View Article : Google Scholar |