You are on page 1of 10

Chemico-Biological Interactions 345 (2021) 109550

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

Research paper

Polystyrene microplastics induce apoptosis via ROS-mediated p53 signaling


pathway in zebrafish
Sathisaran Umamaheswari a, Sheela Priyadarshinee a, Krishna Kadirvelu b, Mathan Ramesh a, *
a
Unit of Toxicology, Department of Zoology, Bharathiar University, Coimbatore, 641 046, India
b
DRDO-BU Centre for Life Sciences, Bharathiar University Campus, Coimbatore, 641 046, India

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

Keywords: Microplastic (MP) pollution is ubiquitous and has become an emerging threat to aquatic biota. Recent scientific
Plastics reports have recorded their toxic impacts at the cellular and organism levels, but the underlying molecular
Enzymes mechanism of their toxicity remains unclear. The present study elucidates an array of molecular events under­
Inflammation
lying apoptosis in the gills of polystyrene microplastics (PS-MPs) exposed zebrafish (Danio rerio). PS-MPs at
Apoptosis
different concentrations (10 and 100 μg L− 1) induced the reactive oxygen species (ROS) generation, in turn
Histology
Genes affecting the oxidative and immune defense mechanism. The expression profile of antioxidant genes cat, sod1,
gpx1a and gstp1 were altered significantly. PS-MPs also significantly inhibited the neurotransmission in zebrafish.
In addition, the PS-MPs exposure upregulated the expression of p53, gadd45ba, and casp3b resulting in apoptosis.
We demonstrate that PS-MPs significantly upregulate the transcriptional pattern of tnfa and ptgs2a which are
essential gene markers in inflammatory mechanism. Further, the oxidative damage induced by PS-MPs exposure
could lead to cytological damage resulting in altered lamellar structures, capillary dilation, and necrosis in gill
histomaps. In conclusion, the findings of this work strongly suggest that PS-MPs induce dose-and time-dependent
ROS mediated apoptotic responses in zebrafish. Furthermore, the physiological responses observed in the gills
correlate with the above observations and helps in unravelling the potential molecular mechanism underpinning
the PS-MPs toxicity in zebrafish.

1. Introduction carriers for other environmental pollutants and influence their toxicity
[14,15].
The worldwide increase in plastics manufacture and disposal has The common plastics that are used in day-to-day life comprise high-
caused release of plastic debris in water bodies, thereby posing an density polyethylene (HDPE), low-density polyethylene (LDPE), poly­
ecological risk to the present and future generations [1,2]. The plastic propylene (PP), polystyrene (PS), polyamide (PA), polyester (PES),
trash entering seawaters was determined to be 4–12 million tons every polyvinylchloride (PVC), and polyethylene terephthalate (PET) [16].
year [3]. The majority of the plastic trashes that are circulated in the Among these plastics, PS stands third in the category of frequently
marine environment in the form of larger plastics as well as debris are identified plastic debris in marine environment [17]. Merchant research
non-biodegradable, and hence they greatly affect the aquatic ecosystem and consulting have reported that approximately 32.7 million tons of PS
[4–6]. Furthermore, in the environment plastics are subjected to were produced worldwide [18]. PS is an aromatic hydrocarbon pro­
degradation which results in the formation of microplastics [7,8]. Klein duced from polymerization of styrene monomers. Due to its versatility
et al. [9] reported that degradation of plastics in the environment may and inert nature, PS is employed in the manufacture of styrofoam and
be the key factor for the formation of microplastics. Microplastics (MPs) other plastic wares such as household appliances, toys, automobile
have been detected in oceans and rivers in the range of 1 μm–5 mm [7, parts, food packaging, medical devices, and consumer electronics [19,
10]. In addition, degradation of MPs via photo-oxidation, thermo-ox­ 20]. Besides its application as a general plastic, PS has been widely used
idative reactions and microorganisms [11] may lead to fragmentation of in bioanalytical and biomedical platforms [21,22]. Studies have also
smaller particles such as nanoplastics [12] which have been recognized reported the migration of styrene monomers into food items from PS
as a serious threat to aquatic organisms [13,14]. Plastics may also act as containers posing a threat to humanity [23–25]. Meanwhile, the PS

* Corresponding author. Unit of Toxicology, Department of Zoology, School of Life Sciences, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
E-mail address: mathanramesh@buc.edu.in (M. Ramesh).

https://doi.org/10.1016/j.cbi.2021.109550
Received 15 March 2021; Received in revised form 19 May 2021; Accepted 8 June 2021
Available online 11 June 2021
0009-2797/© 2021 Elsevier B.V. All rights reserved.
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

microbeads, pellets or fragments from cosmetics, detergents, and textiles purchased as aqueous suspensions from Sigma Aldrich. The morphology
also leach into the aquatic environment [26]. of the beads was characterized using a scanning electron microscope
Polystyrene microplastics (PS-MPs) are one of the most abundant (SEM – FEI QUANTA 200), where a drop of sonicated MPs suspension
MPs found in the marine environment [27,28]. These persisting PS-MPs was placed on the double-sided adhesive carbon tape fixed to an
in the aquatic environment produce a detrimental consequence on ma­ aluminum stub. The stub was air-dried in a sterile environment, coated
rine life, and human health. This is attributable to their conformation, with Au/Pd and examined at low vacuum mode (voltage- 15 kV;
toxicants adsorbing potency, biotransforming and biomagnifying ability working distance- 10 mm). The functional properties of PS-MP beads
in the food chain [29,30]. Unfortunately, the majority of MPs and NPs were analysed using Fourier Transform Infrared (FTIR) spectroscopy
consumed by humans are from food packed in polystyrene containers, (FTIR - 4100 type A). For measuring PS-MP bead size, 20 μL of the
seafood and water with undetectable PS-MPs/PS-NPs [31,32]. The aqueous suspension was taken and analysed using Zetasizer Nano,
major source of seafood includes fishes and shell fishes that were rich in Malvern, UK at a scattering angle 90◦ and wavelength 633 nm.
protein content. Approximately twenty percent of individual shellfish
and fish were loaded with microplastic litters in their gastrointestinal
tracts [33,34]. The ingestion and toxicity of PS-MPs in fishes have been 2.2. Chemicals
enumerated from various studies such as oxidative damage [35],
neurotoxicity [36], gene expression [37], alteration in glucose meta­ Alkaline phosphatase (AKP) and Lactate dehydrogenase (LDH) assay
bolism in zebrafish embryos [38], behavioral responses of zebrafish kits were purchased from Coral Clinical systems, Goa, India. High-
larvae [39], mortality of juvenile Daphnia magna [40], decrease of capacity cDNA Reverse Transcription Kits and SYBR Premix Ex Taq
growth rate and gross energy of fish [41] and intestinal immune cell TM II were purchased from Thermofisher Scientific and Takara respec­
dysfunction [42]. The existing studies on PS-MPs toxicity provide data at tively. The primers used for the study was purchased from Euroflims
the physiological level, it remains hypothetical if PS-MPs induced toxic Genomics. TRI Reagent was purchased from Sigma Aldrich. All the other
effects were ROS dependent and affects the molecular pathways pro­ chemicals were purchased from HiMedia Laboratories Pvt. Ltd., India.
ducing detrimental impacts.
Under normal physiological conditions, there is a balance between 2.3. Zebrafish and exposure
ROS generation and antioxidants activity. Exposure to MPs induces ROS
production causing oxidative stress and increased lipid peroxidation In the present study all the experiments were carried out as per the
[37]. As a compensatory response to oxidative stress, antioxidants are regulations of Organization of Economic Co-operation and Development
produced by living cells [8]. Previous studies suggested that the activity [55] and Committee for Control and Supervision of Experiments on
of antioxidants is influenced by the tempo of ROS induction and cyto­ Animals (CPCSEA). Adult male zebrafish (AB strain) were acquired from
protective genes transcription [15,43,44]. The induction of ROS gen­ the New golden aquarium, Coimbatore and maintained in aerated
eration and the activation of tnfa, a pro-inflammatory cytokine, are freshwater. The physicochemical parameters of water i.e., temperature
interdependent as the production of one trigger the activation of other. 26.4 ± 1.2 ◦ C, pH 6.6 ± 0.53, total hardness 159 ± 2.7 mg L− 1 CaCO3,
The ROS acts as a key initiator in various signaling pathways alkalinity 194 ± 2.04 mg L− 1, and dissolved oxygen 6.8 ± 0.3 mg L− 1
involving cell cycle and energy metabolism [45]. Qiang and Cheng [46] were measured daily as per APHA guidelines [56]. During acclimatiza­
have reported on the ROS mediated p53 apoptotic cascade activation on tion, the fishes were fed with commercial pellets and the water was
exposure to microplastics. The p53 signaling pathway regulates various renewed once a day.
cellular responses associated with cell cycle, senescence, cell survival Two different concentrations of PS-MPs namely 10 μg L− 1 and 100
and apoptosis [47,48]. The p53 gene transduce signals to stimulate μg L− 1 were selected for testing the chronic toxicity [57]. Healthy fish
apoptosis via cas3b and gadd45ba activation [49,50] along with the with 0.28 ± 0.07 g body weight and 2.43 ± 0.06 cm body length (35
expression of ptgs2a, an inflammatory biomarker in zebrafish. The gene Nos) were selected from the stock and introduced in each glass aquaria
gadd45ba has been associated with stress response to any chemical or filled with 35 L of water. Fish were exposed to two concentrations of
environmental stressors, causing growth arrest, DNA damage or PS-MPs (10 μg L− 1 as Treatment I and 100 μg L− 1 as Treatment II) along
apoptosis [51]. A limited number of toxicity studies indicated that with the control in three replicates for each concentration. The glass
oxidative and subsequent DNA damage was linked to the physiological aquaria were replenished with test solution (freshwater for control)
responses induced by PS-MPs [5,52]. every 24 h to sustain the PS-MPs concentration. Every day the physi­
However, the underlying mechanism of PS-MPs induced toxicity are cochemical properties of water were measured for the renewed aquar­
not yet clearly understood. The present study was conducted to further ium water. Experiments were conducted for a period of 35 days with 7
discern these mechanisms, induced oxidative responses and associated days sampling frequency. After exposure, 4 fishes from control and
apoptotic pathway in zebrafish. Zebrafish (Danio rerio) has been widely treated groups were sampled, cleansed with methanol to get rid of
used as a vertebrate model for studying toxicity of any chemical in the particles from the skin, weighed (0.28 ± 0.09 g body weight and 2.55 ±
aquatic environment [53]. Besides a vertebrate model, it is a successful 0.08 cm body length), anesthetized and gill tissues were harvested and
model in MPs toxicity studies for its transparent nature to localise used for biochemical assays. Similarly, gills of 4 fishes from each aquaria
fluorescent MPs/NPs [54]. We hypothesize that ROS mediates the p53 were sampled separately at the end of 7 and 35 d and used for histo­
apoptotic pathway via casp3b activation and subsequently modulates logical analysis (n = 1) and expression analysis (n = 3).
the physiology in PS-MPs exposed zebrafish. In the present study, we
have evaluated the potential of PS-MPs to induce oxidative stress,
biochemical, and histological responses in zebrafish (Danio rerio). In 2.4. Preparation of sample homogenate
addition, we explored the effects of PS-MPs on transcriptional pattern of
genes involved in apoptotic pathway and inflammatory responses of the The pooled tissue samples were homogenized with ice-cold phos­
gills in zebrafish. phate buffer saline (50 mM, pH 7.4). A part of this homogenate was used
for lipid peroxidation (LPO), catalase (CAT), superoxide dismutase
2. Materials and methods (SOD), and glutathione peroxidase (GPx) assays. The leftover homoge­
nate was centrifuged at 12000 g for 30 min at 4 ◦ C and the supernatant
2.1. Polystyrene MPs was used to measure reactive oxygen species (ROS), glutathione S-
transferase (GST), acetylcholinesterase (AChE), and biochemical en­
PS-MP beads (Product no: LB1) of 0.10–0.12 μm in size were zymes (AKP, and LDH).

2
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

2.5. Oxidative stress indices 2.8. Acetylcholinesterase (AChE) activity

2.5.1. Reactive oxygen species (ROS) AChE level was measured using the method adapted by Ellman et al.
The ROS level was measured by the modified dichlorofluorescein- [63] with slight modifications. Shortly, 250 μL of the reaction solution
diacetate (DCF-DA) method [58]. To 20 mL of the supernatant, 200 (30 mL of 0.1 M phosphate buffer, 1 mL of 10 mM 5, 5-dithiobis-2-nitro­
mL of PBS and 8.3 mL of DCF-DA was added and incubated in the dark benzoic acid solution with sodium bicarbonate, and 0.2 mL of 0.075 M
for 30 min at 37 ◦ C. The fluorescence intensity was measured with 485 acetylcholine solution) were added to 50 μL of the supernatant and the
nm excitation and 520 nm emission in a fluorescence spectrometer optical density was measured at 414 nm in a microplate photometer.
(HORIVA) and expressed as relative fluorescence intensity.
2.9. Real-time reverse transcription-polymerase chain reaction (RT-PCR)
2.5.2. Lipid peroxidation (LPO)
The LPO level was estimated following thiobarbituric acid reactive To study the expression pattern, the total RNA was extracted from
substances (TBARS) assay [59]. Shortly, 100 mL of 5% trichloroacetic the pooled gill tissue samples using TRI Reagent following the manu­
acid (TCA) was added to the tissue homogenate and incubated at 4 ◦ C for facturer’s protocol. The purity and quantity of the obtained RNA were
10 min. Then 100 mL of 0.67% thiobarbituric acid was added to the determined using a NanoDrop plate reader (NanoDrop Technologies,
mixture and centrifuged at 2200 g for 10 min. The clear upper phase was Synergy). Total RNA (1 μg/μL) of each sample was reverse transcribed
incubated in a hot water bath for 10 min, cooled and the optical density using High-capacity cDNA Reverse Transcription Kit. Quantitative real-
was measured at 535 nm in a microplate photometer (Synergy H1, time PCR was performed with synthesized cDNAs, SYBR Premix Ex Taq
BioTek). TM II, and gene-specific primers in Light Cycler 480 (Roche). The re­
action involves denaturation (at 95 ◦ C for 10 min), amplification (40
2.6. Antioxidant enzymes cycles at 95 ◦ C for 15 s and at 60 ◦ C for 1 min), followed by quantifi­
cation, with a melting curve program (60–99 ◦ C). The sources and se­
2.6.1. Catalase (CAT) activity quences of used primers are given in Table 1. All PCR reactions were
The CAT activity was measured following the procedure of Sinha performed in triplicates and the β-actin was employed to normalize the
[60] with slight modifications. Briefly, 25 μL of ice-cold Tris-HCl buffer expression curve of the test gene. Relative fold changes of genes were
(100 mM, pH 7.4) was added to the tissue homogenate and cold determined using the 2− ΔΔCT method [64].
centrifuged at 12000 g for 15 min. The clear upper phase was collected,
3 mL of the reaction mixture (acetic acid and 5% potassium dichromate
2.10. Histological analysis
in 3:1 ratio and 10 mM phosphate buffer, pH 7.0) was added and the
mixture was incubated in a hot water bath for 15 min. The optical
Histological analysis of zebrafish gills was carried out using the
density of the solution was measured at 570 nm in a microplate
protocol of Teng et al. [65]. The gill tissues were fixed in 10% formalin
photometer.
for 48 h and subjected to dehydration in an increasing string of ethanol.
Then the tissues were xylene cleared, embedded in blocks of paraffin
2.6.2. Superoxide dismutase (SOD) activity
wax, and sectioned at 3–5 μm thickness. The sectioned tissues were
The activity of SOD was determined by following the method of
hematoxylin and eosin (H&E) stained for microscopic analysis. The
Marklund and Marklund [61] with slight modifications. 25 μL of 100
slides were observed and photographed using a light microscope (DME
mM Tris-HCl buffer (pH 7.4) was added to the tissue homogenate and
light Microscope, Leica).
cold centrifuged at 12000 g for 15 min. The clear upper phase collected
was added to the reaction solution (50 mM Tris-HCl buffer, with 1 mM
EDTA and 2.64 mM Pyrogallol) and the optical density was read at 420 2.11. Statistical analysis
nm in a microplate photometer.
Graph Pad Prism software (version 8) was used for the statistical
2.6.3. Glutathione peroxidase (GPx) activity analysis. The significant differences in all the experimental groups were
Glutathione peroxidase activity was determined by adapting the
method of Adeyemi et al. [59] with slight modifications. Around 25 μL Table 1
of Tris-HCl buffer (100 mM, pH 7.4) was added to the tissue homogenate Primers and the nucleotide sequences.
and cold centrifuged at 12000 g for 15 min. The clear upper phase was S.No Name Primer sequences Accession number
obtained after centrifugation. Then, 880 μL of the reaction mixture (1
1 β actin GCCAACAGAGAGAAGATGAC AF057040.1
mM GSH, 150 mM of NADPH, 100 mM sodium azide in potassium CACCAGAGTCCATCACAATAC
phosphate buffer, pH 7.0) was added, and the optical density was 2 cat GGAGCTCAACTCTTCATCCA AF170069.1
measured photometrically at 340 nm for 1 min. CCCTTCAGCGTTGTGTTTATC
3 sod1 AGTGAAGGTGACTGGTGAAA BC055516.1
ATGCAGCCGTTTGTGTTG
2.6.4. Glutathione S-transferase (GST) activity 4 gstp1 GGCAACGGCAAACAGTAAA NM_131734.3
GST level was measured using the method adapted by Domingues CATTGAGCACCAGTTAAGGC
et al. [62] with slight modifications. In brief, 100 μL of the reaction 5 gpx1a GGCTTCTACGTGTTGTGTTC NM_001007281.2
solution (10 mM GSH and 60 mM 1-chloro2, 4-dinitrobenzene) was ACTGCACAGTCGGTCTATATC
6 ache CACAACAGGGAGATCACAGTA BC076071.1
added to 50 μL of the clear supernatant and the optical density was
GAAGAGGAACGTGAGTAGCA
measured at 340 nm in a microplate photometer for 5 min. 7 p53 CTTGGTGCTGAATGGACAAC NM_001328588.1
CCAGAGTGATGATTGTGAGGA
2.7. Biochemical analysis 8 casp3b GTGTAGGTGACGAGGAAACA NM_001048066.2
AGGAGCCATTAGCGACATT
9 gadd45ba CAATGCCAGCCTCTCAAATG NM_213031.3
The activity of AKP and LDH enzymes was measured using Reagent CGTTCTTGCAGGGACAGATA
Kits following the manufacturer’s protocol. 10 tnfa GACCTTAGACTGGAGAGATGAC NM_212859.2
ACACCTGGCTGTAGACAAA
11 ptgs2a TGAGGAGATGACAGGAGACA NM_153657.1
CACAAGAAGGCCAGGATACA

3
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

determined using two-way ANOVA followed by Duncan’s multiple 3.4. Biochemical enzymes
comparisons test. The significance level was set at P < 0.05.
Throughout the study period, AKP activity shows a significant (P <
3. Results 0.05) decline in PS-MPs groups (10 μg L− 1 and 100 μg L− 1) compared to
the control as shown in Fig. 2g. The decline was more pronounced with
3.1. PS-MPs – characterization an increase in dosage and duration of PS-MPs exposure. Compared to the
control, PS-MPs (10 μg L− 1 and 100 μg L− 1) have significantly induced
In the present study, the SEM image of PS-MP beads were spherical (P < 0.001) the LDH activity time-and dose-dependently in this study
with a size range of 103–113 nm (0.103–0.113 μm) (Fig. 1a). The FTIR (Fig. 2h).
spectrum of the PS-MPs beads show the presence of polystyrene group
(Fig. 1b), verifying the manufacturers’ data. From Fig. S1, the size dis­ 3.5. Neurotransmitter – acetylcholinesterase (AChE) activity
tribution graph of PS-MPs shows the average size distribution of PS-MPs
is 116.5 nm (0.116 μm) in the sample. Compared to the control, PS-MPs (10 μg L− 1 and 100 μg L− 1) have
significantly (P < 0.001) inhibited the AChE activity in this study
3.2. Oxidative stress indices (Fig. 2i). The observed inhibition in the AChE activity showed a time-
and dose-dependent gradual increase.
Compared to the control, PS-MPs (10 μg L− 1 and 100 μg L− 1) have
significantly induced (P < 0.001) the ROS production time-and dose-
dependently in this study (Fig. 2a). Similarly, the depicted results from 3.6. Gene regulation pattern
Fig. 2b shows the significant (P < 0.001) induction of LPO levels in a
time-and dose-dependent manner in PS-MPs groups (10 μg L− 1 and 100 PS-MPs have significantly altered the transcriptional profile of anti­
μg L− 1) compared to the control. oxidants, acetylcholinesterase and apoptosis-related genes (Fig. 3). The
expression pattern of cat, sod1, gpx1a (Fig. 3a–c), and ache (Fig. 3e) tend
3.3. Antioxidant biomarkers to decrease significantly (P < 0.05) with increasing dose and duration of
PS-MPs exposure. However, the observed cat decrease was not statisti­
Compared to the control, PS-MPs (10 μg L− 1 and 100 μg L− 1) have cally significant (P > 0.05) in the 7th day of lower dose group (10 μg
significantly (P < 0.05) inhibited the CAT activity in this study (Fig. 2c) L− 1). Similarly, the decreased expression of sod1 and ache was also noted
with an exception in the 7th and 14th day in lower dose group (10 μg to be insignificant (P > 0.05) in 7th and 35th day of lower dose group
L− 1). Similarly, the SOD activity was significantly (P < 0.05) inhibited (10 μg L− 1). The gene gstp1 was significantly (P < 0.05) upregulated in
in the PS-MPs groups (10 μg L− 1 and 100 μg L− 1) compared to the the higher dose group (100 μg L− 1) whereas it showed no significant
control (Fig. 2d). However, the SOD activity in the lower dose group (10 change (P > 0.05) in the lower dose group (10 μg L− 1) relative to the
μg L− 1) showed no significant (P > 0.05) variation at the end of the 7th control (Fig. 3d). On the other hand, the genes tnfa, p53, casp3b,
day. From Fig. 2e, GPx activity in the PS-MPs groups (10 μg L− 1 and 100 gadd45ba, and ptgs2a (Fig. 3f–j) were significantly (P < 0.05) upregu­
μg L− 1) show significant (P < 0.05) decrease in relation to control lated in the PS-MPs group when compared to control group (except at
group. However, on 7th day in the lower and higher dose group (10 μg the end of 7th day in Treatment 1).
L− 1 and 100 μg L− 1) and 14th day in higher dose group (100 μg L− 1), no
significant (P > 0.05) change in GPx activity was observed. The 3.7. Histological analysis
observed decrease in antioxidants is inversely proportional to the dosage
and duration of PS-MPs exposure. The result Fig. 2f, depicts the signif­ To study the PS-MPs induced histological responses, we analysed the
icant (P < 0.05) increase in GST activity which depends on time and gill histomaps of zebrafish after 7and 35 d of PS-MPs exposure. As
dose of PS-MPs exposure with an exception in the 7th day in lower dose observed in Fig. 4a and b, gill from the control exhibit normal histo­
group (10 μg L− 1) compared to the control group. morphology, such as regular lamellar structure lined with squamous
epithelial cells, conventional orchestrate of pillar cells, and normal fil­
amental structures. However, after 7 d of PS-MPs exposure, the gills
from the lower dose group (10 μg L− 1) possessed aneurysms, capillary
dilation and necrotic cells (Fig. 4c). The occurrence of histological

Fig. 1. SEM image (a) and FT-IR spectrum (b) of PS-MPs (0.1 μm) used in this study.

4
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

Fig. 2. Effects of PS-MPs exposure on reactive oxygen species (a); lipid peroxidation (b); catalase (c); superoxide dismutase (d); glutathione peroxidise (e); gluta­
thione s-transferase (f); alkaline phosphatase (g); lactate dehydrogenase (h); acetylcholinesterase (i) level in the gills of Danio rerio over 35 d exposure. Values are
expressed as ± SEM. P < 0.05 - statistically significant (*P < 0.05, **P < 0.01, and ***P < 0.001) and P > 0.05 - not significant (#).

lesions increased with increase in time causing lamellar fusion after 35 serious environmental concern throughout the world [1,45,66]. Inges­
d of PS-MPs exposure in the lower dose group (Fig. 4d). Similarly, fish tion of MPs by aquatic organisms may accumulate and cause adverse
exposed to higher dose (100 μg L− 1) exhibited abnormal gill histo­ effects such as oxidative stress, inhibition of growth and development,
morphology such as disrupted lamellar structures, aneurysms, epithelial neurotransmission malfunction, endocrine disruption, immunological
lifting of cells and also exhibited increased necrotic zones after 7d and disorders etc., [26,35,67]. However, data on the MPs-induced physio­
35d of PS-MPs exposure in a dosage and duration dependent manner logical responses via apoptosis signaling pathway in freshwater fish is
(Fig. 4e and f). very limited. Therefore, the present study illustrates the mechanistic
pathway of ROS induced toxicity on the modulation of transcriptional
4. Discussion pattern of cytoprotective and apoptosis-related genes in PS-MPs exposed
zebrafish.
The presence of plastics in the marine and freshwater ecosystem is a In general, three different processes in the cell generate ROS when

5
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

Fig. 3. Effects of PS-MPs exposure on the transcription of cat (a); sod1 (b); gpx1a (c); gstp1 (d); ache (e); tnfa (f); p53 (g); casp3b (h); gadd45ba (i); ptgs2a (j) genes in
the gills of Danio rerio over 35 d exposure. Values are expressed as ± SEM. P < 0.05 - statistically significant (*P < 0.05, **P < 0.01, and ***P < 0.001) and P > 0.05
- not significant (#).

exposed to any stress/or under normal conditions. The former one is the related to ROS production has been reported [51]. The increased LPO
production of ROS from mitochondrial respiratory chain whereas the levels in our study might attribute to the formation of DNA adducts
second involves the NADPH oxidases (NOX)-mediated ROS release. The (MDA (Malondialdehyde) and HNE (4-Hydroxynonenal)) inducing
latter one is the production of ROS from several enzymatic (cytochrome epigenetic modifications and cell death [74].
P450, cyclooxygenases, heme oxygenase, lipoxygenases, myeloperox­ Exposure to PS-MPs has influenced the antioxidant defense mecha­
idases, monoamine oxidases, and xanthine oxidizes) reactions [68]. nism in zebrafish. The antioxidant defense system is an array of anti­
Besides the production of ROS from aforementioned sources, cytokines oxidants such as SOD, CAT, GPx, GSH, and GST involved in the
and growth promoters hitch to different classes of receptors leading to neutralization of free radicals [75]. In our study, the activities of CAT
the generation of ROS that acts as second messengers in various and SOD showed depletion in the PS-MPs group compared to the con­
signaling pathways [42,69]. trol. Akin to the enzyme activity, the cat and sod1 gene likely to
Among different classes of receptors, TNFRSF (tumor necrosis factor downregulate in the PS-MPs group in a time- and dose-independent
super family) is a super family of cytokine receptors that bind to tumor manner. Similar results were observed in Eriocheir sinensis and Macro­
necrosis factor (TNF) regulating various cellular processes such as cell brachium nipponense [15,47]. In addition, GPx activity was significantly
survival, growth, multiplication, differentiation, ageing, and death [70, reduced in PS-MPs treated groups which corroborates with the
71]. The fish exposed to PS-MPs exhibited increased tnfa expression and decreased mRNA levels of gpx1a observed in PS-MPs exposed zebrafish.
ROS levels indicating the occurrence of tnfa induced ROS generation. Gu Meanwhile GST, the phase II antioxidant enzyme showed an elevated
et al. [72] and Qiang and Cheng [46] recently reported the elevated ROS response in PS-MPs groups indicating the increased catalytic process of
levels in the larvae and gonads of PS-MPs treated Danio rerio. However, GST in phase II detoxification process. The gstp1 gene also exhibited a
very few studies have reported on the tnfa mediated ROS activity in time-and dose-dependent upregulation in PS-MPs groups substantiating
PS-MPs induced toxicity. the observed enzyme response. Similar results were observed in PS-NPs
The increased ROS levels and the upregulation of tnfa gene in PS-MPs exposed Daphnia pulex and PS-MPs exposed Poecilia reticulate [76,77].
groups in our study manifest the correlation between the ROS and tnfa Up-regulation of the expression of antioxidant has also been docu­
levels. This corroborates with the findings of Choi et al. [5] on MPs mented in response to the ROS induced by microplastics [50,54].
treated Cyprinodon variegatus. The generated ROS oxidizes the lipids, Oxidative stress is closely associated with the inflammation process
proteins, and DNA affecting the metabolic pathways [73]. Li et al. [15] and the stimulation of any of this process might induce the other [78].
recorded an interdependent response of ROS and LPO levels in the Several inflammatory responses like disrupted lamellar structures,
PS-NPs exposed Macrobrachium nipponense, lending support to our study. epithelial lifting, lamellar fusion, capillary dilation, and necrosis were
In this line, Huang et al. [66] recorded an increase of ROS and LPO level observed in the gill histomaps of PS-MPs exposed fishes. The
in micro-PS exposed marine mussels. Dimitriadi et al. [26] observed a up-regulated transcription pattern of inflammation marker genes tnfa,
significant increase of lipid peroxidation in heart of zebrafish upon and ptgs2a, suggest the stimulation of immune defense mechanism
exposure to PS-MPs. Similarly, changes in the expression of genes against PS-MPs induced oxidative stress, producing inflammatory

6
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

Fig. 4. Histomaps of gill tissue. a) Normal histology of gill tissue - control group (7th day). b) Normal histology of gill tissue - control group (35th day). c) PS-MPs
group - 10 μg L− 1 (7th day). d) PS-MPs group - 10 μg L− 1 (35th day). e) PS-MPs group - 100 μg L− 1 (7th day). f) PS-MPs group - 100 μg L− 1 (35th day). CD -
cytoplasmic degeneration, AM - aneurysm, NC - necrotic cells, LF - Lamellar fusion, EL - epithelial lifting; Scale bar - 400 μm. Insets (scale bar – 100 μm).

responses in the fish. Similarly, study of Wang et al. [79] and Karbalaei release of proinflammatory cytokine [83]. In our study, we observed an
et al. [80] depicted the PS-MPs induced inflammations in the gill his­ inhibited AKP activity with an elevated tnfa expression pattern in
toarchitectures of Oryzias melastigma and Oncorhynchus mykiss. The po­ PS-MPs groups providing additional evidence for the PS-MPs induced
tential toxicity of PS-MPs depends on particle size, species, time, and inflammatory responses in Danio rerio. Similarly, Zhang et al. [84] re­
exposure systems [64]. Among these factors, the particle size determines ported the interdependent response of tnfa and AKP activity in delta­
the toxicity of PS-MPs in an organism. The smaller the size of particle, methrin treated Gobiocypris rarus.
the greater surface area enabling easy adhesion of PS-MPs particles to Besides the observed inflammatory responses, the occurrence of
cells (blood cells, hepatic cells/epithelial cells) inducing cellular rup­ increased expression of growth arrest and DNA damage inducible gene
tures [37]. This leads to cell death thereby producing damaged histo­ beta a, gadd45ba in PS-MPs treated groups reveals the genotoxic po­
architectures in tissues. tential of PS-MPs. The gene gadd45ba is associated with the regulation of
In the present study, the decreased AKP activity in PS-MPs groups several cell cycle events such as cell survival, DNA damage repair, cell
indicates the inhibited release of the enzyme from lysosomes of cells cycle arrest, and apoptosis [85] and the increased expression of this gene
[81]. In general, AKP dephosphorylates the toxic moiety of proin­ indicate the activation of apoptotic cascade in PS-MPs groups. Apoptosis
flammatory molecules that are being produced by the cells during stress plays a crucial role in growth, regulation of tissue homeostasis, and in­
conditions. The diminished AKP activity inhibits the dephosphorylation flammatory responses in all organisms [86]. The observed gadd45ba and
process causing the deposition of proinflammatory molecules in the p53 genes upregulation might have caused apoptosis, which is evident
blood stream [82]. Further, this stimulates the immune response and the from the observed tissue damage in the gill histomaps of PS-MPs exposed

7
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

zebrafish. Similarly, Cheng et al. [87] observed significant loss in DNA fish has not been well characterized. Present findings shows that PS-MP
integrity and upregulated transcriptional pattern of p53 in cadmium induces oxidative stress in cells by generating surplus ROS and
exposed Scylla paramamosain. The gene p53 triggers the decreasing antioxidants production. Subsequently, disparity between
caspase-signaling pathway through apoptosis-related molecules such as ROS generation and antioxidant defenses, causes oxidative damage in
Bcl-2/Bax, noxa or cytochrome c activation [53]. In our study, the zebrafish, and alter apoptosis-related genes expression resulting in
transcriptional pattern of casp3b in PS-MPs group shows increased ac­ dysregulated physiology. Fig. 5 summarizes the molecular events trig­
tivity revealing the occurrence of apoptosis. The results of the present gered by PS-MPs in zebrafish. We hypothesize that PS-MPs induce ROS
study are in good agreement with Karami et al. [88] and Choi et al. [5] that affects the oxidative defense system, and neurotransmission in
showing MPs provoked DNA damage and apoptosis in Danio rerio and zebrafish. Furthermore, ROS induction has stimulated p53 apoptotic
Cyprinodon variegates. cascade activation producing DNA damage and inflammatory responses
LDH, a cytosolic enzyme converts pyruvic acid to lactic acid in the that were evident from the gill histomaps of zebrafish.
anaerobic energy production process. An altered LDH response is the
indication of the xenobiotic intervention in the energy metabolism 5. Conclusion
causing cell death/injury [89]. The increased LDH levels in PS-MPs
groups indicate impaired energy production resulting in lethargic Taken together, it is evident that p53 apoptotic pathway induction
swimming activity. Decrease in swimming speed has also been reported and enzymes dysregulation are responsible for the various physiological
in zebrafish exposed to PS microspheres [53] and in goldfish Carassius responses in PS-MPs treated zebrafish. In addition, several genes
auratus upon exposure to MPs [90]. Penetration of MPs across the involved in cytoprotection and inflammatory responses were also found
blood–brain barrier in fish may be the possible reason for the behavioral to be dysregulated. The current study provides us with insights into the
changes [35]. The lethargic swimming pattern might also be resulted toxic mechanism of PS-MPs to induce apoptosis in zebrafish and further
from the inhibition of AChE activity in PS-MPs treated zebrafish. This research on PS-MPs intervention in metabolic pathways could be
inhibition causes dysfunction of neurotransmitters resulting in acetyl­ studied.
choline (ACh) build up at synapses causing a jam in the cholinergic
neurotransmission [91]. CRediT authorship contribution statement
Many researchers have documented the apoptotic effects of MPs in
fish [11,53,59]. However, the molecular mechanism involved in the Sathisaran Umamaheswari: Conceptual framework, Resources,
induction and execution of apoptosis in the tissues of PS-MPs exposed Experiments, Investigation, Data curation, Original manuscript writing.

Fig. 5. Mechanism of PS-MPs induced apoptosis in zebrafish: In general, PS-MPs or the unbound styrene (which forms SO (styrene oxide) later) enter the cell and
are detoxified by the action of cytp450, GSH and GST enzymes. This mitochondrial detoxification results in ROS generation, which stimulates LPO with the release of
MDA and HNE. This leads to the formation of DNA adducts causing cell death. Furthermore, the produced ROS upregulates the transcription of p53 gene which in
turn switches on the casp3b activation. The activated casp3b promotes the transcription of gadd45ba resulting in DNA damage and apoptosis. This results in tissue
damage causing increased expression of ptgs2a and tnfa with the release of LDH enzymes. On the other hand, the produced ROS downregulates the expression of
cytoprotective genes such as cat, sod1, gpx1a and upregulates the expression of detoxification gene gstp1, thereby influencing the translation of antioxidants.

8
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

Sheela Priyadarshinee: Experiments, Investigation, Formal analysis. the Scientific Aspects of Marine Environmental Protection, 2016. London, https://e
c.europa.eu/environment/marine/good-environmental-status/descriptor-10/pdf/
Krishna Kadirvelu: Resources, Visualization, Investigation. Mathan
GESAMP_microplastics%20full%20study.pdf. (Accessed February 2021).
Ramesh: Conceptual framework, Supervision, Resources, Writing – re­ [18] T.A. Farrelly, I.C. Shaw, Polystyrene as Hazardous Household Waste, Household
view & editing. hazardous waste management, 2017.
[19] J.O. Choi, F. Jitsunari, F. Asakawa, D. Sun Lee, Migration of styrene monomer,
dimers and trimers from polystyrene to food simulants, Food Addit. Contam. 22
Declaration of competing interest (2005) 693–699.
[20] S.T. Rabie, A.M. Mahran, E.M. Kamel, N.H. Abdel Hamid, Photodegradation of
polystyrene stabilized with uracil derivative, J. Appl. Sci. Res. 4 (2008)
The authors declare that they have no known competing financial
2015–2026.
interests or personal relationships that could have appeared to influence [21] A.E. Hagan, C.A. Farrington, G.C. Wall, M.M. Belz, Sodium polystyrene sulfonate
the work reported in this paper. for the treatment of acute hyperkalemia: a retrospective study, Clin. Nephrol. 85
(2016) 38–43.
[22] B. Díez-Buitrago, F.J. Fernández-SanArgimiro, J. Lorenzo, N. Briz, V. Pavlov,
Acknowledgment Modification of chlorosulfonated polystyrene substrates for bioanalytical
applications, Mater. Sci. Eng. C 112 (2020), 110912.
We thank the Director, DRDO-BU Center for Life Sciences, Bharathiar [23] International Agency for Research on Cancer, IARC) - IARC Monographs on the
Evaluation of Carcinogenic Risks to Humans: Overall Evaluations of
University Campus Coimbatore, India for providing laboratory and Carcinogenicity: an Updating of IARC Monographs Volumes 1-42, Supplement 7,
instrumentation facilities. vol. 440, IARC, Lyon, 1987, ISBN 92 832 14110.
[24] D.E. Till, D.J. Ehntholt, R.C. Reid, P.S. Schwartz, A.D. Schwopc, R.R. Sidman, R.
H. Whelan, Migration of styrene monomer from crystal polystyrene to food and
Appendix A. Supplementary data food simulating liquid, Ind. Eng. Chem. Prod. Res. Dev. 21 (1982) 161–185.
[25] Z. Pilevar, A. Bahrami, S. Beikzadeh, H. Hosseini, S.M. Jafari, Migration of styrene
Supplementary data to this article can be found online at https://doi. monomer from polystyrene packaging materials into foods: characterization and
safety evaluation, Trends Food Sci. Technol. 91 (2019) 248–261.
org/10.1016/j.cbi.2021.109550. [26] A. Dimitriadi, C. Papaefthimiou, E. Genizegkini, I. Sampsonidis, S. Kalogiannis,
K. Feidantsis, D.C. Bobori, G. Kastrinaki, G. Koumoundouros, D.A. Lambropoulou,
References G.Z. Kyzas, Adverse effects polystyrene microplastics exert on zebrafish heart-
molecular to individual level, J. Hazard Mater. (2021), 125969.
[27] L.C. deSá, M. Oliveira, F. Ribeiro, T.L. Rocha, M.N. Futter, Studies of the effects of
[1] A. Jahnke, H.P.H. Arp, B.I. Escher, B. Gewert, E. Gorokhova, D. Kühnel,
microplastics on aquatic organisms: what do we know and where should we focus
M. Ogonowski, A. Potthoff, C. Rummel, M. Schmitt-Jansen, E. Toorman,
our efforts in the future? Sci. Total Environ. 645 (2018) 1029–1039.
M. MacLeod, Reducing uncertainty and confronting ignorance about the possible
[28] K. Enders, R. Lenz, C.A. Stedmon, T.G. Nielsen, Abundance, size and polymer
impacts of weathering plastic in the marine environment, Environ. Sci. Technol.
composition of marine microplastics ≥ 10μm in the Atlantic Ocean and their
Lett. 4 (2017) 85–90.
modelled vertical distribution, Mar. Pollut. Bull. 100 (2015) 70–81.
[2] T. Kögel, Ø. Bjorøy, B. Toto, A.M. Bienfait, M. Sanden, Micro- and nanoplastic
[29] M. Carbery, W. O’Connor, T. Palanisami, Trophic transfer of microplastics and
toxicity on aquatic life: determining factors, Sci. Total Environ. 709 (2020),
mixed contaminants in the marine food web and implications for human health,
136050.
Environ. Int. 115 (2018) 400–409.
[3] J.R. Jambeck, R. Geyer, C. Wilcox, T.R. Siegler, M. Perryman, A. Andrady,
[30] A. Elizalde-Velázquez, A.M. Carcano, J. Crago, M.J. Green, S.A. Shah, J.E. Cañas-
R. Narayan, K.L. Law, Marine pollution: plastic waste inputs from land into the
Carrell, Translocation, trophic transfer, accumulation and depuration of
ocean, Science 347 (2015) 768–771.
polystyrene microplastics in Daphnia magna and Pimephales promelas, Environ.
[4] L. Lei, S. Wu, S. Lu, M. Liu, Y. Song, Z.H. Fu, Shi, K.M. Raley-Susman, D. He,
Pollut. 259 (2020), 113937.
Microplastic particles cause intestinal damage and other adverse effects in
[31] F. Du, H. Cai, Q. Zhang, Q. Chen, H. Shi, Microplastics in take-out food containers,
zebrafish Danio rerio and nematode Caenorhabditis elegans, Sci. Total Environ.
J. Hazard Mater. 399 (2020), 122969.
619–620 (2018) 1–8.
[32] M. Revel, A. Chatel, C. Mouneyrac, Micro (nano) plastics: a threat to human
[5] J.S. Choi, Y.J. Jung, N.H. Hong, S.H. Hong, J.W. Park, Toxicological effects of
health? Curr Opin Environ Sci Health 1 (2018) 17–23.
irregularly shaped and spherical microplastics in a marine teleost, the sheepshead
[33] C.M. Rochman, A. Tahir, S.L. Williams, D.V. Baxa, R. Lam, J.T. Miller, F.C. Teh,
minnow (Cyprinodon variegatus), Mar. Pollut. Bull. 129 (2018) 231–240.
S. Werorilangi, S.J. Teh, Anthropogenic debris in seafood: plastic debris and fibers
[6] M. Cole, C. Liddle, G. Consolandi, C. Drago, C. Hird, P.K. Lindeque, TS.
from textiles in fish and bivalves sold for human consumption, Sci. Rep. 5 (2015),
Galloway Microplastics, Microfibres and nanoplastics cause variable sub-lethal
14340.
responses in mussels (Mytilus spp.), Mar. Pollut. Bull. 160 (2020), 111552.
[34] J. Hwang, D. Choi, S. Han, S.Y. Jung, J. Choi, J. Hong, Potential toxicity of
[7] M. Karamanlioglu, R. Preziosi, G.D. Robson, Abiotic and biotic environmental
polystyrene microplastic particles, Sci. Rep. 10 (2020) 7391.
degradation of the bioplastic polymer poly (lactic acid): a review, Polym. Degrad.
[35] Y. Lu, Y. Zhang, Y. Deng, W. Jiang, Y. Zhao, J. Geng, L. Ding, H. Ren, Uptake and
Stabil. 137 (2017) 122–130.
accumulation of polystyrene microplastics in zebrafish (Danio rerio) and toxic
[8] X. Zhang, X. Wang, B. Yan, Single and combined effects of phenanthrene and
effects in liver, Environ. Sci. Technol. 50 (2016) 4054–4060.
polystyrene microplastics on oxidative stress of the clam (Mactra veneriformis), Sci.
[36] L. Yin, H. Liu, H. Cui, B. Chen, L. Li, F. Wu, Impacts of polystyrene microplastics on
Total Environ. 771 (2021), 144728.
the behavior and metabolism in a marine demersal teleost, black rockfish (Sebastes
[9] S. Klein, I.K. Dimzon, J. Eubeler, T.P. Knepper, Analysis, occurrence, and
schlegelii), J. Hazard Mater. 380 (2019), 120861.
degradation of microplastics in the aqueous environment, in: M. Wagner,
[37] J.H. Kim, Y.B. Yu, J.H. Choi, Toxic effects on bioaccumulation, hematological
S. Lambert (Eds.), Freshwater Microplastics. The Handbook of Environmental
parameters, oxidative stress, immune responses and neurotoxicity in fish exposed
Chemistry, 2018.
to microplastics, A Rev. J. Hazard Mater (2021), 125423.
[10] J.M. Pereira, Y. Rodríguez, S. Blasco-Monleon, A. Porter, C.K. Pham, C.
[38] N.R. Brun, P. van Hage, E.R. Hunting, A.G. Haramis, S.C. Vink, M.G. Vijver,
K. Microplastic, In the stomachs of open-ocean and deep-sea fishes of the North-
M. Schaaf, C. Tudorache, Polystyrene nanoplastics disrupt glucose metabolism and
East Atlantic, Environ. Pollut. 265 (2020), 115060.
cortisol levels with a possible link to behavioural changes in larval zebrafish,
[11] M.A. Browne, S.J. Niven, T.S. Galloway, S.J. Rowland, R.C. Thompson,
Commun Biol 2 (2019) 382.
Microplastic moves pollutants and additives to worms, reducing functions linked to
[39] Q. Chen, M. Gundlach, S. Yang, J. Jiang, M. Velki, D. Yin, H. Hollert, Quantitative
health and biodiversity, Curr. Biol. 23 (2013) 2388–2392.
investigation of the mechanisms of microplastics and nanoplastics toward zebrafish
[12] S. Lambert, C.J. Sinclair, E.L. Bradley, A.B.A. Boxall, Effects of environmental
larvae locomotor activity, Sci. Total Environ. 584–585 (2017) 1022–1031.
conditions on latex dergadation in aquatic systems, Sci. Total Environ. 447 (2013)
[40] Y.S. Eltemsah, T. Bøhn, Acute and chronic effects of polystyrene microplastics on
225–234.
juvenile and adult Daphnia magna, Environ. Pollut. 254 (2019), 112919.
[13] N.P. Ivleva, A.C. Wiesheu, R. Niessner, Microplastic in aquatic ecosystems, Angew.
[41] W. Wang, J. Ge, X. Yu, Bioavailability and toxicity of microplastics to fish species: a
Chem. Int. Ed. 56 (2017) 1720–1739.
review, Ecotoxicol. Environ. Saf. 189 (2020), 109913.
[14] K. Zhang, A.H. Hamidian, A. Tubi, Y. Zhang, J.K.H. Fang, C. Wu, P.K.S. Lam,
[42] W. Gu, S. Liu, L. Chen, Y. Liu, C. Gu, H.Q. Ren, B. Wu, Single-cell RNA sequencing
Understanding plastic degradation and microplastic formation in the environment:
reveals size-dependent effects of polystyrene microplastics on immune and
a review, Environ. Pollut. 274 (2021), 116554.
secretory cell populations from zebrafish intestines, Environ. Sci. Technol. 54 (6)
[15] Y. Li, Z. Liu, M. Li, Q. Jiang, D. Wu, Y. Huang, Y. Jiao, M. Zhang, Y. Zhao, Effects of
(2020) 3417–3427.
nanoplastics on antioxidant and immune enzyme activities and related gene
[43] C.B. Jeong, H.M. Kang, M.C. Lee, D.H. Kim, J. Han, D.S. Hwang, S. Souissi, S.J. Lee,
expression in juvenile Macrobrachium nipponense, J. Hazard Mater. 398 (2020),
K.H. Shin, H.G. Park, J.S. Lee, Adverse effects of microplastics and oxidative stress-
122990.
induced MAPK/Nrf2 pathway-mediated defense mechanisms in the marine
[16] C.G. Alimba, C. Faggio, Microplastics in the marine environment: current trends in
copepod Paracyclopina nana, Sci. Rep. 7 (2017), 41323.
environmental pollution and mechanisms of toxicological profile, Environ. Toxicol.
[44] K. Yin, Y. Wang, H. Zhao, D. Wang, M. Guo, M. Mu, Y. Liu, X. Nie, B. Li, J. Li,
Pharmacol. 68 (2019) 61–74.
M. Xing, A comparative review of microplastics and nanoplastics: toxicity hazards
[17] Sources, fate and effects of microplastics in the marine environment: part two of a
on digestive, reproductive and nervous system, Sci. Total Environ. (2021), 145758.
global assessment 93, IMO/FAO/UNESCO-IOC/UNIDO/WMO/IAEA/UN/UNEP/
UNDP, in: GESAMP, P.J. Kershaw, C.M. Rochman (Eds.), Joint Group of Experts on

9
S. Umamaheswari et al. Chemico-Biological Interactions 345 (2021) 109550

[45] G.Y. Liou, P. Storz, Reactive oxygen species in cancer, Free Radic. Res. 44 (5) [70] D.W. Banner, A. D’Arcy, W. Janes, R. Gentz, H.J. Schoenfeld, C. Broger,
(2010) 479–496. H. Loetscher, W. Lesslauer, Crystal structure of the soluble human 55 kd TNF
[46] L. Qiang, J. Cheng, Exposure to polystyrene microplastics impairs gonads of receptor-human TNF beta complex: implications for TNF receptor activation, Cell
zebrafish (Danio rerio), Chemosphere 263 (2021), 12816. 73 (1993) 431–445.
[47] P. Yu, Z. Liu, D. Wu, V. Chen, W. Lv, Y. Zhao, Accumulation of polystyrene [71] R.M. Locksley, N. Killeen, M.J. Lenardo, The TNF and TNF receptor superfamilies:
microplastics in juvenile Eriocheir sinensis and oxidative stress effects in the liver, integrating mammalian biology, Cell 104 (2001) 487–501.
Aquat. Toxicol. 200 (2018) 28–36. [72] L. Gu, L. Tian, G. Gao, S. Peng, J. Zhang, D. Wu, J. Huang, Q. Hua, T. Lu, L. Zhong,
[48] J. Li, H. Liu, J. Paul Chen, Microplastics in freshwater systems: a review on Z. Fu, X. Pan, H. Qian, L. Sun, Inhibitory effects of polystyrene microplastics on
occurrence, environmental effects, and methods for microplastics detection, Water caudal fin regeneration in zebrafish larvae, Environ. Pollut. 266 (2020), 114664.
Res. 137 (2018) 362–374. [73] W. Czarnocka, S. Karpiński, Friend or foe? Reactive oxygen species production,
[49] X. Zhai, L. Wang, C. Xu, Q. Hou, L. Zhang, Z. Li, W. Qin, Z. Liu, Z. Chen, Triptolide scavenging and signaling in plant response to environmental stresses, Free Radic.
preserves glomerular barrier function via the inhibition of p53-mediated increase Biol. Med. 122 (2018) 4–20.
of GADD45B, Arch. Biochem. Biophys. 671 (2019) 210–217. [74] K. Linhart, H. Bartsch, H.K. Seitz, The role of reactive oxygen species (ROS) and
[50] S. Park, J.Y. Lee, H. Park, G. Song, W. Lim, Bifenthrin induces developmental cytochrome P-450 2E1 in the generation of carcinogenic etheno-DNA adducts,
immunotoxicity and vascular malformation during zebrafish embryogenesis, Redox Biol 3 (2014) 56–62.
Comp. Biochem. Physiol. C Toxicol. Pharmacol. 228 (2020), 108671. [75] S. Abarghouei, A. Hedayati, M. Raeisi, B.S. Hadavand, H. Rezaei, A. Abed-
[51] J.M. Salvador, J.D. Brown-Clay, A.J. Fornace Jr., Gadd45 in stress signaling, cell Elmdoust, Size-dependent effects of microplastic on uptake, immune system,
cycle control, and apoptosis, Adv. Exp. Med. Biol. 793 (2013) 1–19. related gene expression and histopathology of goldfish (Carassius auratus),
[52] I. Paul-Pont, C. Lacroix, C. González Fernández, H. Hégaret, C. Lambert, N. Le Goïc, Chemosphere 276 (2021), 129977.
L. Frère, A.L. Cassone, R. Sussarellu, C. Fabioux, J. Guyomarch, M. Albentosa, [76] Z. Liu, P. Yu, M. Cai, D. Wu, M. Zhang, Y. Huang, Y. Zhao, Polystyrene nanoplastic
A. Huvet, P. Soudant, Exposure of marine mussels Mytilus spp. to polystyrene exposure induces immobilization, reproduction, and stress defense in the
microplastics: toxicity and influence on fluoranthene bioaccumulation, Environ. freshwater cladoceran Daphnia pulex, Chemosphere 215 (2019) 74–81.
Pollut. 216 (2016) 724–737. [77] J.N. Huang, B. Wen, J.G. Zhu, Y.S. Zhang, J.Z. Gao, Z.Z. Chen, Exposure to
[53] S. Sarmah, J.A. Marrs, Zebrafish as a vertebrate model system to evaluate effects of microplastics impairs digestive performance, stimulates immune response and
environmental toxicants on cardiac development and function, Int. J. Mol. Sci. 17 induces microbiota dysbiosis in the gut of juvenile guppy (Poecilia reticulata), Sci.
(2016) 2123. Total Environ. 733 (2020), 138929.
[54] J. Bhagat, L. Zang, N. Nishimura, Y. Shimada Zebrafish, An emerging model to [78] K. Lu, R. Qiao, H. An, Y. Zhang, Influence of microplastics on the accumulation and
study microplastic and nanoplastic toxicity, Sci. Total Environ. 728 (2020) 138707. 727 chronic toxic effects of cadmium in zebrafish (Danio rerio), Chemosphere 728
[55] OECD, Organisation for Economic Co-operation and Development, Guidelines for 202 (2018) 514–520.
Testing of Chemicals No 305: Bioconcentration: Flow-Through Fish Test, 1996. [79] J. Wang, Y. Li, L. Lu, M. Zheng, X. Zhang, H. Tian, W. Wang, S. Ru, Polystyrene
Paris. microplastics cause tissue damages, sex-specific reproductive disruption and
[56] APHA, Standard methods for the examination of water and wastewater, Stand. transgenerational effects in marine medaka (Oryzias melastigma), Environ. Pollut.
Methods (2005) 541. 254 (2019), 113024.
[57] J. Ding, S. Zhang, R.M. Razanajatovo, H. Zou, W. Zhu, Accumulation, tissue [80] S. Karbalaei, P. Hanachi, G. Rafiee, P. Seifori, T.R. Walker, Toxicity of polystyrene
distribution, and biochemical effects of polystyrene microplastics in the freshwater microplastics on juvenile Oncorhynchus mykiss (rainbow trout) after individual and
fish red tilapia (Oreochromis niloticus), Environ. Pollut. 238 (2018) 1–9. combined exposure with chlorpyrifos, J. Hazard Mater. 403 (2021), 123980.
[58] X. Zhao, S. Wang, Y. Wu, H. You, L. Lv, Acute ZnO nanoparticles exposure induces [81] M. Qiu, Y. Wang, X. Wang, L. Sun, R. Ye, D. Xu, Z. Dai, Y. Liu, S. Bi, Y. Yao,
developmental toxicity, oxidative stress and DNA damage in embryo-larval R. Gooneratne, Effects of T-2 toxin on growth, immune function and
zebrafish, Aquat. Toxicol. 136–137 (2013) 49–59. hepatopancreas microstructure of shrimp (Litopenaeus vannamei), Aquaculture 462
[59] J.A. Adeyemi, A.da Cunha Martins, F. Barbosa, Teratogenicity, genotoxicity and (2016) 35, 39.
oxidative stress in zebrafish embryos (Danio rerio) co-exposed to arsenic and [82] J. Bilski, A. Mazur-Bialy, D. Wojcik, J. Zahradnik-Bilska, B. Brzozowski,
atrazine, Comp. Biochem. Physiol. C Toxicol. Pharmacol. 172–173 (2015) 7–12. M. Magierowski, T. Mach, K. Magierowska, T. Brzozowski, The role of intestinal
[60] A.K. Sinha, Colorimetric assay of catalase, Anal. Biochem. 47 (2) (1972) 389–394. alkaline phosphatase in inflammatory disorders of gastrointestinal tract, Mediat.
[61] S. Marklund, G. Marklund, Involvement of the superoxide anion radical in the Inflamm. 2017 (2017), 9074601.
autoxidation of pyrogallol and a convenient assay for superoxide dismutase, Eur. J. [83] M.L. Rodgers, R. Takeshita, R.J. Griffitt, Deepwater Horizon oil alone and in
Biochem. 47 (3) (1974) 469–474. conjunction with Vibrio anguillarum exposure modulates immune response and
[62] I. Domingues, R. Oliveira, J. Lourenço, C.K. Grisolia, S. Mendo, A.M. Soares, growth in red snapper (Lutjanus campechanus), Aquat. Toxicol. 204 (2018) 91–99.
Biomarkers as a tool to assess effects of chromium (VI): comparison of responses in [84] L. Zhang, X. Hong, X. Zhao, S. Yan, X. Ma, J. Zha, Exposure to environmentally
zebrafish early life stages and adults, Comp. Biochem. Physiol. C Toxicol. relevant concentrations of deltamethrin renders the Chinese rare minnow
Pharmacol. 152 (3) (2010) 338–345. (Gobiocypris rarus) vulnerable to Pseudomonas fluorescens infection, Sci. Total
[63] G.L. Ellman, K.D. Courtney, V. Andres Jr., R.M. Feather-stone, A new and rapid Environ. 715 (2020), 136943.
colorimetric determination of acetylcholinesterase activity, Biochem. Pharmacol. 7 [85] B. Hoffman, D.A. Liebermann, Gadd45 modulation of intrinsic and extrinsic stress
(1961) 88–95. responses in myeloid cells, J. Cell. Physiol. 218 (1) (2009) 26–31.
[64] K.J. Livak, T.D. Schmittgen, Analysis of relative gene expression data using real- [86] P.M. Eimon, A. Ashkenazi, The zebrafish as a model organism for the study of
time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods 25 (4) (2001) apoptosis, Apoptosis 15 (3) (2010) 331–349.
402–408. [87] C.H. Cheng, H.L. Ma, Y.Q. Deng, J. Feng, Y.K. Jie, Z.X. Guo, Oxidative stress, cell
[65] M. Teng, S. Qi, W. Zhu, Y. Wang, D. Wang, K. Dong, C. Wang, Effects of the cycle arrest, DNA damage and apoptosis in the mud crab (Scylla paramamosain)
bioconcentration and parental transfer of environmentally relevant concentrations induced by cadmium exposure, Chemosphere 263 (2021), 128277.
of difenoconazole on endocrine disruption in zebrafish (Danio rerio), Environ. [88] A. Karami, D.B. Groman, S.P. Wilson, P. Ismail, V.K. Neela, Biomarker responses in
Pollut. 233 (2018) 208–217. zebrafish (Danio rerio) larvae exposed to pristine low-density polyethylene
[66] W. Huang, X. Wang, D. Chen, E.G. Xu, X. Luo, J. Zeng, T. Huan, L. Li, Y. Wang, fragments, Environ. Pollut. 223 (2017) 466–475.
Toxicity mechanisms of polystyrene microplastics in marine mussels revealed by [89] V.V. Husak, N.M. Mosiichuk, I.V. Maksymiv, I.Y. Sluchyk, J.M. Storey, K.B. Storey,
high-coverage quantitative metabolomics using chemical isotope labeling liquid V.I. Lushchak, Histopathological and biochemical changes in goldfish kidney due
chromatography mass spectrometry, J. Hazard Mater. 417 (2021), 126003. to exposure to the herbicide Sencor may be related to induction of oxidative stress,
[67] M. Sharifinia, Z.A. Bahmanbeigloo, M. Keshavarzifard, M.H. Khanjani, B.P. Lyons, 768, Aquat Toxicol 155 (2014) 181–189.
Microplastic pollution as a grand challenge in marine research: a closer look at [90] H. Yang, H. Xiong, K. Mi, W. Xue, W. Wei, Y. Zhang, Toxicity comparison of nano-
their adverse impacts on the immune and reproductive systems, Ecotoxicol. sized and micron-sized microplastics to goldfish Carassius auratus Larvae, J. Hazard
Environ. Saf. 204 (2020), 111109. Mater. 388 (2020), 122058.
[68] M.J. Morgan, Y.S. Kim, Z.G. Liu, TNF alpha and reactive oxygen species in necrotic [91] M.B. Colović, Z. Krstić, T.D. Lazarević-Pašti, A.M. Bondžić, V.M. Vasić,
cell death, Cell Res. 18 (3) (2008) 343–349. Acetylcholinesterase inhibitors: pharmacology and toxicology, Curr.
[69] H. Sauer, M. Wartenberg, J. Hescheler, Reactive oxygen species as intracellular Neuropharmacol. 11 (3) (2013) 315–335.
messengers during cell growth and differentiation, Cell. Physiol. Biochem. 11
(2001) 173–186.

10

You might also like