Polystyrene Microplastics Induced Cardiotoxicity in Chic - 2022 - Science of The

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Science of the Total Environment 840 (2022) 156727

Contents lists available at ScienceDirect

Science of the Total Environment


journal homepage: www.elsevier.com/locate/scitotenv

Polystyrene microplastics-induced cardiotoxicity in chickens via the


ROS-driven NF-κB-NLRP3-GSDMD and AMPK-PGC-1α axes
Yue Zhang, Kai Yin, Dongxu Wang, Yu Wang, Hongmin Lu, Hongjing Zhao , Mingwei Xing
⁎ ⁎
College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• PS-MPs exposure triggered oxidative


stress and ROS overload in myocardium.
• PS-MPs induced cardiac pyroptosis and in-
flammation by the NF-κB-NLRP3-GSDMD
axis.
• PS-MPs caused abnormal mitochondrial
and energy metabolism by the AMPK-
PGC-1α axis.
• PS-MPs elicited cardiotoxicity in chickens
may depend on the driving effect of ROS.

A R T I C L E I N F O A B S T R A C T

Editor: Henner Hollert Microplastics (MPs) pollution is getting increasingly prominent, and its dangers have attracted widespread attention.
The heart is the central hub of the organism's survival, and the mechanism of MPs-induced heart injury in chickens is
Keywords: unknown. Here, we investigated the effects of 5 μm polystyrene microplastics (PS-MPs) on the heart and primary car-
Microplastic
diomyocytes of chickens at varied concentrations. We observed that PS-MPs caused severe pathological damage and
Cardiotoxicity
ultrastructural changes in heart, induced myocardial pyroptosis, inflammatory cell infiltration and mitochondrial le-
Reactive oxygen species
NF-κB-NLRP3-GSDMD axis
sions. PS-MPs evoked abnormal antioxidant enzyme content and ROS overproduction. Detailed mechanistic investiga-
AMPK-PGC-1α pathway tion indicated that PS-MPs triggered pyroptosis via NF-κB-NLRP3-GSDMD axis and exacerbated myocardial
Chickens inflammation (NLRP3, Caspase-1, IL-1β, IL-18, ASC, GSDMD, NF-κB, COX-2, iNOS and IL-6 overexpression). Addition-
ally, PS-MPs induced mitochondrial damage (TFAM, OPA1, MFN1 and MFN2 down-expression, DRP1 and Fis1 over-
expression) and energy metabolism disorders (HK2, PKM2, PDHX and LDH up-regulation) by inhibiting AMPK-PGC-
1α pathway. Interestingly, NAC alleviated these aberrant manifestations in vitro. We suggested that PS-MPs driven al-
terations in NF-κB-NLRP3-GSDMD and AMPK-PGC-1α pathways via ROS overload, which in turn triggered oxidative
stress, myocardial pyroptosis, inflammation, mitochondrial and energy metabolism dysfunction. This provided theo-
retical bases for protecting chickens from toxic injury by MPs.

1. Introduction (MPs) have smaller particle size (size < 5 mm), stronger adsorption capacity
and greater difficulty in degradation, thus posing a serious pollution and
With the increasing amount of “white waste”, its harm has gotten a lot threat to the environment (Xu et al., 2020; Yin et al., 2021a). MPs have
of focus. However, compared with the “white plastic”, microplastics been detected in the worldwide environment (Schwabl et al., 2019; C. Li
et al., 2020). According to statistics, approximately 700 species of marine
⁎ Corresponding authors. organisms have been contaminated with MPs (Wright et al., 2013; Avio
E-mail addresses: zhaohongjing@nefu.edu.cn (H. Zhao), xingmingwei@nefu.edu.cn et al., 2015). As research continues, experts have discovered that the accu-
(M. Xing). mulation of MPs on the organism can have irreversible toxic effects on

http://dx.doi.org/10.1016/j.scitotenv.2022.156727
Received 1 April 2022; Received in revised form 23 May 2022; Accepted 12 June 2022
Available online 14 June 2022
0048-9697/© 2022 Published by Elsevier B.V.
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

human and animal bodies (Wright and Kelly, 2017). Polystyrene comprehensive investigation of the detailed damage mechanisms of PS-
microplastics (PS-MPs) have been reported to accumulate in fish, rats, MPs exposure in chickens.
chickens and humans through the transmission of the food chain, causing
severe damage to various organs, such as liver necrosis, dysbiosis of intesti-
nal flora and nephritis (Lu et al., 2016; Y.L. Wang et al., 2021; Banerjee and 2. Materials and methods
Shelver, 2021). Also, PS-MPs of 3–12 μm in the concentration range of
0.5 mg/L to 100 mg/L could break the blood-brain barrier, lead to cranial 2.1. Animals and experimental design
neruritis (Prüst et al., 2020), as well as to cause sperm cell atrophy, abscis-
sion and apoptosis at all levels through the blood-testis barrier, thereby All procedures of the current experiments were approved and super-
inducing reproductive toxicity (Hou et al., 2021). It is worth mentioning vised by the Animal Protection and Utilization Committee of Northeastern
that the myocardium may also be a target organ for environmental expo- Forestry University, and the animal experiments strictly complied with the
sure to MPs. In a study of developmental toxicity in zebrafish embryos, “Guide to Animal Welfare Standards in China” and the “Regulations on the
Pitt et al. found that the toxicity of PS-MPs migrated to the heart, triggering Management of Laboratory Animals (2017)”.
a decrease in heart rate in the offspring (Pitt et al., 2018). Meanwhile, PS-MPs (5 μm) purchased from Tesulang Chemical Company, China.
recent studies have confirmed that PS-MPs promoted myocardial apoptosis Sixty one-day-old chicks (a hatchery in Harbin, Heilongjiang Province,
further aggravating its fibrosis (Z. Li et al., 2020). The health of waterfowl China) were randomly divided into four groups (15 chicks per group).
and birds is also at risk from MPs, mainly in the form of intestinal damage Control group (Con): drinking normal water with neither PS-MPs, low-
and impaired embryonic development (Banerjee and Shelver, 2021; L. dose group (L-MPS): drinking water containing 1 mg/L PS-MPs, which
Wang et al., 2021). Nevertheless, the effects of MPs accumulation on myo- was determined based on previous reports, as an environment-related
cardial injury in avian or bird and their potential molecular mechanisms concentration (Sun et al., 2021a; Sun et al., 2021b). Medium-dose group
have not been clarified. (M-MPS): drinking water containing 10 mg/L PS-MPs, and high-dose
Reactive oxygen species (ROS) plays an important role in regulating the group (H-MPS): drinking water containing 100 mg/L PS-MPs. PS-MPs
physiological functions of the body (Yang et al., 2019). Available evidence was dissolved in ultrapure water and placed in an ultrasonic shaker for
has shown that ROS is one of the key causes of myocardial cell cycle arrest 2 h, 1 h and 30 min, followed by multiple resuspensions between each treat-
(Pei et al., 2020), and triggering ROS can cause lipid peroxidation, leading ment. All chickens were kept in a well-ventilated space with free food and
to disruption of cell membrane structure and cell death (Paulus and drinking water. The temperature of the site was 33 °C, the humidity was
Tschöpe, 2013). Pyroptosis, also named “inflammatory cell necrosis”, is a 65 %, and the light was about 16 h/day. After 7 days of adaptation, the
recently discovered new form of programmed cell death (Shi et al., 2017; temperature was reduced by 1 °C every 3 days, the humidity was reduced
Q. Liu et al., 2022; Gu et al., 2022). Reportedly, the increased production by 5 % per week, and the final temperature was controlled at 22 °C, the
of ROS upregulated NF-κB expression (X.J. Liu et al., 2022), which in humidity was maintained at 55 %. After 42 days, all chickens were eutha-
turn facilitated the activation of NLRP3 inflammasome (Jing et al., 2022), nized, fresh heart samples were extracted, some of the tissue was placed
IL-1β, IL-18 precursor and Caspase-1. Activated Caspase-1, on the one in 4 % paraformaldehyde fixative, and the other part was divided and
hand, cleaved gasdermin-D (GSDMD) to form a peptide containing the labeled, then placed in a − 80 °C refrigerator for subsequent use.
active region of GSDMD-N, induced cell membrane perforation rupture,
released its contents, and triggered pyroptosis (Sun et al., 2019), on the
2.2. Histopathological observation of the heart
other hand, cleaved IL-1β and IL-18 precursors, and distributed them extra-
cellularly, recruiting inflammatory cells to aggregate and amplify the
Hearts were placed in tissue fixative for 24 h. Dehydrated in graded
inflammatory response (Dai et al., 2021).
ethanol and dewaxed in xylene as described by previous experience and
It is well known that the process of cell death is accompanied by al-
the experimental manipulations of Chen et al. (Chen et al., 2022), 5 μm
terations in mitochondrial homeostasis (Horng, 2014). Recent reports
sections were made, followed by hematoxylin eosin (H&E) staining and
have confirmed that exposure to environmental pollutants stimulated
placed under light microscopy for observation.
strong alterations in the AMPK-PGC-1α pathway and induced mitochon-
drial dysfunction (Yang et al., 1987; Cai et al., 2021). And previously,
experimental data by Li et al. simultaneously suggested that ROS- 2.3. Transmission electron microscopy (TEM) observation
AMPK-PGC-1α may be a regulatory site involved in MPs-induced glyco-
lytic metabolism (Li et al., 2021). Alterations in mitochondrial homeo- For the examination of the ultrastructure of the heart tissue, we used the
stasis may affect normal organismal metabolism (Coppi et al., 2021). previous experimental method. 1 mm3 sections of the heart were manipu-
Therefore, we have reason to suspect that the AMPK-PGC-1α pathway lated following the experimental procedure of Zhao et al. (2021). The
plays a key role in PS-MPs induced myocardial mitochondrial energy micrographs were obtained at 80 kV using a transmission electron micro-
metabolism. scope (GEM1200ES, Japan).
Cell death is an essential physiological process in the organism
(Lin et al., 2016). Past studies on myocardial injury have generally focused
on apoptosis (Kung et al., 2011), whereas the role of pyroptosis as a new 2.4. Cardiomyocyte culture and related treatment
mode of cell death and related pathways crosstalk in cardiac diseases has
not been extensively studied, and the specific mechanisms of MPs to induce Extraction and culture of primary chickens embryonic myocardial cells
this cardiotoxicity is not clear. Considering that the effects of PS-MPs according to relevant literature guidance and our appropriate improve-
exposure in the field/natural environment on chickens have not been ments (Yang et al., 2017). We chose 12 or 13-day-old sterile chickens
mentioned. Thus, we chose PS-MPs (5 μm), the most widely applied in embryos for extraction. Heart tissue (Lower third of the heart tip) was re-
environmental pollution studies and constructed experimental models of moved from intact chickens embryos, digested with 0.1 % Collagenase 1II
chickens exposed to different concentrations of PS-MPs for 42 days. More (Biosharp, China), and cardiomyocytes were cultured in DMEM/F12
importantly, we further evaluated the cardiotoxicity of PS-MPs by culturing (Gibco, China) supplemented with 10 % fetal bovine serum (FBS, Biological
primary chicken embryo cardiomyocytes in vitro. We hypothesized that PS- Industries) and 1 % penicillin-streptomycin (Beyotime) at in a humidified
MPs would induce chickens cardiotoxicity via ROS-driven NF-κB-NLRP3- incubator with 5 % CO2 at 37 °C for 48 h. When cardiomyocyte fusion
GSDMD and AMPK-PGC-1α axis systems, possibly involving programmed reaches 80 %, the growth medium was removed and DEMF/F12 containing
impairment of oxidative stress, pyroptosis, inflammation, mitochondria 2 % FBS, 1 % penicillin-streptomycin medium was added prior to PS-MPs
and energy metabolism. This work provided richer theoretical data for a and/or antioxidant 0.5 mM NAC (Beyotime) treatment (Gao et al., 2018).

2
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

2.5. Cardiomyocyte viability analysis 1:200) for 1 h. Nuclei were stained with 4′, 6-diamidino-2-phenylindole
(DAPI) (Beyotime) for 5 min (Miao et al., 2022a). Finally, images were
Counting Kit-8 (CCK-8) assay was used to detect the effect of different captured by a fluorescence microscope (Shang Guang Optical Microscope,
action times (4 h, 12 h, 24 h, and 48 h) and concentrations of NAC China). Correlation quantitative analysis using Image J.
(0.2 mM, 0.5 mM, 1 mM, 2 mM and 2.5 mM) and PS-MPs (0 mg/mL,
0.01 mg/mL, 0.05 mg/mL, 0.1 mg/mL, 0.25 mg/mL, 0.5 mg/mL, 2.9. Detection of antioxidant function
1 mg/mL, 1.5 mg/mL, 2 mg/mL, and 2.5 mg/mL) on the survival of cardio-
myocytes. The absorbance at 450 nm was measured by an enzyme marker Selection of relevant kits to measured T-AOC, CAT, GSH, SOD and MDC
according to the manufacturer's instructions. levels in heart tissues, and the operating rules strictly follow the instructions
of the kit (Jiancheng Bioengineering Institute, Nanjing, China).
2.6. Flow cytometry

2.10. Detection of ROS accumulation in cardiomyocytes


We selected Annexin V-FITC/PI Apoptosis Detection Kit (Beyotime)
to measure the damage of cardiomyocytes treated with different concentra-
We used the ROS assay kit (Nanjing Jiancheng Bioengineering Institute)
tions of PS-MPs. Cells in each group were digested with 0.1 % collagenase,
to examine its accumulation (Chi et al., 2021). The cardiomyocytes were
centrifuged at 1500 rpm/min for 5 min, 5 × 105 cardiomyocytes were
placed in medium containing DCFH-DA and incubated in a 5 % CO2 incuba-
collected, and carefully washed twice with PBS. According to the reagent
tor at 37 °C for 30 min. The images were captured by using fluorescence
instructions, add 500 μL Binding buffer to resuspend the cells, add 5 μL
microscopy and quantified by using Image J.
Annexin V-FITC and 10 μL Propidium Iodide to each tube, and mix well.
Incubate at room temperature for 15 min in the dark, and then perform
the next detection and analysis by CytoFLEX flow cytometry and CytExpert 2.11. Intracellular mitochondrial activity assay in cardiac myocytes
software (Beckman Coulier Life Sciences, China).
The cell culture medium was removed and replaced with a culture
2.7. Immunohistochemistry (IHC) medium containing 100 nM of Mito-Tracker Green (Beyotime) working
solution (Miao et al., 2022b), and the cardiomyocytes were stained in an
Paraffin-embedded heart tissue sections were deparaffinized, hydrated, incubator at 37 °C and 5 % CO2 humidity for 30 min. The absorbance of mi-
and stained with NLRP3 (WanLeiBio, 1:200) and GSDMD antibody tochondrial staining of cardiomyocytes was then quantified using Image J
(Bost Detection of antioxidant function er Biological Technology, 1:200). software.
Staining was visualized by standard peroxidase immunoassays and
counterstaining with Gill II hematoxylin. Pictures were obtained using a 2.12. Primer designs and real-time quantitative PCR (qRT-PCR)
light microscope (Olympus, Tokyo, Japan). The IHC expression level of
the target protein was quantitatively analyzed by Image J software. Similar to previous methods, total RNA from cardiac muscle tissue and
cells was obtained by TRIzol extraction (Liu et al., 2022), after which we
2.8. Immunofluorescence (IF) reverse transcribed them according to the manufacturer's instructions
(Yeasen Biotechnology, China). Meanwhile, we searched the NCBI website
The primary cardiomyocytes were treated with PS-MPs for 24 h, fixed (https://www.ncbi.nlm.nih.gov/) for the upstream and downstream
with 4 % paraformaldehyde and then blocked with a solution containing primer sequences of the desired mRNAs, as shown in Table 1. qRT-PCR
5 % BSA and 0.01 % Triton-X 100. Cardiomyocytes were washed with assays of mRNA were performed using the Applied Biosystems™ 7500
PBS after staining with primary antibodies against NLRP3 and GSDMD at (Thermo Fisher Scientific, USA) and SYBR Green Master Mix (Roche,
4 °C overnight (Xu et al., 2021) and were then further incubated with Switzerland). β-Actin was used as the internal reference gene to complete
Alexa Fluro 488 or FITC-conjugated goat anti-rabbit IgG (WanLeiBio, data normalization according to the 2−ΔΔCt method (Y. Wang et al., 2021).

Table 1
The primers used in the present study.
Gene Serial number Forward (5′–3′) Reverse (5′–3′)

NLRP3 XM_046918111.1 GCTCCTTGCGTGCTCTAAGACC TTGTGCTTCCAGATGCCGTCAG


Caspase-1 XM_040687588.2 GTGCTGCCGTGGAGACAACATAG AGGAGACAGTATCAGGCGTGGAAG
IL-1β XM_046931582.1 GAGGAGGTTTTTGAGCCCGT GCACGAAGCACTTCTGGTTG
IL-18 XM_015297948.4 CTGAAGGTGCGGTGGTTTTG TCTCGAAGCGACTGAAACAA
NF-κB NM_001396038.1 GCAGATAGCCAAGTTCAGGAT CATTTGCTTCCCTGCATTCT
IκB-α NM_001001472.3 GGAGTAGCCCTGGTAGGTCA CCTGCGTAGGTATTGCAGCTT
TNF-α MF000729.1 CTTCCTGCTGGGGTGCATAG AAGAACCAACGTGGGCATTG
COX-2 MN013407.1 TGTCCTTTCACTGCTTTCCAT TTCCATTGCTGTGTTTGAGGT
iNOS NM_204961.2 CCTGGAGGTCCTGGAAGAGT CCTGGGTTTCAGAAGTGGC
IL-6 NM_204628.2 AAGTTCACCGTGTGCGAGAA TCAGGCATTTCTCCTCGTCG
AMPK XM_046922941.1 ACCATCTGTCTCGCCCTCATCC AATGCCACTTCGCTCTTCTTACACC
PGC-1α NM_001006457.2 TGTACTTGGTTCCAAGAAAAGGA CAGACTGGTCGCTGTACCAC
TFAM NM_204100.2 GCAGCTGTTCAGGGGCTG TCAGGAAGCGGAAATAGGCG
OPA1 NM_001396172.1 AGAAGCCCTCCATCAAGAAAA ACAATGCTCCCCCAAAAGGT
DRP1 BM486811.1 TACGTCACAAGAGTCTGCGG CTCCTGTTACTGAGGCACCG
MFN1 NM_001396185.1 CAGGAGACAGAAGGCCCAAG GCTGCAGCTACGTTTATGGC
MFN2 XM_040689233.2 TGAAGCGCAATGTCCCTGTT CATTGATGTACGCAGCCAGC
Fis1 XM_040657193.2 GTCGTGACGTCATTTCACCG ACATCGTCAAACTCCGGGTC
HK2 NM_204212.2 TGCGAGATCCGATTCGTGAG TTTCAACCCTCATCCTCCGC
PKM2 NM_205469.2 CAGCAGGAGACACCGAACTC GAGGCGGGCAACATTCATTC
PDHX NM_001031187.2 GCTGGAGATGCACTGTGTGA ACCAATTAGGGAGCCCAAGC
LDH NM_205284.2 CATCACTTCCAGCACAAACG CGGTGTTTAGGAAAGCCACT
β-actin NM_205518.2 CCGCTCTATGAAGGCTACGC CTCTCGGCTGTGGTGGTGAA

3
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Table 2
The antibodies used in the present study.
Antibody name Dilution ratio kDa Resource Item number

β-Actin 1:2000 42 Abclonal Technology, China AC038


Tubulin 1:500 56 WanLei Biotechnology, China WL01931
p-IκB-α 1:350 35 WanLei Biotechnology, China WL02495
IκB-α 1:350 35 WanLei Biotechnology, China WL01936
NF-κB 1:500 65 WanLei Biotechnology, China WL01273b
IL-1β 1:350 35 WanLei Biotechnology, China WL02257
IL-18 1:350 22 WanLei Biotechnology, China WL01127
NLRP3 1:500 106 WanLei Biotechnology, China WL02635
Pro-Caspase-1 1:350 45 WanLei Biotechnology, China WL03325
Cleaved-caspase-1 1:350 20 WanLei Biotechnology, China WL03325
ASC 1:350 22 WanLei Biotechnology, China WL02462
GSDMD 1:1000 53 Boster Biotechnology, China M02842
iNOS 1:500 130 WanLei Biotechnology, China WL0992a
TNF-α 1:1000 26 Proteintech Group, USA 60291-1-Ig
COX-2 1:1000 68 WanLei Biotechnology, China WL01750
OPA1 1:1000 83 Abclonal Technology, China A9833
MFN1 1:1000 84 Boster Biotechnology, China M02172-1
MFN2 1:1000 86 Proteintech Group, USA 12186-1-AP
DRP1 1:500 80 WanLei Biotechnology, China WL03028
LDH 1:1000 35 WanLei Biotechnology, China WL03271
HK2 1:500 102 WanLei Biotechnology, China WL02454
PKM2 1:350 58 WanLei Biotechnology, China WL03290

Fig. 1. Effects of PS-MPs on myocardial tissue. Chickens were given drinking water with and/or without PS-MPs at different concentrations for 42 days. A: Body weight, heart
weight, and heart/body mass index of chickens. B: Histopathological sections of myocardium. Green arrows: inflammatory cell infiltration. Red arrows: nucleolysis. Blue
arrows: cell expansion or rupture. C: Myocardial injury area. D: Ultrastructural observation of myocardium. Green arrows: mitochondrial damage (swelling or ridge
breakage). Red arrows: loose arrangement of myofilaments. Blue arrows: bubble-like protrusions. Yellow arrows: nuclear solidification. Bar graphs indicate mean ± SD
(n = 3). **P < 0.01 and ***P < 0.001.

4
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

2.13. Western blotting (WB) 3. Results

After extraction of total proteins from myocardial tissues and cells 3.1. Changes in body weight, heart weight and organ index of chickens after
according to the method of Miao et al. (2022c), protein separation was PS-MPs exposure
performed using 8 %–15 % SDS-PAGE gels, followed by transfer of target
proteins to polyvinylidene difluoride (PVDF) membranes, which by trans- After 42 days of exposure to different concentrations of PS-MPs,
fer technique and then overnight incubation of the membranes with the chickens heart tissues were collected and weighed. There was no distinct
antibodies shown in Table 2. After proper washing, the membranes were effect of PS-MPs on body weight, myocardial weight, and heart/body
incubated with HRP-conjugated secondary antibodies (1:10,000, Biosharp, weight index of chickens (P > 0.05) (Fig. 1A).
BL003A) for 90 min (Gong et al., 2022). The protein strip images were
acquired on the ImageQuant LAS 4000 System using the ECL chemilumi- 3.2. PS-MPs promotes myocardial inflammatory injury in chickens
nescence kit (Biosharp). Finally, the optical density (OD) of the protein
bands was analyzed using Image J software. Data are presented as target To assess the effect of PS-MPs on cardiac pathological morphology, H&E
protein (OD)/β-actin or tubulin (OD). staining was used to observe the damage. The myocardial cells in the Con
group showed normal physiological structural and morphology with no
obvious damage conditions. The L-MPS group showed a small amount of in-
2.14. Statistical analysis flammatory cell infiltration and cell swelling, while the M-MPS and H-MPS
group had more obvious abnormal morphology of cardiomyocytes, such as
All data were analyzed with one or/and two-way ANOVA in IBM SPSS inflammatory cell infiltration, nucleolysis and disorganized myofilament
software (version 23, SPSS Inc., Chicago, IL, USA). Bar charts were arrangement (Fig. 1B). Quantitative analysis showed that the degree of
consisted of mean ± standard deviation (SD) of the data, which were myocardial injury was significantly higher in M-MPS (P < 0.01) and H-
replicates of at least three biological samples (n ≥ 3). P values < 0.05 MPS (P < 0.001) compared to Con, when there was no difference in L-
were considered statistically significant, *P < 0.05, **P < 0.01 and MPS (P > 0.05) (Fig. 1C). It suggested that PS-MPs exposure triggered
***P < 0.001 compared to the Con group. inflammation in the chickens myocardium.

Fig. 2. Effect of PS-MPs on myocardial tissue antioxidant enzymes, cardiomyocyte growth and ROS production. A: Expression of MDA, GSH, SOD, CAT and T-AOC in vivo. B:
ROS accumulation in chickens primary cardiomyocytes. C: The influence of different concentrations of PS-MPs on the viability of primary cardiomyocytes at
12 h, 24 h and 48 h, respectively. D: Analysis of the average optical density generated by ROS. Six independent samples (n = 6) for A (GSH) and C, three independent samples
(n = 3) performed in triplicate for others. *P < 0.05, **P < 0.01 and ***P < 0.001.

5
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Fig. 3. PS-MPs stimulation induces pyroptosis in vivo. A: IHC observation and quantitative analysis of NLRP3 and GSDMD by different concentrations of PS-MPs. B: Pyroptosis
marker mRNA levels (NLRP3, Caspase-1, IL-1β and IL-18). C: Pyroptosis marker protein levels (NLRP3, Caspase-1, IL-1β, IL-18, ASC and GSDMD). Bar graphs indicate
mean ± SD (n = 3). *P < 0.05, **P < 0.01 and ***P < 0.001.

3.3. PS-MPs enhances myocardial pyroptosis and mitochondrial damage in the toxic effect time of 12 h did not show striking changes, after 24 h and 48 h
chickens of action we observed that the cell viability presented significant inhibition at
the node of 0.25 mg/mL concentration. Cell survival rate was 85 %–75 %,
Further observation of ultrastructural changes in cardiomyocytes by 80 %–70 %, 70 %–65 %, 60 %–50 %, 50 %–40 %, 40 %–20 % at 0.25, 0.5,
TEM. We found that the organelle structure of the Con group was relatively 1.0, 1.5, 2.0 and 2.5 mg/mL PS-MPs, respectively. Allowing in vitro survival
intact. In the L-MPS group, the arrangement of muscle filaments was loose, time and sensitivity of primary cardiomyocytes. We chose the 0.25 mg/mL of
the mitochondria were slightly enlarged, and a small amount of bubble-like PS-MPs at 24 h as the lowest concentration, and 2 and 4 fold the medium and
protrusions were observed before the plasma membrane ruptured, high concentration groups to set as low dose, respectively. Briefly, four
which was formed by pyroptosis cells, also called “pyroptosis body”. In groups were divided in vitro: Con (without PS-MPS), L-PS (0.25 mg/mL
the M-MPS group, the myocardial fibers were obviously broken, the mito- PS-MPS), M-PS (0.5 mg/mL PS-MPS), H-PS (1 mg/mL PS-MPS).
chondrial ridge was also slightly deformed and broken, and the “pyroptosis Microscopic observation showed that the cells in the Con group were
body” increased. The above state was more obvious and accompanied by normal in morphology and structure, with tight junctions between cells,
nuclear pyknosis in the H-MPS group (Fig. 1D). accompanied by extension of pseudopodia. The PS-MPs-treated groups
exhibited abnormal cell morphology, including: decreased growth density,
3.4. PS-MPs inhibits cardiomyocyte growth reduced cell junctional pseudopodia, and vacuolation of cells. Among them,
cell vacuolation (yellow mark) was more obvious (Fig. S1B). Further flow
To further verify the effect of PS-MPs on myocardium, chickens embry- cytometry analysis revealed that with respect to the Con, the PS-MPs
onic primary cardiomyocytes were used as in vitro study subjects. CCK8 groups with different concentrations exhibited an inhibitory effect on
data confirmed that cardiomyocytes pretreated with 0.2–1.0 mM NAC for cardiomyocytes. NAC treatment largely rescued cardiomyocyte injury
4 h did not affect the viability of normal cells (Fig. S1A). As Fig. 2C showed, invoked by PS-MPs (Fig. S1C).

6
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Fig. 4. PS-MPs induces pyroptosis in vitro. Primary cardiomyocytes were given cultures containing and/or not containing different concentrations of PS-MPs stimulated for
24 h. A and C: The IF expression levels of NLRP3 and GSDMD in primary cardiomyocytes. B and D: Mean optical density analysis of NLRP3 and GSDMD. E: Effect of different
concentrations of PS-MPs (0, 0.25, 0.5, 1.0 mg/mL PS-MPs) with or without 0.5 mM NAC on the growth state of cardiomyocytes. F: Pyroptosis marker mRNA levels
(consistent with myocardial tissue). G: Pyroptosis marker protein levels (consistent with myocardial tissue). Data are expressed as the mean ± SD (n = 3). *P < 0.05,
**P < 0.01 and ***P < 0.001.

7
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

3.5. PS-MPs causes myocardial oxidative stress and ROS overload Then, the expression status of mitochondrial and energy metabolic fac-
tors in cardiomyocytes were tested by Mito-Tracker Green and mRNA levels
PS-MPs dramatically increased the activity of MDA and inhibited the (Fig. 8A–D). As expected, there was a significant effect of mitochondrial
expression of GSH, SOD, CAT and T-AOC compared to the Con (P < 0.05 green fluorescence and AMPK-PGC-1α pathway expression only at H-PS
to P < 0.001). Particularly, the above altered antioxidant enzyme activity (P < 0.05 to P < 0.001). Further WB analysis showed that compared with
was overall dependent on the elevated dose of PS-MPS exposure. In addi- the Con, AMPK, PGC-1α, OPA1, MFN1 and MFN2 were decreased, DRP1,
tion, we also measured the content of ROS in cardiac myocytes. Unlike HK2, PKM2 and LDH were increased in PS-MPs treated cardiomyocytes
the in vivo data, the in vitro experiments showed that even though the (Fig. 8E). Vitally, NAC treatment also restored these abnormal expressions
PS-MPs treatments showed a progressive enhancement in ROS fluorescence (Fig. 8E). Collectively, we demonstrated that PS-MPs might cause mito-
intensity (Fig. 2B), the quantitative analysis implied that there was no note- chondrial damage and impaired energy metabolism in the myocardium
worthy difference between the L-PS and M-PS versus the Con (P > 0.05), by inhibiting the AMPK-PGC-1α pathway.
and only the H-PS was statistically significant (P < 0.001). As we presumed,
adding NAC prominently reduced the activity of ROS (P < 0.05) (Fig. 2D). 4. Discussion
Taken together, PS-MPs exposure induced oxidative stress and ROS
overload, while NAC was effective in treating the excessive accumulation MPs have been shown to be “the primary instigator” of global
of ROS. environmental pollution. The size of MPS particles can influence their
inter-transfer between ecological environments (soil, water and air) (Yang
3.6. PS-MPS triggers NF-κB-NLRP3-GSDMD axis heterotopia in vivo and in vitro et al., 2021; Yin et al., 2021b). Evidence suggests that MPs reached ecosys-
tems through multiple pathways, and notably, which are far more prevalent
NLRP3 inflammasome and GSDMD are the “star factors” in the in terrestrial than in aquatic ecosystems (Rillig et al., 2021; Nizzetto et al.,
pyroptotic pathway. IHC results of cardiac tissues revealed that the expres- 2016). Currently, the information about toxic damage of MPs is generally
sion of NLRP3 and GSDMD were sequentially raised than Con (P < 0.05 to focused on aquatic ecosystems, and its prominent impact in agroforestry
P < 0.001) (Fig. 3A). To further validate the occurrence of pyroptosis, we ecosystems has been overlooked. Hence, it is extremely necessary to
examined the qRT-PCR and WB levels of markers of the NLRP3 study typical species in terrestrial ecosystems. 3–12 μm PS-MPs was usually
inflammasome-dependent pyroptotic pathway (Fig. 3B–C). Consistently, considered to be able to penetrate the blood barrier to reach the various
the expression levels of NLRP3, Caspase-1, IL-1β, IL-18, ASC and GSDMD organ tissues of the body (Wei et al., 2021; Shan et al., 2022; Dimitriadi
were increased in a concentration-dependent manner (Fig. 3B–C). It is et al., 2021). Herein, we applied chickens as the research object and focused
suggested that PS-MPs induced myocardial pyroptosis in a concentration- on the specific mechanism of myocardial toxic damage influenced by MPs
dependent manner. Subsequently, we detected the levels of markers in through in vitro and in vivo validation. We demonstrated for the first
the NF-κB inflammation pathway. As the shown in Fig. 5. The IHC level time that MPs induced myocardial pyroptosis in chickens via triggering
of NF-κB was elevated in the PS-MPs treated group, more pronounced in ROS overload, accompanied through amplified inflammation, imbalanced
H-MPS (P < 0.001) and M-MPS (P < 0.01), while P values < 0.05 of L- of mitochondrial homeostasis and abnormal energy metabolism. Specifi-
MPS rather than Con (Fig. 5A). Coherently, the nuclear accumulation of cally, the addition of NAC effectively declined the accumulation of ROS,
NF-κB displayed markedly higher in the PS-MPs groups (P < 0.001). which in turn alleviated myocardial pyroptosis and the consequent
Furthermore, the mRNA and protein expression of pro-inflammatory inflammatory response by driving the NF-κB-NLRP3-GSDMD axis, and im-
factors TNF-α, iNOS, COX-2, and IL-6 were also clearly upregulated proved mitochondrial dysfunction and energy metabolism abnormalities
and IκB-α was significantly downregulated than the Con (P < 0.05 to via regulating the AMPK-PGC-1α pathway.
P < 0.001) (Fig. 5B–C). Early reported point to MPs exposure led to the excessive accumulation
IF and qRT-PCR showed conspicuous shifts in the levels of all the above of ROS and oxidative stress in the organism (An et al., 2021). Our experi-
factors at H-PS compared to the Con, while there was no significant differ- mental results showed that PS-MPs stimulation strongly altered the activi-
ence at L-PS and M-PS (P > 0.05) (Figs. 4A–F and 5C). Therefore, we ties of GSH, CAT, MDA and T-AOC in myocardial tissue, suggesting the
assayed the protein expression of pyroptosis and inflammatory markers remarkable increased in oxidative stress. Besides, fluorescence staining of
under the effect of H-PS. In contrast to the Con, H-PS remarkably enhanced cardiac myocytes demonstrated that ROS accumulation was more pro-
the levels of NLRP3, pro-Caspase-1, Cleaved-Caspase-1, IL-1β, IL-18, ASC nounced in the PS-MPs treatment. External stimuli mediated the activation
and GSDMD, augmented the NF-κB (nuclear), TNF-α, iNOS, COX-2, and of NLRP3 by driving ROS, NLRP3 would be converted into a polymer com-
IL-6 and greatly decreased IκB-α (P < 0.05 to P < 0.001) (Fig. 4F). Meaning- plex through the junction protein ASC, also known as Caspase-1-dependent
fully, NAC rescued the onset of pyroptosis and inflammation in vitro NLRP3 inflammasome (Qiu et al., 2019). As mentioned before, activation of
(Figs. 4A-D, F and 5E). Overall, we concluded that PS-MPs triggered Caspase-1 cleaved GSDMD was a key “executioner” in the occurrence of
myocardial pyroptosis and amplify the inflammation in chickens via initiat- pyroptosis. Consequently, our experimental resulted also supported that
ing the NF-κB-NLRP3-GSDMD axis in vivo and in vitro. PS-MPs treatment altered the expression of pyroptosis markers to varying
degrees. More noteworthy, pyroptosis is an important factor in promoting
3.7. PS-MPS induces mitochondrial and energy metabolism dysfunction in vivo the inflammatory response (Dai et al., 2021). Stimulants also drove down-
and in vitro stream inflammatory factor expression through triggered ROS activation
of NF-κB typical inflammatory signaling molecules, promoting the secre-
To determine the specific molecular mechanisms by PS-MPs exposure tion of pro-inflammatory factors, such as TNF-α, iNOS, COX2, and IL-6
affects myocardial mitochondrial function and impaired energy metabo- (L. Zhao et al., 2021; Park et al., 2019). Similarly, our data indicated that
lism, we examined the mRNA and protein levels of AMPK-PGC-1α pathway PS-MPs induced NLRP3 inflammasome-mediated typical myocardial
and its downstream markers of mitochondrial fusion and division as well as pyroptotic pathway and amplified myocardial inflammatory damage by
energy metabolism markers in myocardium. As shown in Figs. 6A and 7A, indirectly activating the NF-κB inflammatory pathway. Concretely, the
PS-MPs treatment dramatically inhibited AMPK-PGC-1α pathway, induced PS-MPs treated overexpressed NLRP3, Caspase-1, IL-1β, IL-18, ASC
the TFAM, OPA1, MFN1 and MFN2 mRNA levels were down-regulated and and GSDMD, upregulated NF-κB, TNF-α, COX2, iNOS, IL-6 and decreased
DRP1, Fis1, HK2, PKM2, PDHX and LDH were up-regulated, while AMPK, IκB-α expression. It is worth mentioning that the NAC treatment reversed
PGC-1α, OPA1, DRP1, MFN1, MFN2, HK2, PKM2 and LDH protein the above situation to a great extent.
levels were approximately the same as mRNA (Figs. 6C and 7C). Actually, the AMPK-PGC-1α pathway is an essential sensor for the reg-
There were equally substantiated by IHC analysis of OPA1, DRP1 and ulation of energy metabolism in the body (Zmijewski et al., 2010), whereas
HK2 (Figs. 6B and 7B). numerous scientific studies have demonstrated that the overproduction of

8
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Fig. 5. PS-MPs stimulation leads to inflammatory response in vivo and in vitro. A: IHC observation and quantitative analysis of NF-κB in vivo. B and D: Analysis of mRNA
levels of NF-κB, IκB-α, TNF-α, COX-2, iNOS and IL-6. C and E: Analysis of protein levels of NF-κB, IκB-α, TNF-α, COX-2 and iNOS. Data are expressed as the mean ± SD
(n = 3). *P < 0.05, **P < 0.01 and ***P < 0.001.

ROS obviously modulates the alteration of the AMPK-PGC-1α pathway Nevertheless, these were urgent need for data to confirm whether the
(Li et al., 2021), which will affect mitochondrial function by stimulating ROS-AMPK-PGC-1α axis triggers alterations in both of these mechanisms
downstream mitochondrial fusion-related factors (Cai et al., 2021). in microplastic-induced myocardial injury in chickens. Our work showed

9
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Fig. 6. Effects of PS-MPs exposure on mitochondrial dynamics in vivo. A: Detection of mRNA expression of AMPK, PGC-1α, TFAM, OPA1, DRP1, MFN1, MFN2 and Fis1. B:
IHC observation and quantitative analysis of OPA1 and DRP1. C: Detection of protein expression of AMPK, PGC-1α, OPA1, DRP1, MFN1 and MFN2. Data are expressed as the
mean ± SD (n = 3). *P < 0.05, **P < 0.01 and ***P < 0.001.

that PS-MPs treatment visibly reduced the expression levels of AMPK and AMPK-PGC-1α axis. Explanatorily, in vivo experiments, we observed a
PGC-1α with ROS overaccumulation, triggering abnormal levels of mito- certain dose-dependence of the overall myocardial toxicity affected by PS-
chondrial fusion and fission factors TFAM, OPA1, DRP1, MFN1, MFN2 MPs exposure, but this trend was not verified in vitro experiments, which
and Fis1, and inducing the overexpression of energy metabolism kinases was probably influenced with the duration of PS-MPs action.
HK2, PKM2, PDHX and LDH. Equally, the addition of NAC could rescue In summary, we discovered for the first time the potential mechanism of
the above abnormal expression. Accordingly, this report provided prelimi- MPs induced cardiotoxicity in chickens. Firstly, PS-MPs triggered NF-κB-
nary data to support our view that PS-MPs induced mitochondrial and en- NLRP3-GSDMD axis overexpression by driving ROS overaccumulation,
ergy metabolism dysfunction in chickens myocardium through the launching myocardial pyroptosis and amplifying inflammatory responses in

10
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Fig. 7. Effects of PS-MPs exposure on energy metabolism in vivo. A: Detection of mRNA expression of HK2, PKM2, PDHX and LDH. B: IHC observation and quantitative
analysis of HK2. C: Detection of protein expression of HK2, PKM2, and LDH. Data are expressed as the mean ± SD (n = 3). *P < 0.05, **P < 0.01 and ***P < 0.001.

chickens. Secondly, PS-MPs overtly stimulated the dysfunction of mitochon- IκB-α Nuclear factor-κB alpha
dria and energy metabolism in chickens myocardium via regulating the ex- TNF-α Tumor necrosis factor-α
pression of AMPK-PGC-1α pathway through the overproduction of ROS. COX-2 Cyclooxygenase-2
Moreover, the NAC greatly reversed the above damage in chickens myocar- iNOS Inducible nitric oxide synthase
dium. Emphatically, ROS driven crosstalk of NF-κB-NLRP3-GSDMD and AMPK AMP-activated protein kinase
AMPK-PGC-1α pathways are critical in PS-MPs induced chicken heart injury. PGC-1α PPARγ coactivator-1α
This information fills a gap in the mechanism of MPs damage to myocardium TFAM Recombinant transcription factor A, mitochondrial
in chickens in terrestrial ecosystems and provides direction for protecting OPA1 Optic atrophy type 1
chickens from MPs poisoning. DRP1 Dynamin-related protein 1
MFN1 Mitofusin 1
MFN2 Mitofusin 2
Abbreviations Fis1 Mitochondrial fission 1
HK2 Hexokinase 2
MPs Microplastics PKM2 Pyruvate kinase isozyme type M2
PS-MPs Polystyrene microplastics PDHX Recombinant pyruvate dehydrogenase complex component X
GSH Glutathione LDH Lactate dehydrogenase
SOD Superoxide dismutase NAC N-Acetyl-L-cysteine
CAT Catalase
MDA Malondialdehyde Supplementary data to this article can be found online at https://doi.
T-AOC Total antioxidant capacity org/10.1016/j.scitotenv.2022.156727.
ROS Reactive oxygen species
NLRP3 Recombinant NLR family, pyrin domain containing protein 3 CRediT authorship contribution statement
Caspase-1 Cysteinyl aspartate specific proteinase-1
IL-1β Interleukin-1β Yue Zhang: Conceptualization, Methodology, Writing-Original draft,
IL-6 Interleukin-6 Writing-review. Kai Yin: Investigation, Formal analysis. Dongxu Wang:
IL-18 Interleukin-18 Investigation, Resources. Yu Wang: Software, Formal analysis. Hongmin
ASC Apoptosis-associated speck-like protein containing Lu: Resources, Data review. Hongjing Zhao: Supervision, Project adminis-
GSDMD Gasdermin D tration. Mingwei Xing: Supervision, Project administration, Funding
NF-κB Nuclear factor factor-κB acquisition.

11
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Fig. 8. Effects of PS-MPs exposure on mitochondrial dynamics and energy metabolism in vitro. A: Effect of PS-MPs with and without NAC on mitochondria of cardiomyocytes.
B: Mitochondrial mean optical density analysis. C and D: Relative mRNA expression level of AMPK, PGC-1α, TFAM, OPA1, DRP1, MFN1, MFN2, Fis1, HK2, PKM2, PDHX and
LDH. E: Relative protein expression levels of AMPK, PGC-1α, OPA1, DRP1, MFN1, MFN2, HK2, PKM2 and LDH. Data are expressed as the mean ± SD (n = 3). *P < 0.05,
**P < 0.01 and ***P < 0.001.

Declaration of competing interest References

The authors declare that they have no known competing financial inter- An, R., Wang, X., Yang, L., Zhang, J., Wang, N., Xu, F., Hou, Y., Zhang, H., Zhang, L., 2021. Poly-
ests or personal relationships that could have appeared to influence the styrene microplastics cause granulosa cells apoptosis and fibrosis in ovary through oxida-
work reported in this paper. tive stress in rats. Toxicology 449, 152665. https://doi.org/10.1016/j.tox.2020.152665.
Avio, C.G., Gorbi, S., Milan, M., Benedetti, M., Fattorini, D., d'Errico, G., Pauletto, M.,
Bargelloni, L., Regoli, F., 2015. Pollutants bioavailability and toxicological risk from
Acknowledgments microplastics to marine mussels. Environ. Pollut. 198, 211–222. https://doi.org/10.
1016/j.envpol.2014.12.021.
Banerjee, A., Shelver, W.L., 2021. Micro- and nanoplastic-mediated pathophysiological
This study acknowledges the support of the Natural Science Foundation changes in rodents, rabbits, and chickens: a review. J. Food Prot. 84, 1480–1495.
of Heilongjiang Province of China (ZD2020C005). https://doi.org/10.4315/jfp-21-117.

12
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Cai, J., Guan, H., Jiao, X., Yang, J., Chen, X., Zhang, H., Zheng, Y., Zhu, Y., Liu, Q., Park, M.H., Gutiérrez-García, A.K., Choudhury, M., 2019. Mono-(2-ethylhexyl) phthalate
Zhang, Z., 2021. NLRP3 inflammasome mediated pyroptosis is involved in cadmium aggravates inflammatory response via sirtuin regulation and inflammasome activation
exposure-induced neuroinflammation through the IL-1beta/IkB-alpha-NF-kappaB- in RAW 264.7 cells. Chem. Res. Toxicol. 32, 935–942. https://doi.org/10.1021/acs.
NLRP3 feedback loop in swine. Toxicology 453, 152720. https://doi.org/10.1016/ chemrestox.9b00101.
j.tox.2021.152720. Paulus, W.J., Tschöpe, C., 2013. A novel paradigm for heart failure with preserved ejection
Chen, X., Bi, M., Yang, J., Cai, J., Zhang, H., Zhu, Y., Zheng, Y., Liu, Q., Shi, G., Zhang, Z., fraction: comorbidities drive myocardial dysfunction and remodeling through coronary
2022. Cadmium exposure triggers oxidative stress, necroptosis, Th1/Th2 imbalance microvascular endothelial inflammation. J. Am. Coll. Cardiol. 62, 263–271. https://doi.
and promotes inflammation through the TNF-alpha/NF-kappaB pathway in swine small org/10.1016/j.jacc.2013.02.092.
intestine. J. Hazard. Mater. 421, 126704. https://doi.org/10.1016/j.jhazmat.2021. Pei, J., Wang, F., Pei, S., Bai, R., Cong, X., Nie, Y., Chen, X., 2020. Hydrogen sulfide promotes
126704. cardiomyocyte proliferation and heart regeneration via ROS scavenging. Oxidative Med.
Chi, Q., Hu, X., Liu, Z., Han, Y., Tao, D., Xu, S., Li, S., 2021. H2S exposure induces cell death in Cell. Longev. 2020, 1412696. https://doi.org/10.1155/2020/1412696.
the broiler thymus via the ROS-initiated JNK/MST1/FOXO1 pathway. Ecotoxicol. Envi- Pitt, J.A., Kozal, J.S., Jayasundara, N., Massarsky, A., Trevisan, R., Geitner, N., Wiesner, M.,
ron. Saf. 222, 112488. https://doi.org/10.1016/j.ecoenv.2021.112488. Levin, E.D., Di Giulio, R.T., 2018. Uptake, tissue distribution, and toxicity of polystyrene
Coppi, L., Ligorio, S., Mitro, N., Caruso, D., De Fabiani, E., Crestani, M., 2021. PGC1s and be- nanoparticles in developing zebrafish (Danio rerio). Aquat. Toxicol. 194, 185–194.
yond: disentangling the complex regulation of mitochondrial and cellular metabolism. https://doi.org/10.1016/j.aquatox.2017.11.017.
Int. J. Mol. Sci. 22. https://doi.org/10.3390/ijms22136913. Prüst, M., Meijer, J., Westerink, R.H.S., 2020. The plastic brain: neurotoxicity of micro-
Dai, X.-Y., Li, X.-W., Zhu, S.-Y., Li, M.-Z., Zhao, Y., Talukder, M., Li, Y.-H., Li, J.-L., 2021. Ly- and nanoplastics. Part. Fibre Toxicol. 17, 24. https://doi.org/10.1186/s12989-020-
copene ameliorates Di(2-ethylhexyl) phthalate-induced pyroptosis in spleen via suppres- 00358-y.
sion of classic Caspase-1/NLRP3 pathway. J. Agric. Food Chem. 69, 1291–1299. https:// Qiu, Z., He, Y., Ming, H., Lei, S., Leng, Y., Xia, Z.Y., 2019. Lipopolysaccharide (LPS) aggravates
doi.org/10.1021/acs.jafc.0c06534. high glucose- and Hypoxia/Reoxygenation-induced injury through activating ROS-
Dimitriadi, A., Papaefthimiou, C., Genizegkini, E., Sampsonidis, I., Kalogiannis, S., Feidantsis, dependent NLRP3 inflammasome-mediated pyroptosis in H9C2 cardiomyocytes.
K., Bobori, D.C., Kastrinaki, G., Koumoundouros, G., Lambropoulou, D.A., Kyzas, G.Z., J. Diabetes Res. 2019, 8151836. https://doi.org/10.1155/2019/8151836.
Bikiaris, D.N., 2021. Adverse effects polystyrene microplastics exert on zebrafish heart - Rillig, M.C., Ryo, M., Lehmann, A., 2021. Classifying human influences on terrestrial ecosys-
molecular to individual level. J. Hazard. Mater. 416, 125969. https://doi.org/10.1016/ tems. Glob. Chang. Biol. 27, 2273–2278. https://doi.org/10.1111/gcb.15577.
j.jhazmat.2021.125969. Schwabl, P., Köppel, S., Königshofer, P., Bucsics, T., Trauner, M., Reiberger, T., Liebmann, B.,
Gao, X., Zheng, Y., Peng, L., Ruan, X., Ji, H., Qiu, Y., Liu, X., Teng, P., Guo, D., Jiang, S., 2018. 2019. Detection of various microplastics in human stool: a prospective case series. Ann.
Maduramicin induces apoptosis in chicken myocardial cells via intrinsic and extrinsic Intern. Med. 171, 453–457. https://doi.org/10.7326/m19-0618.
pathways. Toxicol. in Vitro 50, 190–200. https://doi.org/10.1016/j.tiv.2018.03.008. Shan, S., Zhang, Y., Zhao, H., Zeng, T., Zhao, X., 2022. Polystyrene nanoplastics penetrate
Gong, Z.G., Zhao, Y., Wang, Z.Y., Fan, R.F., Liu, Z.P., Wang, L., 2022. Epigenetic regulator across the blood-brain barrier and induce activation of microglia in the brain of mice.
BRD4 is involved in cadmium-induced acute kidney injury via contributing to lysosomal Chemosphere 298, 134261. https://doi.org/10.1016/j.chemosphere.2022.134261.
dysfunction, autophagy blockade and oxidative stress. J. Hazard. Mater. 423, 127110. Shi, J., Gao, W., Shao, F., 2017. Pyroptosis: gasdermin-mediated programmed necrotic cell
https://doi.org/10.1016/j.jhazmat.2021.127110. death. Trends Biochem. Sci. 42, 245–254. https://doi.org/10.1016/j.tibs.2016.10.004.
Gu, X., Wang, Y., He, Y., Zhao, B., Zhang, Q., Li, S., 2022. MiR-1656 targets GPX4 to trigger Sun, L., Ma, W., Gao, W., Xing, Y., Chen, L., Xia, Z., Zhang, Z., Dai, Z., 2019. Propofol directly
pyroptosis in broilers kidney tissues by activating NLRP3 inflammasome under se defi- induces caspase-1-dependent macrophage pyroptosis through the NLRP3-ASC inflamma-
ciency. J. Nutr. Biochem. 105, 109001. https://doi.org/10.1016/j.jnutbio.2022.109001. some. Cell Death Dis. 10, 542. https://doi.org/10.1038/s41419-019-1761-4.
Horng, T., 2014. Calcium signaling and mitochondrial destabilization in the triggering of the Sun, T., Zhan, J., Li, F., Ji, C., Wu, H., 2021. Effect of microplastics on aquatic biota: a
NLRP3 inflammasome. Trends Immunol. 35, 253–261. https://doi.org/10.1016/j.it. hormetic perspective. Environ. Pollut. (Barking, Essex : 1987) 285, 117206. https://
2014.02.007. doi.org/10.1016/j.envpol.2021.117206.
Hou, B., Wang, F., Liu, T., Wang, Z., 2021. Reproductive toxicity of polystyrene microplastics: Sun, T., Zhan, J., Li, F., Ji, C., Wu, H., 2021. Environmentally relevant concentrations of
in vivo experimental study on testicular toxicity in mice. J. Hazard. Mater. 405, 124028. microplastics influence the locomotor activity of aquatic biota. J. Hazard. Mater. 414,
https://doi.org/10.1016/j.jhazmat.2020.124028. 125581. https://doi.org/10.1016/j.jhazmat.2021.125581.
Jing, H., Wang, F., Gao, X.J., 2022. Lithium intoxication induced pyroptosis via ROS/NF-κB/ Wang, Y.L., Lee, Y.H., Hsu, Y.H., Chiu, I.J., Huang, C.C., Huang, C.C., Chia, Z.C., Lee, C.P., Lin,
NLRP3 inflammasome regulatory networks in kidney of mice. Environ. Toxicol. https:// Y.F., Chiu, H.W., 2021. The kidney-related effects of polystyrene microplastics on human
doi.org/10.1002/tox.23446. kidney proximal tubular epithelial cells HK-2 and male C57BL/6 mice. Environ. Health
Kung, G., Konstantinidis, K., Kitsis, R.N., 2011. Programmed necrosis, not apoptosis, in the Perspect. 129, 57003. https://doi.org/10.1289/ehp7612.
heart. Circ. Res. 108, 1017–1036. https://doi.org/10.1161/circresaha.110.225730. Wang, L., Xue, N., Li, W., Wufuer, R., Zhang, D., 2021. Ecotoxicological effects of
Li, C., Busquets, R., Campos, L.C., 2020. Assessment of microplastics in freshwater systems: a microplastics on bird embryo development by hatching without eggshell. J. Visual.
review. Sci. Total Environ. 707, 135578. https://doi.org/10.1016/j.scitotenv.2019. Exp. https://doi.org/10.3791/61696.
135578. Wang, Y., Zhao, H., Liu, Y., Li, J., Nie, X., Huang, P., Xing, M., 2021. Environmentally relevant
Li, Z., Zhu, S., Liu, Q., Wei, J., Jin, Y., Wang, X., Zhang, L., 2020. Polystyrene microplastics concentration of sulfamethoxazole-induced oxidative stress-cascaded damages in the in-
cause cardiac fibrosis by activating Wnt/beta-catenin signaling pathway and promoting testine of grass carp and the therapeutic application of exogenous lycopene. Environ.
cardiomyocyte apoptosis in rats. Environ. Pollut. 265, 115025. https://doi.org/10. Pollut. (Barking, Essex: 1987) 274, 116597. https://doi.org/10.1016/j.envpol.2021.
1016/j.envpol.2020.115025. 116597.
Li, S., Ma, Y., Ye, S., Tang, S., Liang, N., Liang, Y., Xiao, F., 2021. Polystyrene microplastics Wei, Y., Zhou, Y., Long, C., Wu, H., Hong, Y., Fu, Y., Wang, J., Wu, Y., Shen, L., Wei, G., 2021.
trigger hepatocyte apoptosis and abnormal glycolytic flux via ROS-driven calcium over- Polystyrene microplastics disrupt the blood-testis barrier integrity through ROS-mediated
load. J. Hazard. Mater. 417, 126025. https://doi.org/10.1016/j.jhazmat.2021.126025. imbalance of mTORC1 and mTORC2. Environ. Pollut. (Barking, Essex: 1987) 289,
Lin, J., Kumari, S., Kim, C., Van, T.M., Wachsmuth, L., Polykratis, A., Pasparakis, M., 2016. 117904. https://doi.org/10.1016/j.envpol.2021.117904.
RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation. Nature 540, Wright, S.L., Kelly, F.J., 2017. Plastic and human health: a micro Issue? Environ. Sci. Technol.
124–128. https://doi.org/10.1038/nature20558. 51, 6634–6647. https://doi.org/10.1021/acs.est.7b00423.
Liu, Q., Du, P., Zhu, Y., Zhang, X., Cai, J., Zhang, Z., 2022. Thioredoxin reductase 3 suppres- Wright, S.L., Thompson, R.C., Galloway, T.S., 2013. The physical impacts of microplastics on
sion promotes colitis and carcinogenesis via activating pyroptosis and necrosis. Cell. Mol. marine organisms: a review. Environ. Pollut. 178, 483–492. https://doi.org/10.1016/j.
Life Sci. 79, 106. https://doi.org/10.1007/s00018-022-04155-y. envpol.2013.02.031.
Liu, X.J., Wang, Y.Q., Shang, S.Q., Xu, S., Guo, M., 2022. TMT induces apoptosis and Xu, S., Ma, J., Ji, R., Pan, K., Miao, A.J., 2020. Microplastics in aquatic environments: occur-
necroptosis in mouse kidneys through oxidative stress-induced activation of the NLRP3 rence, accumulation, and biological effects. Sci. Total Environ. 703, 134699. https://doi.
inflammasome. Ecotoxicol. Environ. Saf. 230, 113167. https://doi.org/10.1016/j. org/10.1016/j.scitotenv.2019.134699.
ecoenv.2022.113167. Xu, S., Xiaojing, L., Xinyue, S., Wei, C., Honggui, L., Shiwen, X., 2021. Pig lung fibrosis is
Liu, J.B., Li, Z.F., Lu, L., Wang, Z.Y., Wang, L., 2022. Glyphosate damages blood-testis barrier active in the subacute CdCl(2) exposure model and exerts cumulative toxicity through
via NOX1-triggered oxidative stress in rats: long-term exposure as a potential risk for the M1/M2 imbalance. Ecotoxicol. Environ. Saf. 225, 112757. https://doi.org/10.
male reproductive health. Environ. Int. 159, 107038. https://doi.org/10.1016/j.envint. 1016/j.ecoenv.2021.112757.
2021.107038. Yang, Q., Han, B., Xue, J., Lv, Y., Li, S., Liu, Y., Wu, P., Wang, X., Zhang, Z., 1987. Hexavalent
Lu, Y., Zhang, Y., Deng, Y., Jiang, W., Zhao, Y., Geng, J., Ding, L., Ren, H., 2016. Uptake and chromium induces mitochondrial dynamics disorder in rat liver by inhibiting AMPK/
accumulation of polystyrene microplastics in zebrafish (Danio rerio) and toxic effects in PGC-1α signaling pathway, environmental pollution (Barking. Essex 265 (2020),
liver. Environ. Sci. Technol. 50, 4054–4060. https://doi.org/10.1021/acs.est.6b00183. 114855. https://doi.org/10.1016/j.envpol.2020.114855.
Miao, Z., Miao, Z., Teng, X., Xu, S., 2022a. Chlorpyrifos triggers epithelioma papulosum Yang, J., Hamid, S., Cai, J., Liu, Q., Xu, S., Zhang, Z., 2017. Selenium deficiency-induced thi-
cyprini cell pyroptosis via miR-124-3p/CAPN1 axis. J. Hazard. Mater. 424, 127318. oredoxin suppression and thioredoxin knock down disbalanced insulin responsiveness in
https://doi.org/10.1016/j.jhazmat.2021.127318. chicken cardiomyocytes through PI3K/Akt pathway inhibition. Cell. Signal. 38, 192–200.
Miao, Z., Miao, Z., Wang, S., Wu, H., Xu, S., 2022b. Exposure to imidacloprid induce oxidative https://doi.org/10.1016/j.cellsig.2017.07.012.
stress, mitochondrial dysfunction, inflammation, apoptosis and mitophagy via NF- Yang, B., Chen, Y., Shi, J., 2019. Reactive oxygen species (ROS)-based nanomedicine. Chem.
kappaB/JNK pathway in grass carp hepatocytes. Fish Shellfish Immunol. 120, 674–685. Rev. 119, 4881–4985. https://doi.org/10.1021/acs.chemrev.8b00626.
https://doi.org/10.1016/j.fsi.2021.12.017. Yang, L., Zhang, Y., Kang, S., Wang, Z., Wu, C., 2021. Microplastics in soil: a review on
Miao, Z., Miao, Z., Shi, X., Wu, H., Yao, Y., Xu, S., 2022c. The antagonistic effect of selenium methods, occurrence, sources, and potential risk. Sci. Total Environ. 780, 146546.
on lead-induced apoptosis and necroptosis via P38/JNK/ERK pathway in chicken kidney. https://doi.org/10.1016/j.scitotenv.2021.146546.
Ecotoxicol. Environ. Saf. 231, 113176. https://doi.org/10.1016/j.ecoenv.2022.113176. Yin, K., Wang, D., Zhao, H., Wang, Y., Guo, M., Liu, Y., Li, B., Xing, M., 2021. Microplastics
Nizzetto, L., Futter, M., Langaas, S., 2016. Are agricultural soils dumps for microplastics of pollution and risk assessment in water bodies of two nature reserves in Jilin Province:
urban origin? Environ. Sci. Technol. 50, 10777–10779. https://doi.org/10.1021/acs. correlation analysis with the degree of human activity. Sci. Total Environ. 799,
est.6b04140. 149390. https://doi.org/10.1016/j.scitotenv.2021.149390.

13
Y. Zhang et al. Science of the Total Environment 840 (2022) 156727

Yin, K., Wang, Y., Zhao, H., Wang, D., Guo, M., Mu, M., Liu, Y., Nie, X., Li, B., Li, J., Xing, M., Zhao, L., Shi, W., Hu, F., Song, X., Cheng, Z., Zhou, J., 2021. Prolonged oral ingestion of
2021. A comparative review of microplastics and nanoplastics: toxicity hazards on diges- microplastics induced inflammation in the liver tissues of C57BL/6J mice through polar-
tive, reproductive and nervous system. Sci. Total Environ. 774. https://doi.org/10.1016/ ization of macrophages and increased infiltration of natural killer cells. Ecotoxicol. Envi-
j.scitotenv.2021.145758. ron. Saf. 227, 112882. https://doi.org/10.1016/j.ecoenv.2021.112882.
Zhao, H., Wang, Y., Liu, Y., Yin, K., Wang, D., Li, B., Yu, H., Xing, M., 2021. ROS-induced hep- Zmijewski, J.W., Banerjee, S., Bae, H., Friggeri, A., Lazarowski, E.R., Abraham, E., 2010.
atotoxicity under cypermethrin: involvement of the crosstalk between Nrf2/Keap1 and Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated
NF-kappaB/ikappaB-alpha pathways regulated by proteasome. Environ. Sci. Technol. protein Kinase*. J. Biol. Chem. 285, 33154–33164. https://doi.org/10.1074/jbc.
55, 6171–6183. https://doi.org/10.1021/acs.est.1c00515. M110.143685.

14

You might also like