-
ST segment elevation on an electrocardiogram monitor represented a successful myocardial infarction (MI) model surgery. The sinus rhythm and heart rate of mice in the sham group were normal. However, after MI surgery, the ST segment significantly increased, and exposure to UPM exacerbated ST segment elevation (Figure 1B). Echocardiographic measurements of ejection fraction (EF), fractional shortening (FS), and the peak velocities of aortic flow in the heart are shown in Figure 1 and Table 1. Ligation of the LAD coronary artery (MI group) led to significantly lower LVEF and LVFS compared with the sham group (P < 0.01, Figure 1C–E). UPM exposure further reduced LVEF and LVFS (MI vs. MI+UPM, P < 0.01 and P < 0.05, respectively). SLL, SLM, and SLH groups all exhibited significantly increased LVEF (P < 0.001, P < 0.001, and P < 0.01, respectively) and LVFS (P < 0.01) as well as improved cardiac systolic function. The effects of SL on the peak velocities of aortic flow were examined using color Doppler mode at 24 h after MI and UPM exposure. As shown in Table 1, the aortic valve (AV) peak velocity decreased markedly in the MI group compared with the sham group (P < 0.05). When exposed to UPM, the AV peak velocity exhibited a persistent decline. After treatment, each dose of SL increased blood flow in the aorta outflow tract, especially the medium and high doses (P < 0.01 and P < 0.05, respectively). These results suggest that SL improves cardiac dysfunction activity comparable to that of the positive drug aspirin group.
Table 1. Effects of UPM exposure and SL intervention on aortic valve peak flow velocity after myocardial ischemia
Group n AV Peak Vel (mm/s) Sham 6 1189.12 ± 256.85 MI 6 965.36 ± 131.21▲ MI+UPM 6 795.9 ± 120.58 SLL 6 972.6 ± 185.15 SLM 6 1039.69 ± 332.54** SLH 6 1002.02 ± 343.94* AS 6 1017.1 ± 187.43* Note. Compared with Sham group,▲P<0.05 ;Compared with the MI+UPM group,*P<0.05,**P<0.01 -
To further confirm myocardial damage, TTC staining was performed to measure infarct sizes (Fig. 2A, B). We observed an increase in myocardial infarct size in the MI group compared with the sham group (P < 0.001). After UPM exposure, the infarct size increased further. Compared with the MI+UPM group, the infarct size decreased significantly in response to all doses of SL (P < 0.05, P < 0.0001, P < 0.05, respectively). The results of hematoxylin–eosin staining are shown in Fig. 2C. In the sham group, myocardial cells were neatly arranged, myocardial fibers were regularly arranged, and there was no evident inflammatory cell infiltration. In the MI group, the arrangement of myocardial fibers was evidently disordered. Part of the tissue exhibited fibrous edema, and the number of myocardial fibers between the gaps increased, accompanied by inflammatory cell infiltration. Compared with the MI group, the deformation of myocardial fibers, swelling and rupture of myocardial cells, and infiltration of inflammatory cells were more severe in the MI+UPM group. After SL or aspirin treatment, the structural disorder of cardiac muscle fibers improved, and the arrangement of cell nuclei was orderly. Elevated levels of cTnT and LDH in the blood can reflect the degree of myocardial tissue injury after MI. As shown in Fig. 2D–2E, the serum levels of cTnT and LDH were higher in the MI group than in the sham group; additionally, we found that UPM further increased the levels of cTnT and LDH, which decreased after SL and aspirin treatment.
Figure 2. SL extract relieve UPM-induced cardiac injury. (A, B) Effect of SL on infarct size in the experimental mice and the infarct rate as a percentage of the total size. Heart sections of the experimental mice stained with 2, 3, 5-triphenyltetrazolium chloride. Normal tissue was stained red, and infarcted tissue was pale. (C) Histological micrograph of sof hematoxylin & eosin staining myocardial sections, (hematoxylin & eosin:×400) (D, E) LDH and cTnT levels in mice;All values are mean ± SD (n = 6/group). ▲▲▲P < 0.001, ▲▲▲▲P < 0.0001, comparing with the Sham group; ####P < 0.0001 comparing with the MI group; * P < 0.05, ****P < 0.0001 comparing with the MI+UPM group.
-
Inflammatory cytokines in the serum of mice were assayed via ELISA to determine the anti-inflammatory effects of SL. The results are illustrated in Fig. 3; the expression of IL-6 and MCP-1 was markedly increased in response to the MI surgery (P < 0.0001). Additionally, expression of the inflammatory cytokines IL-6, TNF-α, and MCP-1 markedly increased after tracheal instillation of UPM (P < 0.0001, P < 0.0001, P < 0.01, respectively) and then significantly decreased upon treatment with SL (P < 0.0001) (Fig. 3A–3C). Subsequently, we used immunofluorescence to detect the expression of the macrophage cell marker CD68 in myocardial tissue (Fig. 3D). After permanent ligation of the LAD coronary artery, CD68 protein staining intensity in ischemic areas of mice heart was significantly increased, indicating the infiltration of macrophages in the ischemic areas of the heart. Cardiac macrophage infiltration was further exacerbated by tracheal instillation of UPM, and SL administration reduced the infiltration of macrophages.
Figure 3. SL extract alleviated UPM-induced inflammation. (A–C) The changes in inflammatory cytokine interleukin (IL)-6 (IL-6), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1(MCP-1) levels in the serum of mice and the effects of SL; (D) CD68 immunofluorescence staining for macrophages; values are mean ± SD (n = 6/group). ▲▲▲▲P < 0.0001 , comparing with the Sham group; ####P < 0.0001 comparing with the MI group; **P < 0.01, ****P < 0.0001 comparing with the MI+UPM group.
-
Apoptotic death was examined using TUNEL assay and by determining the proportion of caspase-3-positive cells in the myocardial tissue. Compared with Sham, the ratio of apoptotic-positive cells in myocardial cells of the MI group significantly increased (P < 0.01); compared with MI, there was no statistical difference in MI+UPM; compared with MI+UPM, the myocardial cell apoptotic-positive cell rate in SL (P < 0.01). (Fig. 4A, B). Subsequently, the expression of caspase-3 protein in the myocardial tissue was detected via immunohistochemical analysis; the results are shown in Fig. 4C, D. The level of caspase-3 protein in the heart tissue of the MI group was increased (but not significantly) compared with the sham group. Compared with the MI group, UPM exposure significantly increased the expression of caspase-3 in the myocardium tissue. Additionally, we found that UPM triggered myocardial apoptosis, which was markedly attenuated by SL treatment.
Figure 4. SL extract alleviated UPM-induced H9c2 cells apoptosis. (A, B) TUNEL-positive cells and quantification. (C, D) Caspase 3 immunohistochemical staining of apoptosis and quantification; values are mean ± SD (n = 6/group). ▲▲▲P < 0.001 , comparing with the Sham group; ###P < 0.001 comparing with the MI group; *P < 0.05, **P < 0.01 comparing with the MI+UPM group.
-
H9c2 cells were incubated with various concentrations of SL (0, 1.25, 2.5, 5, 10, 20 μg/mL) for 24 h, and cell viability was assessed via the CCK8 assay. The results revealed that cell viability did not decrease among the SL-treated groups compared with the control group. Interestingly, cell viability increased by 8.47% in the 5 μg/mL SL-treated group (Fig. 5A). Changes in morphological characteristics of 5 μg/mL SL-treated H9c2 cells were assessed microscopically. The microscopic images demonstrated that SL was not cytotoxic and was able to promote cell growth (Fig. 5B). Furthermore, the morphological changes of H9c2 cells were observed after UPM exposure combined with hypoxia treatment. The H9c2 cells in the control group were fusiform with a full cytoplasm and clear edges. Conversely, after 12 h of hypoxia, the cells showed shrinkage and inconspicuous edges; however, with UPM exposure, the number of H9c2 cells reduced, and many cells dislodged from the bottom of the cell culture dish. Morphological observation of H9c2 cells revealed that the cells were shrunken and broken, with blurred edges and large deposits of black particles (Fig. 5C). This phenotype could be rescued by SL (5 μg/mL) (Fig. 5C). Therefore, SLE not only exhibited no toxicity toward H9c2 cells but also improved cell viability and prevented cell damage in H9c2 cells exposed to UPM under hypoxia.
Figure 5. SL extract prevented cell damage in H9c2 cells with UPM exposure. (A) H9c2 cells were incubated with various concentrations of SL (0, 1.25, 2.5, 5, 10, 20 μg/mL) for 24 h and cell viability was measured by CCK8 assay. (B) The microscopy images of cell state after 24 h incubation of SL; (C) Images of cell state were taken at the end point of the experiment.
-
To detect the effects of SL on UPM exposure combined with hypoxia-induced H9c2 cell apoptosis, we investigated cell apoptosis and the expression of apoptosis-associated proteins in H9c2 cells. As shown in Fig. 6A–6D, we found that hypoxia treatment of H9c2 cells induced a significant decrease in Bcl-2 expression (P < 0.0001) but had no effect on Bax expression. Exposure to UPM led to the downregulation of Bcl-2 (P < 0.0001) and the upregulation of Bax (P < 0.01), which, in turn, led to an increased ratio of Bax/Bcl-2 compared with the Hyp group (P < 0.0001). SL treatment increased the expression of the antiapoptotic protein Bcl-2 and inhibited that of the proapoptotic marker Bax, resulting in an overall decrease in the Bax/Bcl-2 ratio (all P < 0.0001). Subsequently, apoptosis was examined after Annexin V-FITC/PI fluorescence staining. The results confirmed that UPM exposure promoted cell apoptosis and that SL treatment reversed apoptosis (Fig. 6F). Additionally, PCR was performed to determine the gene expression level of the inflammatory mediator NFκB (Fig. 6E), and the results showed that NFκB expression was slightly increased under UPM exposure but not significantly. After SL treatment, NFκB expression was significantly decreased (P < 0.01).
Figure 6. SL extract prevented cell apoptosis and decreased expression of NFκB in H9c2 cells with UPM exposure. (A-D) The expression of Bax and Bcl-2 in the H9c2 cells; quantitative analysis of Bax, Bcl-2 and Bax/Bcl-2. (E) Real-time PCR analysis shows changes in the relative expression of NFκB genes relative to the housekeeping β-actin gene; (F) Fluorescence microscope were used to examine the results of staining of FITC and PI. values are mean ± SD (n = 3/group). ▲▲▲P < 0.001 , ▲▲▲▲P < 0.0001 comparing with the Control group; ##P < 0.01, ####P < 0.0001 comparing with the Hyp group; **P < 0.01, ****P < 0.0001 comparing with the Hyp+UPM group.
doi: 10.3967/bes2024.137
Shenlian Extract Protects Against Ultrafine Particulate Matter-aggravated Myocardial Ischemic Injury by Inhibiting Inflammation and Cell Apoptosis
-
Abstract:
Objective Emerging evidence suggests that exposure to ultrafine particulate matter (UPM, aerodynamic diameter < 0.1 µm) is associated with adverse cardiovascular events. Previous studies have found that Shenlian (SL) extract possesses anti-inflammatory and antiapoptotic properties and has a promising protective effect at all stages of the atherosclerotic disease process. In this study, we aimed to investigated whether SL improves UPM-aggravated myocardial ischemic injury by inhibiting inflammation and cell apoptosis. Methods We established a mouse model of MI+UPM. Echocardiographic measurement, measurement of myocardialinfarct size, biochemical analysis, ELISA, histopathological analysis, TUNEL, WB , PCR and so on were used to explore the anti-inflammatory and anti-apoptotic effects of SL in vivo and in vitro. Results SL treatment can attenuate UPM-induced cardiac dysfunction by improving left ventricular ejection fraction, fractional shortening, and decreasing cardiac infarction area. SL significantly reduced the levels of myocardial enzymes and attenuated UPM-induced morphological alterations. Moreover, SL significantly reduced expression levels of the inflammatory cytokines IL-6, TNF-α, and MCP-1. UPM further increased the infiltration of macrophages in myocardial tissue, whereas SL intervention reversed this phenomenon. UPM also triggered myocardial apoptosis, which was markedly attenuated by SL treatment. The results of in vitro experiments revealed that SL prevented cell damage caused by exposure to UPM combined with hypoxia by reducing the expression of the inflammatory factor NF-κB and inhibiting apoptosis in H9c2 cells. Conclusion Overall, both in vivo and in vitro experiments demonstrated that SL attenuated UPM-aggravated myocardial ischemic injury by inhibiting inflammation and cell apoptosis. The mechanisms were related to the downregulation of macrophages infiltrating heart tissues. -
Key words:
- Ultrafine particulate matter /
- Shenlian extract /
- Inflammation /
- Apoptosis /
- Macrophage
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
&These authors contributed equally to this work.
注释:1) CREDIT AUTHORSHIP CONTRIBUTION STATEMENT: 2) DECLARATION OF COMPETING INTEREST: -
Figure 1. SL extract can relieve UPM-induced cardiac dysfunction. (A) Flowchart of experiments. (B) Effects of UPM on the ST segment. (C) M-mode ultrasound of the heart to evaluate cardiac function. (D,E) An ultrahigh-resolution small animal ultrasound imaging system was used to obtain left ventricular ejection fraction (LVEF), LV fractional shortening (LVFS); all values are mean ± SD (n = 6/group). ▲▲P < 0.01, comparing with the Sham group; # P < 0.05, ## P < 0.01 comparing with the MI group; ** P < 0.01, **** P < 0.0001 comparing with the MI+UPM group.
Figure 2. SL extract relieve UPM-induced cardiac injury. (A, B) Effect of SL on infarct size in the experimental mice and the infarct rate as a percentage of the total size. Heart sections of the experimental mice stained with 2, 3, 5-triphenyltetrazolium chloride. Normal tissue was stained red, and infarcted tissue was pale. (C) Histological micrograph of sof hematoxylin & eosin staining myocardial sections, (hematoxylin & eosin:×400) (D, E) LDH and cTnT levels in mice;All values are mean ± SD (n = 6/group). ▲▲▲P < 0.001, ▲▲▲▲P < 0.0001, comparing with the Sham group; ####P < 0.0001 comparing with the MI group; * P < 0.05, ****P < 0.0001 comparing with the MI+UPM group.
Figure 3. SL extract alleviated UPM-induced inflammation. (A–C) The changes in inflammatory cytokine interleukin (IL)-6 (IL-6), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1(MCP-1) levels in the serum of mice and the effects of SL; (D) CD68 immunofluorescence staining for macrophages; values are mean ± SD (n = 6/group). ▲▲▲▲P < 0.0001 , comparing with the Sham group; ####P < 0.0001 comparing with the MI group; **P < 0.01, ****P < 0.0001 comparing with the MI+UPM group.
Figure 4. SL extract alleviated UPM-induced H9c2 cells apoptosis. (A, B) TUNEL-positive cells and quantification. (C, D) Caspase 3 immunohistochemical staining of apoptosis and quantification; values are mean ± SD (n = 6/group). ▲▲▲P < 0.001 , comparing with the Sham group; ###P < 0.001 comparing with the MI group; *P < 0.05, **P < 0.01 comparing with the MI+UPM group.
Figure 5. SL extract prevented cell damage in H9c2 cells with UPM exposure. (A) H9c2 cells were incubated with various concentrations of SL (0, 1.25, 2.5, 5, 10, 20 μg/mL) for 24 h and cell viability was measured by CCK8 assay. (B) The microscopy images of cell state after 24 h incubation of SL; (C) Images of cell state were taken at the end point of the experiment.
Figure 6. SL extract prevented cell apoptosis and decreased expression of NFκB in H9c2 cells with UPM exposure. (A-D) The expression of Bax and Bcl-2 in the H9c2 cells; quantitative analysis of Bax, Bcl-2 and Bax/Bcl-2. (E) Real-time PCR analysis shows changes in the relative expression of NFκB genes relative to the housekeeping β-actin gene; (F) Fluorescence microscope were used to examine the results of staining of FITC and PI. values are mean ± SD (n = 3/group). ▲▲▲P < 0.001 , ▲▲▲▲P < 0.0001 comparing with the Control group; ##P < 0.01, ####P < 0.0001 comparing with the Hyp group; **P < 0.01, ****P < 0.0001 comparing with the Hyp+UPM group.
Table 1. Effects of UPM exposure and SL intervention on aortic valve peak flow velocity after myocardial ischemia
Group n AV Peak Vel (mm/s) Sham 6 1189.12 ± 256.85 MI 6 965.36 ± 131.21▲ MI+UPM 6 795.9 ± 120.58 SLL 6 972.6 ± 185.15 SLM 6 1039.69 ± 332.54** SLH 6 1002.02 ± 343.94* AS 6 1017.1 ± 187.43* Note. Compared with Sham group,▲P<0.05 ;Compared with the MI+UPM group,*P<0.05,**P<0.01 -
[1] Joshi SS, Miller MR, Newby DE. Air pollution and cardiovascular disease: the Paul Wood Lecture, British Cardiovascular Society 2021. Heart, 2022; 108, 1267−73. doi: 10.1136/heartjnl-2021-319844 [2] Lelieveld J, Pozzer A, Pöschl U, et al. Loss of life expectancy from air pollution compared to other risk factors: a worldwide perspective. Cardiovasc Res, 2020; 116, 1910−7. doi: 10.1093/cvr/cvaa025 [3] Zhang JW, Chen Z, Shan D, et al. Adverse effects of exposure to fine particles and ultrafine particles in the environment on different organs of organisms. J Environ Sci (China), 2024; 135, 449−73. doi: 10.1016/j.jes.2022.08.013 [4] Miller MR, Newby DE. Air pollution and cardiovascular disease: car sick. Cardiovasc Res, 2020; 116, 279−94. [5] Jaafari J, Naddafi K, Yunesian M, et al. Associations between short term exposure to ambient particulate matter from dust storm and anthropogenic sources and inflammatory biomarkers in healthy young adults. Sci Total Environ, 2021; 761, 144503. doi: 10.1016/j.scitotenv.2020.144503 [6] Lu X, Li RQ, Yan XX. Airway hyperresponsiveness development and the toxicity of PM2.5. Environ Sci Pollut Res Int, 2021; 28, 6374−91. doi: 10.1007/s11356-020-12051-w [7] Kleinman MT, Araujo JA, Nel A, et al. Inhaled ultrafine particulate matter affects CNS inflammatory processes and may act via MAP kinase signaling pathways. Toxicol Lett, 2008; 178, 127−30. doi: 10.1016/j.toxlet.2008.03.001 [8] Bhargava A, Shukla A, Bunkar N, et al. Exposure to ultrafine particulate matter induces NF-κβ mediated epigenetic modifications. Environ Pollut, 2019; 252, 39−50. doi: 10.1016/j.envpol.2019.05.065 [9] Li J, An Z, Song J, et al. Fine particulate matter-induced lung inflammation is mediated by pyroptosis in mice. Ecotoxicol Environ Saf, 2021; 219, 112351. doi: 10.1016/j.ecoenv.2021.112351 [10] Li RJ, Zhao LF, Tong JL, et al. Fine particulate matter and sulfur dioxide coexposures induce rat lung pathological injury and inflammatory responses via TLR4/p38/NF-κB pathway. Int J Toxicol, 2017; 36, 165−73. doi: 10.1177/1091581816682225 [11] Dou CM, Zhang J, Qi CC. Cooking oil fume-derived PM2.5 induces apoptosis in A549 cells and MAPK/NF-кB/STAT1 pathway activation. Environ Sci Pollut Res Int, 2018; 25, 9940−8. doi: 10.1007/s11356-018-1262-5 [12] Badamjav R, Zhang L, Sonom D, et al. Thalictrum minus L. ameliorates particulate matter-induced acute lung injury in mice. J Ethnopharmacol, 2021; 264, 113379. doi: 10.1016/j.jep.2020.113379 [13] Wei HY, Yuan WJ, Yu H, et al. Cytotoxicity induced by fine particulate matter (PM2.5) via mitochondria-mediated apoptosis pathway in rat alveolar macrophages. Environ Sci Pollut Res Int, 2021; 28, 25819−29. doi: 10.1007/s11356-021-12431-w [14] Geng XW, Wang XH, Luo M, et al. Induction of neutrophil apoptosis by a Bcl-2 inhibitor reduces particulate matter-induced lung inflammation. Aging (Albany NY), 2018; 10, 1415−23. [15] Ren F, Ji C, Huang YJ, et al. AHR-mediated ROS production contributes to the cardiac developmental toxicity of PM2.5 in zebrafish embryos. Sci Total Environ, 2020; 719, 135097. doi: 10.1016/j.scitotenv.2019.135097 [16] Li XL, Geng J, Chen YH, et al. Exposure to particulate matter induces cardiomyocytes apoptosis after myocardial infarction through NFκB activation. Biochem Biophys Res Commun, 2017; 488, 224−31. doi: 10.1016/j.bbrc.2017.05.047 [17] Guo Y, Yang Q, Weng XG, et al. Shenlian extract against myocardial injury induced by ischemia through the regulation of NF-κB/IκB signaling axis. Front Pharmacol, 2020; 11, 134. doi: 10.3389/fphar.2020.00134 [18] Li YJ, Chen Y, You Y, et al. Effects of Shenlian extracts (参莲提取物) on atherosclerosis by inhibition of the inflammatory response. J Tradit Chin Med, 2011; 31, 344-8. [19] Liu L, Li Q, Yin J, et al. ShenLian extract enhances TGF-β functions in the macrophage-SMC unit and stabilizes atherosclerotic plaques. Front Pharmacol, 2021; 12, 669730. doi: 10.3389/fphar.2021.669730 [20] Qu SQ, Li K, Yang T, et al. Shenlian extract protects against ultrafine particulate matter-aggravated myocardial ischemic injury by inhibiting inflammation response via the activation of NLRP3 inflammasomes. Environ Toxicol, 2021; 36, 1349−61. doi: 10.1002/tox.23131 [21] Qu SQ, Deng SQ, Yang T, et al. Shengmai Yin alleviated plaque vulnerability and ischemic myocardial damage in diesel exhaust particle-aggravated atherosclerosis with myocardial ischemia. Ecotoxicol Environ Saf, 2022; 234, 113379. doi: 10.1016/j.ecoenv.2022.113379 [22] Hou Y, Fan FH, Xie N, et al. Rhodiola crenulata alleviates hypobaric hypoxia-induced brain injury by maintaining BBB integrity and balancing energy metabolism dysfunction. Phytomedicine, 2024; 128, 155529. doi: 10.1016/j.phymed.2024.155529 [23] Zeng YY, Cao YF, Qiao X, et al. Air pollution reduction in China: recent success but great challenge for the future. Sci Total Environ, 2019; 663, 329−37. doi: 10.1016/j.scitotenv.2019.01.262 [24] Mannucci PM, Harari S, Franchini M. Novel evidence for a greater burden of ambient air pollution on cardiovascular disease. Haematologica, 2019; 104, 2349−57. doi: 10.3324/haematol.2019.225086 [25] Peters A, Dockery DW, Muller JE, et a yocardial infarction. Circulation, 2001; 103, 2810-5. [26] Chen C, Zhu PF, Lan L, et al. Short-term exposures to PM2.5 and cause-specific l. Increased particulate air pollution and the triggering of m mortality of cardiovascular health in China. Environ Res, 2018; 161, 188−94. doi: 10.1016/j.envres.2017.10.046 [27] Shi LH, Zanobetti A, Kloog I, et al. Low-concentration PM2.5 and mortality: estimating acute and chronic effects in a population-based study. Environ Health Perspect, 2016; 124, 46−52. doi: 10.1289/ehp.1409111 [28] Beber LCC, da Silva MOAF, dos Santos AB, et al. The association of subchronic exposure to low concentration of PM2.5 and high-fat diet potentiates glucose intolerance development, by impairing adipose tissue antioxidant defense and eHSP72 levels. Environ Sci Pollut Res Int, 2020; 27, 32006−16. doi: 10.1007/s11356-020-09581-8 [29] Hoffmann B, Boogaard H, de Nazelle A, et al. WHO air quality guidelines 2021-aiming for healthier air for all: a joint statement by medical, public health, scientific societies and patient representative organisations. Int J Public Health, 2021; 66, 1604465. doi: 10.3389/ijph.2021.1604465 [30] Chang ET, Lau EC, Moolgavkar SH. Smoking, air pollution, and lung cancer risk in the Nurses' Health Study cohort: time-dependent confounding and effect modification. Crit Rev Toxicol, 2020; 50, 189−200. doi: 10.1080/10408444.2020.1727410 [31] Miller KA, Siscovick DS, Sheppard L, et al. Long-term exposure to air pollution and incidence of cardiovascular events in women. N Engl J Med, 2007; 356, 447−58. doi: 10.1056/NEJMoa054409 [32] Hung CS, Huang CC, Pan SC, et al. Acute particulate matter exposure is associated with disturbances in heart rate complexity in patients with prior myocardial infarction. Sci Total Environ, 2020; 733, 138842. doi: 10.1016/j.scitotenv.2020.138842 [33] Chen H, Burnett RT, Copes R, et al. Ambient fine particulate matter and mortality among survivors of myocardial infarction: population-based cohort study. Environ Health Perspect, 2016; 124, 1421−8. doi: 10.1289/EHP185 [34] Tibuakuu M, Michos ED, Navas-Acien A, et al. Air pollution and cardiovascular disease: a focus on vulnerable populations worldwide. Curr Epidemiol Rep, 2018; 5, 370−8. doi: 10.1007/s40471-018-0166-8 [35] Pope III CA, Turner MC, Burnett RT, et al. Relationships between fine particulate air pollution, cardiometabolic disorders, and cardiovascular mortality. Circ Res, 2015; 116, 108−15. doi: 10.1161/CIRCRESAHA.116.305060 [36] Aryal A, Harmon AC, Dugas TR. Particulate matter air pollutants and cardiovascular disease: strategies for intervention. Pharmacol Ther, 2021; 223, 107890. doi: 10.1016/j.pharmthera.2021.107890 [37] Chen LN, Guo Y, Qu SQ, et al. The protective effects of Shengmai formula against myocardial injury induced by ultrafine particulate matter exposure and myocardial ischemia are mediated by the PI3K/AKT/p38 MAPK/Nrf2 pathway. Front Pharmacol, 2021; 12, 619311. doi: 10.3389/fphar.2021.619311 [38] Wang Y, Tang M. PM2.5 induces autophagy and apoptosis through endoplasmic reticulum stress in human endothelial cells. Sci Total Environ, 2020; 710, 136397. doi: 10.1016/j.scitotenv.2019.136397 [39] Pei YH, Chen J, Wu X, et al. LncRNA PEAMIR inhibits apoptosis and inflammatory response in PM2.5 exposure aggravated myocardial ischemia/reperfusion injury as a competing endogenous RNA of miR-29b-3p. Nanotoxicology, 2020; 14, 638−53. doi: 10.1080/17435390.2020.1731857 [40] Jiang Y, Zhao XH, Chen J, et al. PM2.5 induces cardiac malformations via PI3K/akt2/mTORC1 signaling pathway in zebrafish larvae. Environ Pollut, 2023; 323, 121306. doi: 10.1016/j.envpol.2023.121306 [41] Wang YF, Fei YX, Zhao B, et al. Ma Xing Shi Gan decoction protects against PM2.5-induced lung injury through suppression of epithelial-to-mesenchymal transition (EMT) and epithelial barrier disruption. Evid Based Complement Alternat Med, 2020; 2020, 7176589. doi: 10.1155/2020/7176589 [42] Wang PL, Liu H, Fan XS, et al. Effect of San'ao decoction on aggravated asthma mice model induced by PM2.5 and TRPA1/TRPV1 expressions. J Ethnopharmacol, 2019; 236, 82−90. doi: 10.1016/j.jep.2019.02.043 [43] Liu H, Fan XS, Zhu Y. Effects of Wu'ao decoction on the expressions of TRPA1 and TRPV1 in lung in PM2.5 and OVA induced severe asthma mice model. Chin J Integr Tradit West Med, 2019; 39, 997−1003. (In Chinese [44] Shen ZY, Fu SG, Yang AD, et al. Effects of modified Qianjin Weijing decoction on TNF-α and NF-κB in rats with lung injury induced by particulate matter. Chin J Inf Tradit Chin Med, 2018; 25, 38−42. (In Chinese [45] Zhang JB, Sun L, Li SQ, et al. Therapeutic effect of Qingzao Runfei Huazhuo Xingxue decoction on PM2.5-induced respiratory disease in mice. Chin Crit Care Med, 2016; 28, 916−20. (In Chinese [46] Kim HY, Yoon JJ, Kim DS, et al. YG-1 extract improves acute pulmonary inflammation by inducing bronchodilation and inhibiting inflammatory cytokines. Nutrients, 2021; 13, 3414. doi: 10.3390/nu13103414 [47] Xia YL, Dolgor S, Jiang SY, et al. YiQiFuMai lyophilized injection attenuates particulate matter-induced acute lung injury in mice via TLR4-mTOR-autophagy pathway. Biomed Pharmacother, 2018; 108, 906−13. doi: 10.1016/j.biopha.2018.09.088 [48] Chen LN, Guo Y, Qu SQ, et al. The protective effects of Shengmai formula against myocardial injury induced by ultrafine particulate matter exposure and myocardial ischemia are mediated by the PI3K/AKT/p38 MAPK/Nrf2 pathway. Front Pharmacol, 2021; 12, 619311. [49] Li JJ, Wang CM, Wang YJ, et al. Network pharmacology analysis and experimental validation to explore the mechanism of Shenlian extract on myocardial ischemia. J Ethnopharmacol, 2022; 288, 114973. doi: 10.1016/j.jep.2022.114973 [50] Wang CM. Protective effects of Shenlian extract on myocardial infarction of rats by regulating mitochondrial autophagy and intervening inflammasomes activation. China Academy of Chinese Medical Sciences. 2021. (In Chinese [51] Arefnezhad R, Nejabat A, Behjati F, et al. Tanshinone IIA against cerebral ischemic stroke and ischemia-reperfusion injury: a review of the current documents. Mini Rev Med Chem, 2024; 24, 1701−9. doi: 10.2174/0113895575299721240227070032 [52] Gupta S, Mishra KP, Kumar B, et al. Andrographolide attenuates complete Freund's adjuvant induced arthritis via suppression of inflammatory mediators and pro-inflammatory cytokines. J Ethnopharmacol, 2020; 261, 113022. doi: 10.1016/j.jep.2020.113022 [53] Chen YW, Huang MZ, Chen CL, et al. PM2.5 impairs macrophage functions to exacerbate pneumococcus-induced pulmonary pathogenesis. Part Fibre Toxicol, 2020; 17, 37. doi: 10.1186/s12989-020-00362-2 [54] Chen S, Chen LP, Ye LZ, et al. PP2A-mTOR-p70S6K/4E-BP1 axis regulates M1 polarization of pulmonary macrophages and promotes ambient particulate matter induced mouse lung injury. J Hazard Mater, 2022; 424, 127624. doi: 10.1016/j.jhazmat.2021.127624 [55] Cui XJ, Lai WJ, Zhao Y, et al. The exosome-mediated cascade reactions for the transfer and inflammatory responses of fine atmospheric particulate matter in macrophages. Environ Sci Technol, 2023; 57, 7891−901. doi: 10.1021/acs.est.3c01436 [56] Hu XQ, Chen M, Cao X, et al. TGF-β-containing small extracellular vesicles from PM2.5-activated macrophages induces cardiotoxicity. Front Cardiovasc Med, 2022; 9, 917719. doi: 10.3389/fcvm.2022.917719 [57] Vendrov AE, Xiao H, Lozhkin A, et al. Cardiomyocyte NOX4 regulates resident macrophage-mediated inflammation and diastolic dysfunction in stress cardiomyopathy. Redox Biol, 2023; 67, 102937. doi: 10.1016/j.redox.2023.102937