Open Access

N‑acetyl cysteine prevents ambient fine particulate matter‑potentiated atherosclerosis via inhibition of reactive oxygen species‑induced oxidized low density lipoprotein elevation and decreased circulating endothelial progenitor cell

  • Authors:
    • Yixin Xu
    • Haoran Bu
    • Yufan Jiang
    • Xiaoqing Zhuo
    • Ke Hu
    • Zhihua Si
    • Yong Chen
    • Qiwei Liu
    • Xianwei Gong
    • Haihui Sun
    • Qingyi Zhu
    • Lianqun Cui
    • Xiaochun Ma
    • Yuqi Cui
  • View Affiliations

  • Published online on: May 27, 2022     https://doi.org/10.3892/mmr.2022.12752
  • Article Number: 236
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ambient fine particulate matter (PM) serves an important role in the development of cardiovascular disease, including atherosclerosis. Antioxidant N‑acetyl cysteine (NAC) has protective effects in the cardiovascular system. However, it is unknown if NAC prevents PM‑potentiated atherosclerosis in hyperlipidemia. Low‑density lipoprotein (LDL) receptor knockout mice were pretreated with 1 mg/ml NAC in drinking water for 1 week and continued to receive NAC, high‑fat diet and intranasal instillation of PM for 1 week or 6 months. Blood plasma was collected for lipid profile, oxidized (ox‑)LDL, blood reactive oxygen species (ROS) and inflammatory cytokine (TNF‑α, IL‑1β and IL‑6) measurement. Blood cells were harvested for endothelial progenitor cell (EPC) population and intracellular ROS analysis. Murine aorta was isolated for atherosclerotic plaque ratio calculation. NAC treatment maintained circulating EPC level and significantly decreased blood ox‑LDL and ROS, inflammatory cytokines, mononuclear and EPC intracellular ROS levels as well as aortic plaque ratio. NAC prevented PM‑potentiated atherosclerosis by inhibiting plasma ROS‑induced ox‑LDL elevation, mononuclear cell and EPC intracellular ROS‑induced circulating EPC reduction and inflammatory cytokine production.

Introduction

Ambient fine particulate matter (PM) is associated with cardiovascular disease (CVD), including coronary artery disease (CAD), heart failure and hypertension (1). However, the underlying mechanism is not well studied. Different types of CVD may be caused by duration of PM exposure; acute PM exposure increases myocardial infarction (2), stroke (3) and other acute cardiovascular events (4), while chronic exposure contributes to development of hypertension, diabetes and other cardiometabolic conditions (4). Following six months exposure of PM with diameter ≤2.5 µm, atherosclerosis development in apolipoprotein E knockout mice significantly increases and is accompanied by vasomotor tone alteration and vascular inflammation (5). However, the detailed mechanisms underlying PM-potentiated atherosclerosis in hyperlipidemia is not well studied.

Endothelial dysfunction or injury is a key factor that contributes to the development of atherosclerosis and CAD (6,7). Bone marrow (BM)-derived endothelial progenitor cells (EPCs) serve a key role in vascular reendothelialization, angiogenesis and promotion of neointima formation following vascular injury (811). In addition, EPC number and function are significantly decreased in patients with CAD and hyperlipidemia (12,13). However, the exact mechanism underlying decreased EPC levels in patients with hyperlipidemia remains unknown. Oxidized low-density lipoprotein (ox-LDL) is a key component in hyperlipidemia and serves an important role in development of atherosclerosis, primarily via oxidative stress (14). Our previous study indicated that ox-LDL has a similar effect to chronic hyperlipidemia on BM and blood EPC populations (15). However, the effect of PM exposure on ox-LDL and EPC population in hyperlipidemia is still unknown.

Reactive oxygen species (ROS) are small molecules, such as hydrogen peroxide (H2O2), that regulate tissue oxidative stress (16). ROS-induced oxidative stress also regulates BM stem cell (BMSC) and BMSC-derived progenitor cell self-renewal, proliferation, mobilization, homing, migration, differentiation, apoptosis and senescence (17,18). Rate of ROS generation in peripheral blood monocytes is increased in hyperlipidemic patients, along with elevated plasma ox-LDL levels, which increases intracellular ROS formation in cultured endothelial cells (19,20). Our previous data showed that PM exposure also increases EPC intracellular ROS production in wild-type (WT) mice (21). To the best of our knowledge, however, ROS production following short- and long-term PM exposure in mice with hyperlipidemia has not been evaluated.

N-acetyl cysteine (NAC) is an antioxidant widely used to investigate CVD (22). Our previous data indicated that NAC prevents atherosclerosis by maintaining EPC population, decreasing both intracellular and extracellular ROS production in hyperlipidemic mice and preventing LDL oxidation in WT mice (15,23). NAC also protects EPCs from apoptosis and decreases levels of inflammatory cytokines following PM exposure in WT mice (21). To the best of our knowledge, however, the effects of NAC on development of atherosclerosis in hyperlipidemic mice with acute and chronic PM exposure has not been investigated. The present study amid to determine the protective effect and underlying mechanism of NAC on atherosclerosis in mice with hyperlipidemia following acute and chronic PM exposure.

Materials and methods

Animal model

All animal experiments were performed in accordance with the Guidelines of the Animal Care Committee of Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China. The experimental protocols for the present study were reviewed and approved by the Animal Care Committee of Shandong First Medical University (approval no. 2021-228). A total of 48 4–6-week-old male homozygous C57BL/6J LDL receptor knockout (LDLR KO) mice were obtained from Shanghai Model Organisms Center, Inc. and randomized into 6 groups (n=8/group) as follows: Normal diet (ND); high-fat diet (HFD); HFD + NAC; PM + ND; PM + HFD and PM + HFD + NAC. In addition to ad libitum access to food and water, all mice were kept at room temperature with 40–60% humidity and a 12/12-h light/dark cycle. Following 1 week acclimation, mice were challenged with HFD (17% anhydrous milk fat; 0.2% cholesterol) to induce hyperlipidemia for 1 week or 6 months, as previously described (23). PM (cat. no. NIST2786; mean particle diameter, <4 µm) was purchased from MilliporeSigma. PM was dispersed in solution by ultrasonication in endotoxin-free water for 30 min at a concentration of 0.5 µg/µl as previously described (24,25). Hyperlipidemic mice were treated with 10 µg PM three times/week for 1 week or 6 months via intranasal instillation, as previously described (26). Endotoxin-free water was used as control. There are multiple routes for PM to invade the body following inhalation. The primary route is via the respiratory system; other routes include olfactory epithelium and digestive (swallowing sputum with PM) (27). Intranasal instillation was used to mimic natural PM exposure. To evaluate the protective effect of NAC on hyperlipidemic mice with acute and chronic PM exposure, animals were pre-treated with NAC (1 mg/ml in drinking water) for 1 week and continued NAC treatment for 1 week or 6 months, as previously described (23). Mice fed ND were used as control. Although animal health and behavior were monitored every 7 days, 6 mice in the 6-month PM group died before the end of experiment; this may have been due to advanced atherosclerosis development.

Following 1-week or 6-month PM, HFD and NAC treatment, 3.0 and 1.5% isoflurane was used to induce and maintain anesthesia, respectively. A total of 300–500 µl murine blood was collected via cardiac puncture. Then, carbon dioxide (50–70% of chamber volume/min) was used to euthanize the animals. Death was confirmed by cardiac and respiratory arrest or fixed and dilated pupils. Murine aorta was isolated after confirming the death of mice.

Blood lipid, plasma ROS and inflammatory cytokine measurement and atherosclerotic plaque ratio calculation

At the end of the experiment, murine blood was centrifuged at 300 g and room temperature for 20 min to collect plasma for lipid profile, ox-LDL, plasma ROS and inflammatory cytokine analysis. A total of 40 µl plasma was applied in the lipid profile test cassette (Cholestech LDX; cat. no. 10–989; Thermo Fisher Scientific, Inc.) for measuring total cholesterol (TC), triglyceride (TG), LDL, high-density lipoprotein (HDL), non-HDL cholesterol (28) and the ratio of TC/HDL by using Alere Cholestech LDX System. Murine plasma ox-LDL was measured with Ox-LDL ELISA kit (cat. no. CSB-E07933m; Cusabio Technology LLC) according to the manufacturer's instruction. Murine plasma was collected to detect blood ROS levels by using In Vitro ROS/Reactive Nitrogen Species (RNS) Assay (cat. no. STA-347; Cell Biolabs, Inc.) and Pierce™ Quantitative Peroxide Assay kit (lipid-compatible formulation; cat. no. 23285; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. Following incubation of plasma with reagent at room temperature for 30 min, samples were transferred to a 48-well plate for analysis using a microplate reader (Multiskan™ FC Microplate Photometer) at the wavelength of 595 nm to measure the optical density (OD). Plasma levels of inflammatory cytokines TNF-α (cat. no. 430915), IL-1β (cat. no. 432615) and IL-6 (cat. no. 431315) were measured using ELISA kits (BioLegend, Inc.) according to the manufacturer's instructions. Murine aorta was dissected and directly stained with oil red (MilliporeSigma) for plaque formation measurement at room temperature for 5 min. The ratio of plaque area to total inner surface of aorta was calculated as previously described (29).

Mononuclear cell and EPC intracellular ROS detection and EPC measurement

An endothelial cell marker combined with a stem cell marker CD34+/CD133+ was used to identify EPC as previously described (30). Murine blood cells were harvested after sacrifice to detect EPC and mononuclear cell intracellular ROS formation via flow cytometry by using ROS Detection Reagents-FITC (cat. no. D399; Invitrogen; Thermo Fisher Scientific, Inc.) as previously described (31). After removing red blood cells (RBCs) using 1X RBC lysis buffer (cat. no. 00433357; Thermo Fisher Scientific), a total of 50,000 cells in each sample were incubated with 5 µg/ml ROS Detection Reagents-FITC for 10 min at 37°C. BD™ LSRII (BD Biosciences) at a wavelength of 525 nm was used to calculate the positively fluorescent cells for intracellular ROS detection. For EPC measurement, cells were incubated with anti-mouse CD34-AF700 (cat. no. 560518; BD Biosciences) and CD133-PE (cat. no. 12-1331-82; eBioscience; Thermo Fisher Scientific, Inc.) as previously described (21), then incubated with 5 µg/ml ROS Detection Reagents-FITC for 10 min at 37°C. Labeled cells were washed twice with PBS and suspended in warm PBS for analysis by flow cytometry on a BD™ LSRII system (BD Biosciences). All antibodies were diluted 1:100.

Statistical analysis

All data are presented as the mean ± SD (5–8 repeats) and were analyzed using one- or two-way ANOVA (PRISM Version 5.0.; GraphPad Software, Inc.) followed by post hoc conservative Bonferroni's test to minimize type I error. Normal distribution of data was tested using Shapiro-Wilk W-test; equal variance was tested using F-test. When the null hypothesis of normality and/or equal variance was rejected, non-parametric Mann-Whitney U-test was used. Two-tailed P<0.05 was considered to indicate a statistically significant difference.

Results

NAC prevents PM-potentiated atherosclerosis in hyperlipidemia

To test the effect of NAC on atherosclerosis development in hyperlipidemic mice following PM exposure, LDLR KO mice were pre-treated with NAC for 1 week, and continued with NAC, HFD and PM exposure for 6 months. Atherosclerotic plaque formation was increased up to 1.3 fold in PM + HFD compared with HFD mice (Fig. 1). NAC significantly decreased plaque formation in PM + HFD + NAC (0.43±0.20%) and HFD + NAC (0.42±0.22%) compared with PM + HFD (0.80±0.29%) and HFD (0.62±0.21%) (Fig. 1). There was no plaque formation in any 1 week treatment (data not shown). These results indicated that NAC prevented PM-potentiated atherosclerosis formation.

NAC decreases blood ox-LDL levels following chronic PM exposure in hyperlipidemic mice

The blood lipid levels, including TC, TG and LDL, were significantly increased in LDLR KO mice treated with HFD for 1 week or 6 months with or without PM exposure compared with ND mice (Table I). NAC had no effect on blood lipid profile in hyperlipidemic mice with or without PM exposure (Table I). Blood ox-LDL levels raised to 2.0±0.5 nmol/ml in LDLR KO 6-month PM + ND mice compared with 1-week PM + ND mice (0.44±0.10 nmol/ml; Fig. 2). Furthermore, blood ox-LDL level significantly increased to 14.00±2.00 nmol/ml in 6-month PM + HFD mice compared with 6-month HFD mice (10.02±2.21 nmol/ml; Fig. 2). NAC treatment significantly decreased murine plasma ox-LDL levels in LDLR KO mice following 6 month HFD treatment with (PM + HFD + NAC, 6.03±0.21 nmol/ml) or without PM (HFD + NAC, 5.80±3.00 nmol/ml) compared with 6-month PM + HFD (14.01±2.00 nmol/ml) or HFD groups (10.02±2.21 nmol/ml; Fig. 2). Blood ox-LDL levels were not elevated in any 1 week group compared with ND (Fig. 2). These data suggested that NAC prevented atherosclerosis in association with reducing PM-induced elevation of ox-LDL level in hyperlipidemia.

Table I.

Lipid profile of LDL receptor knockout mice.

Table I.

Lipid profile of LDL receptor knockout mice.

A, 1 week

LipidNDHFDHFD + NACPM + NDPM + HFDPM + HFD + NAC
TC, mg/dl228.60±25.66 1162.80±148.13a 1132.80±123.22a230.60±27.88 1252.80±233.23b 1255.60±114.34b
HDL, mg/dl88.00±9.6482.80±6.8785.40±7.5490.00±10.2485.50±7.0782.50±6.34
TG, mg/dl148.40±35.47 673.20±123.82a 632.80±87.32a150.80±36.62 658.30±130.94b 642.50±90.54b
LDL, mg/dl140.00±0.74 920.60±119.92a 983.80±108.50a139.00±1.55 898.40±152.72b 884.20±110.30b
Non-HDL, mg/dl146.40±24.25 1074.80±136.13a 1054.80±103.22a148.30±26.72 1088.80±140.24b 1059.20±112.32b
TC/HDL2.61±0.32 14.00±0.74a 13.20±0.85a2.56±0.52 14.65±0.24b 15.22±0.62b

B, 6 months

LipidNDHFDHFD + NACPM + NDPM + HFDPM + HFD + NAC

TC, mg/dl228.70±29.02 1704.0±228.70a 1728.00±234.80a232.50±28.77 1789.00±242.30b 1732.00±243.80b
HDL, mg/dl76.00±9.8074.00±24.4092.00±21.3078.00±10.1078.00±25.2095.00±32.40
TG, mg/dl103.70±35.00 597.50±217.30a 545.50±197.20a105.50±42.00 566.40±220.40b 567.40±188.40b
LDL, mg/dl132.00±23.10 1583.00±100.50a 1576.00±112.20a140.00±34.20 1602.00±103.20b 1610.00±122.20b
Non-HDL, mg/dl119.30±83.30 1700.00±90.50a 1720.00±100.60a122.50±90.50 1689.00±89.50b 1656.00±112.40b
TC/HDL2.80±0.40 23.00±5.40a 18.78±7.20a2.98±0.70 22.90±4.40b 18.23±6.90b

{ label (or @symbol) needed for fn[@id='tfn1-mmr-26-01-12752'] } Data are presented as the mean ± standard deviation (n=5-8).

a P<0.01 vs. ND;

b P<0.01 vs. PM + ND. HDL, high-density lipoprotein; LDL, low-density lipoprotein; TG, triglyceride; TC, total cholesterol; ND, normal diet; HFD, high-fat diet; NAC, N-acetyl cysteine; PM, ambient fine particulate matter.

NAC maintains EPC levels following acute and chronic PM exposure in mice with hyperlipidemia

PM significantly decreased levels of circulating CD34+/CD133+ cell in LDLR KO mice fed ND for 1 week (0.044±0.003%) and 6 months (0.010±0.003%) and further decreased the cell population in PM + HFD mice (1 week, 0.02±0.01%; 6 months, 0.006±0.003%; Fig. 3). NAC treatment effectively reversed the effects of PM and hyperlipidemia on EPC (PM + HFD + NAC) in both 1 week (0.05±0.01%) and 6 month (0.03±0.01%) groups compared with 1-week PM + HFD (0.02±0.01%) and 6-month PM + HFD (0.0056±0.0029%) groups respectively. Of note, EPC levels were not altered in 1-week HFD mice (HFD, 0.050±0.002% vs. ND, 0.050±0.005%) and were significantly decreased in 6-month HFD mice (HFD, 0.02±0.01% vs. ND, 0.040±0.002%). These data demonstrated that NAC prevented PM-induced decrease of EPC in hyperlipidemia.

NAC blocks plasma, mononuclear cell and EPC intracellular ROS production in hyperlipidemic mice with acute and chronic PM exposure

Our previous study indicated that increased blood ROS production is associated with increased ox-LDL levels (23) and circulating EPCs are derived from mononuclear cells (32). To determine the mechanism underlying the protective effect of NAC on elevated plasma ox-LDL and decreased circulating EPC levels in mice with hyperlipidemia following acute and chronic PM exposure, plasma ROS, as well as circulating mononuclear cell and EPC intracellular ROS production, was measured in hyperlipidemic mice following 1-week and 6-month PM exposure with or without NAC. Total plasma ROS/RNS and H2O2 levels were significantly increased in 1-week PM + HFD mice (530±48 µM/ml; 0.33±0.02 OD) or 6 months (1,255±110 µM/ml; 0.60±0.01 OD) compared with 1 week HFD mice (239±57 µM/ml; 0.12±0.05 OD) or 6 month (845±107 µM/ml; 0.45±0.04 OD). NAC significantly blocked plasma ROS production in 1-week (PM + HFD + NAC, 318±35 µM/ml; 0.13±0.01 OD) or 6-month (PM + HFD + NAC, 480±88 µM/ml; 0.24±0.02 OD) PM and HFD treated mice compared with 1 week or 6-month PM + HFD mice (Fig. 4A and B). Similarly, mononuclear cell and EPC intracellular ROS production were significantly elevated up to 1.3-1.6-fold in LDLR KO 1-week or 6-month PM + HFD mice compared with HFD mice (Fig. 4C and D). The increased EPC and mononuclear cell intracellular ROS production were effectively blocked by NAC in 1 week and 6 month PM + HFD + NAC mice (Fig. 4C and D). Of note, ROS production increased in 1 week and 6 month PM + ND mice and 6 month HFD mice compared with 1 week and 6 month ND mice. There was no ROS elevation in 1-week HFD compared with ND mice (Fig. 4A-D). These results indicated that NAC prevented atherosclerosis formation mainly through inhibition of ROS production in hyperlipidemic mice following PM exposure.

NAC effectively inhibits plasma inflammatory cytokine production in hyperlipidemic mice with acute and chronic PM exposure

Our previous study reported that NAC exerts powerful anti-inflammatory effects in WT mice with PM exposure (21). Therefore, the present study investigated whether PM inhibits production of inflammatory cytokines, including TNF-α, IL-1β and IL-6, in LDLR KO HFD mice. Inflammatory cytokine levels were significantly increased up to 1.3-2.0-fold in LDLR KO 1-week and 6-month PM + HFD mice compared with mice with either HFD or PM + ND (Fig. 5). NAC effectively prevented production of all three cytokines in PM + HFD mice (Fig. 5).

Discussion

The present study demonstrated that PM exposure potentiated atherosclerosis formation by promoting ox-LDL levels and inflammatory cytokine production and decreasing circulating EPC levels in hyperlipidemic mice. The mechanisms primarily involved elevated ROS production. NAC effectively reversed both acute and chronic PM exposure-potentiated effects in LDLR KO HFD mice.

Cardiovascular risk factors, such as hyperlipidemia, diabetes and age are associated with incomplete revascularization and decreased re-reendothelialization following arterial injury in both humans and animals (3335). Chronic hyperlipidemia exerts harmful effects on the cardiovascular system, including aggravation of atherosclerosis due to elevation of ox-LDL and TG levels and decreased HDL (36). In addition, short-term hyperlipidemia affects organs primarily via initiating inflammation (3739). On the other hand, PM exposure is associated with atherosclerosis development in both human and animal studies (1,40). The mechanisms primarily involved in PM-potentiated atherosclerosis include early changes in vascular tone via elevated oxidative stress and inflammation, cholesterol modification and promotion of pro-thrombotic reactions (40). The present data indicated that acute and chronic PM exposure in hyperlipidemic mice further increased plasma ox-LDL and inflammatory cytokine levels and decreased circulating EPCs, which serve an important role in endothelial repair (1). The mechanisms primarily involve increased plasma ROS as well as EPC and mononuclear intracellular ROS production. In addition, males have a higher risk for CVD than females; females are better protected than males against CVD due to undefined mechanisms (41). Our previous study demonstrated that CD34+/CD133+ EPC population was selectively decreased in male mice following PM exposure via elevated oxidative stress, while PM exposure had no effects on estrogen-independent EPC changes in female mice (42). PM exposure-induced decrease in EPC population in male mice may be due to decreased expression of pulmonary superoxide dismutase 1 (43). Therefore, the regulation of anti-atherosclerotic mechanisms in females requires further investigation. To the best of our knowledge, there is no way to determine the exact amount of PM in respiratory tissue following exposure; however, our previous study indicated that there is increased inflammation in murine pulmonary tissue following PM exposure compared with control (21).

NAC is an antioxidant with therapeutic value for decreasing endothelial dysfunction, inflammation, fibrosis, invasion, cartilage erosion, acetaminophen detoxification and prolong transplant life (44). In cardiovascular studies, NAC protects diabetic heart at risk of myocardial infarction and prevents ischemia- and non-ischemia-associated cardiac damage and ischemia-reperfusion-induced injury primarily via inhibition of oxidative stress in humans (22,45). NAC is also reported to prevent hypertrophy and fibrosis in β-MyHC-Q403 transgenic rabbits and cTnT-Q92 transgenic mice with hypertrophic cardiomyopathy via multiple thiol-responsive mechanisms (46) and inhibit platelet aggregation and reperfusion injury in both human and animals (47). In accordance with our and other previous studies (23,4850), the present study demonstrated that NAC had no effect on lipid profile in hyperlipidemia, whereas Korou et al (51) found NAC decreases serum LDL levels in hyperlipidemic mice. This difference may be due to different administration methods. Additionally, NAC decreases ox-LDL levels via inhibition of oxidative stress; to the best of our knowledge, however, the detailed mechanism has not previously been investigated (5254). NAC prevents atherosclerosis formation via suppression of inflammation by inhibiting NF-κB-induced TNF-α production and blood homocysteine levels (55,56). NAC maintains EPC population, decrease conversion of LDL to ox-LDL in vivo, prevent atherosclerosis in humans and mice with hyperlipidemia and promote hind limb ischemia recovery in mice (15,23,30,57).

NAC exhibits protective effects against PM-associated damage in different organs (1). NAC could abolishes PM-induced suppression of lymphocyte function in rats and mice (58), inhibits pulmonary inflammation in mice following 5 h and 1 month PM exposure (21,59) and prevents PM-induced ROS production in human endothelial cells (60). To the best of our knowledge, however, the effect of NAC on air pollution-potentiated atherosclerosis has not been reported. Certain studies have reported that NAC prevents senescence of porcine coronary artery endothelial cells in vitro (61), attenuates carbon monoxide-induced ischemic heart failure (62), prevents cardiomyocyte apoptosis (63) and inhibits vascular smooth muscle proliferation (64) and heart arrhythmia (65) in rats following PM exposure. The mechanisms include inhibition of oxidative stress, deactivation of c-Jun N-terminal kinase, p38 MAP kinase and inhibition of inflammation via prevention of NF-κB transcription factor activity (44). NAC prevents apoptosis and promotes cell survival via extracellular signal-regulated kinase activation (44). Our previous studies indicated that NAC inhibits EPC apoptosis, promotes BMSC proliferation and inhibits inflammatory cytokine production via blocking ROS generation in mice following PM exposure for 1 month (21,66). The present data showed that NAC effectively attenuated PM-potentiated atherosclerosis formation via inhibition of plasma ROS-induced ox-LDL elevation and mononuclear cell and EPC intracellular ROS-induced decreases in circulating EPC levels. Of note, blood inflammatory cytokine levels in mice with hyperlipidemia following 1 week or 6 month PM exposure were also significantly decreased by NAC treatment.

Further studies are needed to determine the mechanism of NAC against PM-potentiated atherosclerosis including that underlying NAC-induced decrease in ox-LDL levels, inability of NAC to completely prevent atherosclerosis development and the cell signaling pathways involved in NAC suppression of inflammatory cytokine production.

In conclusion, the present study demonstrated that NAC effectively prevented PM-potentiated atherosclerosis via inhibition of plasma ROS-induced ox-LDL elevation and mononuclear cell and EPC intracellular ROS-induced decrease in circulating EPC levels, as well as suppression of inflammatory cytokine production.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Nature Science Foundation of China (grant nos. 81600222 and 81800255), Young Experts of Taishan Scholar Program of Shandong Province (grant no. tsqn201812142), Academic Promotion Programme of Shandong First Medical University (grant nos. 2019RC017), The Natural Science Foundation of Shandong Province (grant nos. ZR2016HM22, ZR2018BH002, ZR2020MH044 and ZR2021MH112) and Clinical Medical Science and Technology Innovation Development Plan Project of Jinan in China (grant no. 201704106).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

YQC, XCM and LQC designed the experiments. YXX, HRB, YFJ, QWL, XWG and XQZ performed the experiments. KH, ZHS, QYZ, YC and HHS collected and analyzed the data. YQC and YXX wrote the manuscript. All authors have read and approved the final manuscript. YQC, XCM and YXX confirm the authenticity of all the raw data.

Ethics approval and consent to participate

All animal procedures were performed in accordance with the Guidelines of the Animal Care Committee of the Shandong Provincial Hospital affiliated to Shandong First Medical University (Jinan, China). The Animal Care Committee of Shandong Provincial Hospital affiliated to Shandong First Medical University (Jinan, China) approved the experimental protocols (approval no. 2021-228).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Cui Y, Sun Q and Liu Z: Ambient particulate matter exposure and cardiovascular diseases: A focus on progenitor and stem cells. J Cell Mol Med. 20:782–793. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Liu Y, Pan J, Fan C, Xu R, Wang Y, Xu C, Xie S, Zhang H, Cui X, Peng Z, et al: Short-term exposure to ambient air pollution and mortality from myocardial infarction. J Am Coll Cardiol. 77:271–281. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Shah AS, Lee KK, McAllister DA, Hunter A, Nair H, Whiteley W, Langrish JP, Newby DE and Mills NL: Short term exposure to air pollution and stroke: Systematic review and meta-analysis. BMJ. 350:h12952015. View Article : Google Scholar : PubMed/NCBI

4 

Rajagopalan S, Al-Kindi SG and Brook RD: Air pollution and cardiovascular disease: JACC state-of-the-art review. J Am Coll Cardiol. 72:2054–2070. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Sun Q, Wang A, Jin X, Natanzon A, Duquaine D, Brook RD, Aguinaldo JG, Fayad ZA, Fuster V, Lippmann M, et al: Long-term air pollution exposure and acceleration of atherosclerosis and vascular inflammation in an animal model. JAMA. 294:3003–3010. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Davignon J and Ganz P: Role of endothelial dysfunction in atherosclerosis. Circulation. 109 (23 Suppl 1):III27–III32. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Heitzer T, Schlinzig T, Krohn K, Meinertz T and Munzel T: Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease. Circulation. 104:2673–2678. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D, Ramaswami P, Pippen AM, Annex BH, Dong C and Taylor DA: Aging, progenitor cell exhaustion, and atherosclerosis. Circulation. 108:457–463. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Strehlow K, Werner N, Berweiler J, Link A, Dirnagl U, Priller J, Laufs K, Ghaeni L, Milosevic M, Böhm M and Nickenig G: Estrogen increases bone marrow-derived endothelial progenitor cell production and diminishes neointima formation. Circulation. 107:3059–3065. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Urbich C and Dimmeler S: Endothelial progenitor cells: Characterization and role in vascular biology. Circ Res. 95:343–353. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Werner N, Junk S, Laufs U, Link A, Walenta K, Bohm M and Nickenig G: Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ Res. 93:e17–e24. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Chen JZ, Zhang FR, Tao QM, Wang XX and Zhu JH and Zhu JH: Number and activity of endothelial progenitor cells from peripheral blood in patients with hypercholesterolaemia. Clin Sci (Lond). 107:273–280. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM and Dimmeler S: Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ Res. 89:E1–E7. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Kattoor AJ, Pothineni NVK, Palagiri D and Mehta JL: Oxidative stress in atherosclerosis. Curr Atheroscler Rep. 19:422017. View Article : Google Scholar : PubMed/NCBI

15 

Cui Y, Narasimhulu CA, Liu L, Li X, Xiao Y, Zhang J, Xie X, Hao H, Liu JZ, He G, et al: Oxidized low-density lipoprotein alters endothelial progenitor cell populations. Front Biosci (Landmark Ed). 20:975–988. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Auten RL and Davis JM: Oxygen toxicity and reactive oxygen species: The devil is in the details. Pediatr Res. 66:121–127. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Ogasawara MA and Zhang H: Redox regulation and its emerging roles in stem cells and stem-like cancer cells. Antioxid Redox Signal. 11:1107–1122. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Pervaiz S, Taneja R and Ghaffari S: Oxidative stress regulation of stem and progenitor cells. Antioxid Redox Signal. 11:2777–2789. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Cominacini L, Garbin U, Pasini AF, Davoli A, Campagnola M, Pastorino AM, Gaviraghi G and Lo Cascio V: Oxidized low-density lipoprotein increases the production of intracellular reactive oxygen species in endothelial cells: Inhibitory effect of lacidipine. J Hypertens. 16:1913–1919. 1998. View Article : Google Scholar : PubMed/NCBI

20 

Vasconcelos EM, Degasperi GR, de Oliveira HC, Vercesi AE, de Faria EC and Castilho LN: Reactive oxygen species generation in peripheral blood monocytes and oxidized LDL are increased in hyperlipidemic patients. Clin Biochem. 42:1222–1227. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Cui Y, Xie X, Jia F, He J, Li Z, Fu M, Hao H, Liu Y, Liu JZ, Cowan PJ, et al: Ambient fine particulate matter induces apoptosis of endothelial progenitor cells through reactive oxygen species formation. Cell Physiol Biochem. 35:353–363. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Dludla PV, Dias SC, Obonye N, Johnson R, Louw J and Nkambule BB: A systematic review on the protective effect of N-Acetyl cysteine against diabetes-associated cardiovascular complications. Am J Cardiovasc Drugs. 18:283–298. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Cui Y, Narasimhulu CA, Liu L, Zhang Q, Liu PZ, Li X, Xiao Y, Zhang J, Hao H, Xie X, et al: N-acetylcysteine inhibits in vivo oxidation of native low-density lipoprotein. Sci Rep. 5:163392015. View Article : Google Scholar : PubMed/NCBI

24 

Mutlu GM, Green D, Bellmeyer A, Baker CM, Burgess Z, Rajamannan N, Christman JW, Foiles N, Kamp DW, Ghio AJ, et al: Ambient particulate matter accelerates coagulation via an IL-6-dependent pathway. J Clin Invest. 117:2952–2961. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Gavett SH, Haykal-Coates N, Highfill JW, Ledbetter AD, Chen LC, Cohen MD, Harkema JR, Wagner JG and Costa DL: World Trade Center fine particulate matter causes respiratory tract hyperresponsiveness in mice. Environ Health Perspect. 111:981–991. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Shadie AM, Herbert C and Kumar RK: Ambient particulate matter induces an exacerbation of airway inflammation in experimental asthma: Role of interleukin-33. Clin Exp Immunol. 177:491–499. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Holt GR: Effects of air pollution on the upper aerodigestive tract. Otolaryngol Head Neck Surg. 114:201–204. 1996. View Article : Google Scholar : PubMed/NCBI

28 

Aggarwal DJ, Kathariya MG and Verma DPK: LDL-C, NON-HDL-C and APO-B for cardiovascular risk assessment: Looking for the ideal marker. Indian Heart J. 73:544–548. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Zhuo X, Bu H, Hu K, Si Z, Chen L, Chen Y, Yang L, Jiang Y, Xu Y, Zhao P, et al: Differences in the reaction of hyperlipidemia on different endothelial progenitor cells based on sex. Biomed Rep. 15:642021. View Article : Google Scholar : PubMed/NCBI

30 

Cui Y, Liu L, Xiao Y, Li X, Zhang J, Xie X, Tian J, Sen CK, He X, Hao H and Liu Z: N-acetylcysteine differentially regulates the populations of bone marrow and circulating endothelial progenitor cells in mice with limb ischemia. Eur J Pharmacol. 881:1732332020. View Article : Google Scholar : PubMed/NCBI

31 

Rosenkranz AR, Schmaldienst S, Stuhlmeier KM, Chen W, Knapp W and Zlabinger GJ: A microplate assay for the detection of oxidative products using 2′,7′-dichlorofluorescin-diacetate. J Immunol Methods. 156:39–45. 1992. View Article : Google Scholar : PubMed/NCBI

32 

Eggermann J, Kliche S, Jarmy G, Hoffmann K, Mayr-Beyrle U, Debatin KM, Waltenberger J and Beltinger C: Endothelial progenitor cell culture and differentiation in vitro: A methodological comparison using human umbilical cord blood. Cardiovasc Res. 58:478–486. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Ii M, Takenaka H, Asai J, Ibusuki K, Mizukami Y, Maruyama K, Yoon YS, Wecker A, Luedemann C, Eaton E, et al: Endothelial progenitor thrombospondin-1 mediates diabetes-induced delay in reendothelialization following arterial injury. Circ Res. 98:697–704. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Kinnaird T, Stabile E, Zbinden S, Burnett MS and Epstein SE: Cardiovascular risk factors impair native collateral development and may impair efficacy of therapeutic interventions. Cardiovasc Res. 78:257–264. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Sodha NR, Clements RT, Boodhwani M, Xu SH, Laham RJ, Bianchi C and Sellke FW: Endostatin and angiostatin are increased in diabetic patients with coronary artery disease and associated with impaired coronary collateral formation. Am J Physiol Heart Circ Physiol. 296:H428–H434. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Panahi Y, Ahmadi Y, Teymouri M, Johnston TP and Sahebkar A: Curcumin as a potential candidate for treating hyperlipidemia: A review of cellular and metabolic mechanisms. J Cell Physiol. 233:141–152. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Krauzova E, Kracmerova J, Rossmeislova L, Mališová L, Tencerová M, Koc M, Štich V and Šiklová M: Acute hyperlipidemia initiates proinflammatory and proatherogenic changes in circulation and adipose tissue in obese women. Atherosclerosis. 250:151–157. 2016. View Article : Google Scholar : PubMed/NCBI

38 

He L, Hao L, Fu X, Huang M and Li R: Severe hypertriglyceridemia and hypercholesterolemia accelerating renal injury: A novel model of type 1 diabetic hamsters induced by short-term high-fat/high-cholesterol diet and low-dose streptozotocin. BMC Nephrol. 16:512015. View Article : Google Scholar : PubMed/NCBI

39 

Krysiak R, Zmuda W, Marek B and Okopien B: Comparison of the effects of short-term hypolipidaemic treatment on plasma adipokine levels in men and women with isolated hypercholesterolaemia. Endokrynol Pol. 66:114–120. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Bevan GH, Al-Kindi SG, Brook R and Rajagopalan S: Ambient air pollution and atherosclerosis: Recent updates. Curr Atheroscler Rep. 23:632021. View Article : Google Scholar : PubMed/NCBI

41 

Kander MC, Cui Y and Liu Z: Gender difference in oxidative stress: A new look at the mechanisms for cardiovascular diseases. J Cell Mol Med. 21:1024–1032. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Liu X, Xiao Y, Zhu Q, Cui Y, Hao H, Wang M, Cowan PJ, Korthuis RJ, Li G, Sun Q and Liu Z: Circulating endothelial progenitor cells are preserved in female mice exposed to ambient fine particulate matter independent of estrogen. Int J Mol Sci. 22:72002021. View Article : Google Scholar : PubMed/NCBI

43 

Liu X, Wang A, Chen Z, Cui Y, Hao H, Domeier TL, Sun Q and Liu Z: Tempol preserves endothelial progenitor cells in male mice with ambient fine particulate matter exposure. Biomedicines. 10:3272022. View Article : Google Scholar : PubMed/NCBI

44 

Zafarullah M, Li WQ, Sylvester J and Ahmad M: Molecular mechanisms of N-acetylcysteine actions. Cell Mol Life Sci. 60:6–20. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Bartekova M, Barancik M, Ferenczyova K and Dhalla NS: Beneficial effects of N-acetylcysteine and N-mercaptopropionylglycine on ischemia reperfusion injury in the heart. Curr Med Chem. 25:355–366. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Marian AJ: Experimental therapies in hypertrophic cardiomyopathy. J Cardiovasc Transl Res. 2:483–492. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Nikbakht M, Ahmadi F, Vaseghi G, Talasaz AH, Eshraghi A, Naderi J and Daneshmehr MA: The role of N-acetylcysteine in platelet aggregation and reperfusion injury in recent years. Curr Clin Pharmacol. 12:83–91. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Lin CP, Huang PH, Lai CF, Chen JW, Lin SJ and Chen JS: Simvastatin attenuates oxidative stress, NF-κB activation, and artery calcification in LDLR-/-mice fed with high fat diet via down-regulation of tumor necrosis factor-α and TNF receptor 1. PLoS One. 10:e01436862015. View Article : Google Scholar : PubMed/NCBI

49 

Subrahmanian S, Varshney R, Subramani K, Murphy B, Woolington S and Ahamed J: N-Acetylcysteine inhibits aortic stenosis progression in a murine model by blocking Shear-induced activation of platelet latent transforming growth factor beta 1. Antioxid Redox Signal. Dec 7–2021.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

50 

Spartalis M, Siasos G, Mastrogeorgiou M, Spartalis E, Kaminiotis VV, Mylonas KS, Kapelouzou A, Kontogiannis C, Doulamis IP, Toutouzas K, et al: The effect of per os colchicine administration in combination with fenofibrate and N-acetylcysteine on triglyceride levels and the development of atherosclerotic lesions in cholesterol-fed rabbits. Eur Rev Med Pharmacol Sci. 25:7765–7776. 2021.PubMed/NCBI

51 

Korou LM, Agrogiannis G, Pantopoulou A, Vlachos IS, Iliopoulos D, Karatzas T and Perrea DN: Comparative antilipidemic effect of N-acetylcysteine and sesame oil administration in diet-induced hypercholesterolemic mice. Lipids Health Dis. 9:232010. View Article : Google Scholar : PubMed/NCBI

52 

Meng XP, Yin CS, Cui JH, Li ZX, Wang L, Wang YW and Li YL: Inhibitory effect of N-acetylcysteine upon atherosclerotic processes in rabbit carotid. Zhonghua Yi Xue Za Zhi. 89:1850–1853. 2009.(In Chinese). PubMed/NCBI

53 

Sung HJ, Kim J, Kim Y, Jang SW and Ko J: N-acetyl cysteine suppresses the foam cell formation that is induced by oxidized low density lipoprotein via regulation of gene expression. Mol Biol Rep. 39:3001–3007. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Thiele H, Hildebrand L, Schirdewahn C, Eitel I, Adams V, Fuernau G, Erbs S, Linke A, Diederich KW, Nowak M, et al: Impact of high-dose N-acetylcysteine versus placebo on contrast-induced nephropathy and myocardial reperfusion injury in unselected patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. The LIPSIA-N-ACC (Prospective, Single-Blind, Placebo-Controlled, Randomized Leipzig Immediate PercutaneouS Coronary Intervention Acute Myocardial Infarction N-ACC) Trial. J Am Coll Cardiol. 55:2201–2209. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Poitevin S, Garnotel R, Antonicelli F, Gillery P and Nguyen P: Type I collagen induces tissue factor expression and matrix metalloproteinase 9 production in human primary monocytes through a redox-sensitive pathway. J Thromb Haemost. 6:1586–1594. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Wiklund O, Fager G, Andersson A, Lundstam U, Masson P and Hultberg B: N-acetylcysteine treatment lowers plasma homocysteine but not serum lipoprotein(a) levels. Atherosclerosis. 119:99–106. 1996. View Article : Google Scholar : PubMed/NCBI

57 

Zhu Q, Hao H, Xu H, Fichman Y, Cui Y, Yang C, Wang M, Mittler R, Hill MA, Cowan PJ, et al: Combination of antioxidant enzyme overexpression and N-acetylcysteine treatment enhances the survival of bone marrow mesenchymal stromal cells in ischemic limb in mice with type 2 diabetes. Am Heart Assoc. 10:e0234912021. View Article : Google Scholar : PubMed/NCBI

58 

Omara FO, Fournier M, Vincent R and Blakley BR: Suppression of rat and mouse lymphocyte function by urban air particulates (Ottawa dust) is reversed by N-acetylcysteine. J Toxicol Environ Health A. 59:67–85. 2000. View Article : Google Scholar : PubMed/NCBI

59 

Rhoden CR, Lawrence J, Godleski JJ and Gonzalez-Flecha B: N-acetylcysteine prevents lung inflammation after short-term inhalation exposure to concentrated ambient particles. Toxicol Sci. 79:296–303. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Wang T, Wang L, Moreno-Vinasco L, Lang GD, Siegler JH, Mathew B, Usatyuk PV, Samet JM, Geyh AS, Breysse PN, et al: Particulate matter air pollution disrupts endothelial cell barrier via calpain-mediated tight junction protein degradation. Part Fibre Toxicol. 9:352012. View Article : Google Scholar : PubMed/NCBI

61 

Sharma K, Lee HH, Gong DS, Park SH, Yi E, Schini-Kerth V and Oak MH: Fine air pollution particles induce endothelial senescence via redox-sensitive activation of local angiotensin system. Environ Pollut. 252((Pt A)): 317–329. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Reboul C, Boissiere J, Andre L, Meyer G, Bideaux P, Fouret G, Feillet-Coudray C, Obert P, Lacampagne A, Thireau J, et al: Carbon monoxide pollution aggravates ischemic heart failure through oxidative stress pathway. Sci Rep. 7:397152017. View Article : Google Scholar : PubMed/NCBI

63 

Kim JB, Kim C, Choi E, Park S, Park H, Pak HN, Lee MH, Shin DC, Hwang KC and Joung B: Particulate air pollution induces arrhythmia via oxidative stress and calcium calmodulin kinase II activation. Toxicol Appl Pharmacol. 259:66–73. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Tzeng HP, Yang RS, Ueng TH and Liu SH: Upregulation of cyclooxygenase-2 by motorcycle exhaust particulate-induced reactive oxygen species enhances rat vascular smooth muscle cell proliferation. Chem Res Toxicol. 20:1170–1176. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Rhoden CR, Wellenius GA, Ghelfi E, Lawrence J and Gonzalez-Flecha B: PM-induced cardiac oxidative stress and dysfunction are mediated by autonomic stimulation. Biochim Biophys Acta. 1725:305–313. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Cui Y, Jia F, He J, Xie X, Li Z, Fu M, Hao H, Liu Y, Liu DZ, Cowan PJ, et al: Ambient fine particulate matter suppresses in vivo proliferation of bone marrow stem cells through reactive oxygen species formation. PLoS One. 10:e01273092015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2022
Volume 26 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu Y, Bu H, Jiang Y, Zhuo X, Hu K, Si Z, Chen Y, Liu Q, Gong X, Sun H, Sun H, et al: N‑acetyl cysteine prevents ambient fine particulate matter‑potentiated atherosclerosis via inhibition of reactive oxygen species‑induced oxidized low density lipoprotein elevation and decreased circulating endothelial progenitor cell. Mol Med Rep 26: 236, 2022
APA
Xu, Y., Bu, H., Jiang, Y., Zhuo, X., Hu, K., Si, Z. ... Cui, Y. (2022). N‑acetyl cysteine prevents ambient fine particulate matter‑potentiated atherosclerosis via inhibition of reactive oxygen species‑induced oxidized low density lipoprotein elevation and decreased circulating endothelial progenitor cell. Molecular Medicine Reports, 26, 236. https://doi.org/10.3892/mmr.2022.12752
MLA
Xu, Y., Bu, H., Jiang, Y., Zhuo, X., Hu, K., Si, Z., Chen, Y., Liu, Q., Gong, X., Sun, H., Zhu, Q., Cui, L., Ma, X., Cui, Y."N‑acetyl cysteine prevents ambient fine particulate matter‑potentiated atherosclerosis via inhibition of reactive oxygen species‑induced oxidized low density lipoprotein elevation and decreased circulating endothelial progenitor cell". Molecular Medicine Reports 26.1 (2022): 236.
Chicago
Xu, Y., Bu, H., Jiang, Y., Zhuo, X., Hu, K., Si, Z., Chen, Y., Liu, Q., Gong, X., Sun, H., Zhu, Q., Cui, L., Ma, X., Cui, Y."N‑acetyl cysteine prevents ambient fine particulate matter‑potentiated atherosclerosis via inhibition of reactive oxygen species‑induced oxidized low density lipoprotein elevation and decreased circulating endothelial progenitor cell". Molecular Medicine Reports 26, no. 1 (2022): 236. https://doi.org/10.3892/mmr.2022.12752