Next Article in Journal
The Glutathione Peroxidase Gene Family in Chenopodium quinoa: Genome-Wide Identification, Classification, Gene Expression and Functional Analysis
Previous Article in Journal
High-Intensity Interval Training as Redox Medicine: Targeting Oxidative Stress and Antioxidant Adaptations in Cardiometabolic Disease Cohorts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidative Stress and Neurotoxicity Biomarkers in Fish Toxicology

1
Institute of Biology and Earth Sciences, University of the National Education Commission, Podchorążych 2, 31-054 Krakow, Poland
2
Department of Animal Biology and Environment, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
3
Department of Animal Environment Biology, Institute of Animal Science, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(8), 939; https://doi.org/10.3390/antiox14080939 (registering DOI)
Submission received: 28 May 2025 / Revised: 25 July 2025 / Accepted: 29 July 2025 / Published: 30 July 2025
(This article belongs to the Special Issue Reactive Oxygen Species Signalling and Oxidative Stress in Fish)

Abstract

Exposure to xenobiotics causes pathophysiological changes in fish, including oxidative stress and neurotoxicity. Here, we describe the biochemical mechanisms underlying oxidative stress (i.e., redox imbalance) and the biochemical markers commonly used to assess its level. Neurotoxicity biomarkers used in fish include behavioral, histological, molecular, neurotransmitter-related, and enzymatic parameters, among which acetylcholinesterase (AChE) activity is the most commonly measured. We therefore also review the changes in AChE activity in fish exposed to common xenobiotics. In most cases, AChE activity decreased in a concentration- and time-dependent manner, although some studies reported no change or even an increase. We emphasize the relevance of all the parameters discussed in the context of fish toxicology studies.

1. Introduction

Oxidative stress and neurotoxicity often co-occur in fish exposed to toxic agents [1,2,3,4,5]. Therefore, this review aims to summarize and discuss the most relevant and recent literature on the use of oxidative stress parameters and neurotoxicity biomarkers in fish toxicological studies.
Model organisms used in monitoring and laboratory studies possess biological characteristics that are representative of a broader group of aquatic organisms, particularly those belonging to the same taxonomic units. Therefore, it can be assumed that aquatic pollutants inducing oxidative stress or altering AChE activity act in a similar manner across a wider range of aquatic species. Such changes affect animal behavior, motor activity, and metabolism, ultimately leading to a decline in the animals’ physiological condition. Based on this, it can be inferred that certain contaminants, through mechanisms involving oxidative stress and disruption of neural transmission, may influence relationships within ecosystems. For instance, they may increase mortality, reduce reproductive capacity, and affect various types of interactions within the environment. However, it is important to keep in mind that the outcomes of scientific research are significantly influenced by the conditions under which the studies were conducted. Our experience shows that different researchers apply varying fish stocking densities, which can influence water quality parameters, including the concentration of unionized ammonia (NH3)—a compound toxic to fish and whose concentration is often not reported in experimental studies. Therefore, general conclusions should be drawn with caution, particularly when they address broader phenomena than those directly investigated.

2. Indicators of Redox Imbalance in Fish Toxicology

In aerobic organisms, oxygen undergoes four electron reductions to produce adenosine phosphates, which leads to the formation of reactive oxygen species (ROS). Some ROS are essential to physiological regulation and processes. Free radicals are ROS with unpaired electrons. Primary free radicals are superoxide radical (O2•−), hydroxyl radical (OH), hydroperoxyl radical (HOO), alkyl peroxyl radical (LOO), alkoxyl radical (LO), and nitric oxide (NO). They are unstable because they tend to form pairs with electrons of other compounds [6]. When oxidation and reduction reactions are unbalanced (i.e., redox imbalance), excessive amounts of free radicals are produced, which may lead to significant cellular damage [7,8].
The formation of free radicals may be assessed directly by the measurements of the O2•− or H2O2 formation [9,10]. This method is rarely used in ecotoxicological studies because of the extremely short lifetime of the O2•− anion or H2O2. Nonetheless, Falfushynska and Stoliar [11] indicated increased production of O2•− in the gills and liver of crucian carp (Carassius carassius) obtained from contaminated waters. Free radicals cause lipid peroxidation, oxidation of DNA bases followed by DNA fragmentation, inhibition of enzymes, and damage to numerous structural compounds [12,13,14]. The products of oxidative damage of chemical molecules are direct evidence of the occurrence of reactive oxygen species in the organism. They are often used as markers of oxidative stress (Table 1). Indication of lipid peroxides as a level of thiobarbituric acid reactive substances (TBARS) belongs to the most popular methods.
Several studies reported increased levels of TBARS in different organs of fish from polluted water. A correlation also occurred between TBARS and the production of O2•− in the liver and gills of common carp (Cyprinus carpio) [11]. On the other hand, in fish organs with high antioxidative capacity, increased activity of antioxidative enzymes may not be followed by increased TBARS levels [39]. Products of guanosine hydroxylation such as 8-hydroxy-2′-deoxyguanosine (8-OHdG), 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxodG), and 8-hydroxyguanine (8-OHGua) indicate genotoxic effects of ROS. Analytical assays focused on the measurements of oxidative damage of guanosine indicated increased urinary levels of 8-OHGua in fish from polluted environments. However, most studies on the genotoxic effects of ROS in fish are conducted in controlled laboratory conditions. Other products of oxidative damage are protein carbonyls. Protein carbonyls may be a helpful indicator of oxidative stress under controlled laboratory conditions in the studies of model oxidative stress. Measurements of the level of protein carbonyls are rarely used in ecotoxicological studies. Despite their infrequent use in ecotoxicology, protein carbonyl measurements have revealed elevated levels in various tissues of gibel carp (Carassius gibelio) inhabiting an aquatic ecosystem influenced by a hydropower plant [40].
Organisms are protected against oxidative stress by nonenzymatic and enzymatic antioxidants. Nonenzymatic antioxidants provide electrons to free radicals, preventing oxidative damage. They include both endogenous and exogenous substances: vitamins (e.g., vitamin E, ascorbic acid), carotenoids, thiols (e.g., glutathione), uric acid, metallothioneins (MTs), polyphenols, flavonoids, and other compounds. Nonenzymatic antioxidants are non-specific and constitute the first defense against ROS [41]. Enzymatic antioxidants comprise specific antioxidative enzymes. Superoxide dismutase (SOD) converts O2•− to H2O2. Manganese superoxide dismutase (MnSOD) expresses activity in mitochondria, while Cu,ZnSOD (copper-zinc superoxide dismutase) shows activity in cytosol and extracellular fluid [42]. Catalase (CAT) and glutathione peroxidase (GPx) catalyze the reduction of H2O2 to molecular oxygen and H2O. Their activity is usually coupled with SOD to prevent excessive H2O2 synthesis. Glutathione S-transferase (GST) also plays a vital role in preventing oxidative stress, which catalyzes the conjugation of harmful electrophiles to reduced glutathione (GSH) [42].
Both enzymatic and nonenzymatic antioxidants are often used as markers of oxidative stress. Nonenzymatic molecules are oxidized in the ROS reactions, so their levels may decrease during ROS excess. Metallothioneins (MTs) are induced mainly by zinc, cadmium, copper, and lead and are also engaged in metabolizing many other metals. Another essential function of MTs is protection against free radicals, where their methionine serves as a free radical scavenger [43]. Ecotoxicological studies mainly focus on MTs in the context of metal exposure and/or metal induction of oxidative stress. It was reported that fish from metal-contaminated environments showed increased MT gene expression, accompanied by elevated expression of antioxidative enzymes [44]. Increased MT concentration, oxidative damage, and enhanced activity of enzymatic antioxidants in the liver and gills of pelagic fish from metal-contaminated sites have also been documented [45].
Glutathione is one of the most studied molecules in fish subjected to oxidative stress. Most studies report increased concentrations of GSH in fish liver, kidneys, and gills from contaminated environments [9,46,47]. Increased GSH may suggest activating the antioxidative system or the phase II biotransformation reactions. The oxidized to reduced glutathione GSSG/GSH ratio may be used as the tissue redox state indicator. The relationship between GSSG/GSH and environmental contamination was found in crucian carp (Carassius carassius): the fish from contaminated water had higher GSSG/GSH than those from relatively clean water [48].
Adaptation of the cells to increased ROS production includes increased expression of genes encoding antioxidative enzymes and an increase in enzyme activity. Specific enzyme activity against ROS provides a valuable indicator of the redox balance in environmental studies. It has been shown that the activity of antioxidative enzymes (SOD, CAT, GR—glutathione reductase, GPx) is significantly correlated with oxidative damage products (TBARS, protein carbonyls) in the liver of sea trout (Salmo trutta) from the Słupia River (Poland) at different developmental stages [49]. Another study demonstrated differences in GPx gene expression in common carp (Cyprinus carpio) inhabiting two distinct water reservoirs in India [44].

Organic Pollutants and Redox Imbalance

The influence of organic pollution on redox balance is complex. It mainly depends on the chemical properties of the compounds and their metabolism in fish organisms. Most of the organic pollutants affect the redox balance by several different mechanisms. A good example is the redox imbalance caused by ammonia. It occurs in two forms, NH4+ and NH3, which can be converted into each other. Exposure to ammonia causes the formation of ROS such as O2•−, OH, H2O2, and ROO [50]. In estuarine teleost European sea bass (Dicentrarchus labrax), exposure to ammonia caused ROS formation and MDA accumulation in the brain, liver, gills, muscles, and kidneys. This effect was accompanied by the increased activity of SOD, CAT, GPx, and GR and increased concentration of GSH. However, these effects showed different dynamics in different tissues [50], which suggests different modulation of response to ammonia and free radicals in various tissues. Several mechanisms participate in ammonia-induced oxidative stress. First, and probably the most important to fish, is gill damage and subsequent hypoxia. Hypoxia shifts the mitochondrial redox balance toward a more reduced state, which may increase ROS production [7,51]. During hypoxia, the biosynthesis of nuclear factor-κb (NF-κb), an essential transcriptional regulator of proinflammatory factors, increases in fish gills [52]. Upregulation of proinflammatory cytokines TNF-α and IL-1β was also reported in other fish organs after exposure to ammonia [53,54]. Generally, inflammation is induced by free radicals, although some ROS are released by immune cells during the inflammation [55]. It may be expected that the immune system generates significant amounts of ROS in hypoxia. Ammonia has also influenced the expression of genes controlling redox balance. In great blue-spotted mudskippers (Boleophthalmus pectinirostris), exposure to ammonia resulted in the downregulation of the nuclear factor erythroid 2-related factor 2 (NRF2) [54]. It is known that the downregulation of NRF2 causes the downregulation of downstream genes encoding SOD, CAT, and GPx and the subsequent redox imbalance. Notably, glyphosate-based herbicides induce the exact mechanism studied in the liver tissue of common carp [17]. Another mechanism of oxidative stress induction by ammonia occurs in fish brains. Ammonia overstimulates the NMDA receptor. This causes a large influx of Ca2+ into the neural cells, followed by the generation of ROS and excitotoxicity [56].
Organic compounds undergo biotransformation by cytochrome P450 enzymes (CYPs). In fish, the CYP1 subfamily is expressed, and CYP1A is often used to indicate exposure to organic pollution [57]. Induction of CYP1A activates mixed-function oxygenases (MFOs), which catalyze the oxygenation of xenobiotics by single oxygen addition [58]. Redox reactions leading to the biotransformation of xenobiotics cause the formation of ROS. One of the most studied monooxygenases in fish is ethoxyresorufin-O-deethylase (EROD). Fish organs, mainly gills and liver, exposed to organic xenobiotics show increased activity of EROD and an increased rate of free radicals’ formation [59]. The participation of CYPs in oxidative stress induction in fish was confirmed by the studies on the toxicity of different organic compounds, including pyrethroid pesticides, hydrocarbons, chlorinated hydrocarbons, dioxins, organophosphates, and carbamates [58,59,60,61,62,63].
Other mechanisms mediating the induction of oxidative stress by organic xenobiotics are disturbances in mitochondrial electron transport, inhibition of enzymes, and depletion of nonenzymatic antioxidants. An example of a pesticide that induces oxidative stress by inhibiting electron transport in mitochondria is fenpyroximate [61]. Exposure to fenpyroximate caused increased activity of SOD, GPx, and CAT in the gill cell line and liver tissue of flounder (Paralichthys olivaceus) [64]. Electron transport in mitochondria may be interrupted by enzyme inhibition. Chlorothalonil, an organochlorine pesticide, inhibits the activity of NADPH oxidase by binding to its SH groups. This was found to cause disturbances in electron transport in mitochondria and the generation of free radicals in macrophages of striped bass (Morone saxatilis) [61]. Other mechanisms of oxidative imbalance may be related to the depletion of antioxidant levels. Pesticides may cause GSH depletion through two mechanisms, i.e., generating free radicals and conjugating with GSH in the second detoxification phase. Both effects may lead to oxidative stress. Such a mechanism of induction of oxidative stress was indicated after exposure of rainbow trout to organophosphates, methylparathion, and diazinon. Oxidative imbalance also occurred in the rainbow trout’s liver, gills, and muscles [65]. An interesting mechanism occurs in fish exposed to paraquat, a bipyridyl herbicide. Paraquat is reduced to its cation radical by glutathione reductase (GR); next, the radical is neutralized by the conjugation to GSH. This way, the radical chain reaction is stopped, although it leads to the depletion of GSH and lower protection against ROS. Moreover, GSH depletion leads to increased GR activity and a generation rate of paraquat cation radicals. The occurrence of oxidative stress accompanied by depletion of GSH and increased activity of GR seem to confirm the above mechanism in Nile tilapia (Oreochromis niloticus) [61].
The effects of toxic agents on fish oxidative stress parameters are presented in Table 2. In the cited studies, the most frequently determined parameters of oxidative stress in fish were glutathione; enzymatic antioxidants: SOD, CAT, GPx, and GST; and oxidative damage products: MDA and TBARS. Only in approximately 15% of the analyzed cases were no changes in the mentioned oxidative stress parameters noted. In comparison, in 85% of cases, their increase and/or decrease in activity/concentration under the influence of various toxicants was reported. In particular groups of substances, a diverse biological response was observed in fish, probably due to the mechanism of action of the antioxidant barrier, which may be species-specific. The results (Table 2) show that most aquatic pollutants induced oxidative stress in fish, which is indicated by increased levels of oxidation products (e.g., TBARS, MDA, and LPO). Various effects on antioxidant defense were observed: an increase, sometimes accompanied by no symptoms of oxidative stress, indicating efficiency; or a decrease, suggesting a breakdown of the antioxidant system. These effects were related to the toxicant’s concentrations.
Differences in individual research results may also be related to the differences in experimental conditions (water parameters, exposure duration, etc.). Despite discrepancies in the research results and difficulties in determining changes typical for given substances and/or fish species, the enzymatic and nonenzymatic parameters discussed in this review are commonly used in fish toxicology (both in field and laboratory studies) and provide valuable information about the organisms’ status.

3. Neurotoxicity Biomarkers in Fish Toxicology

The development and use of neurotoxicity biomarkers is an essential issue in toxicology because of numerous emerging pollutants, such as microplastics, pharmaceuticals, and other novel chemical compounds, as well as “old” contaminants. Metals and pesticides exhibit neurotoxic potential, leading to diverse deficits in neural systems representing varying levels of evolutionary advancement [109]. Neurotoxicity in fish may result in disturbed schooling, migration, spatial distribution, feeding, reproduction, and predator avoidance. Neurotoxicity biomarkers used in fish include histopathological [110,111] and morphometric [112] evaluation of the brain, various behavioral endpoints [113,114,115], expression of marker genes associated with neuron development and growth [116], and the levels of neurotransmitters in brain [117,118]. However, changes in the activity of cholinesterases (ChEs) are the most often used biomarkers of aquatic pollution because these enzymes are frequent targets for toxic effects of contaminants such as insecticides or metals [119]. This group of enzymes includes, among others, acetylcholinesterase (AChE), butyrylcholinesterase (BChE), propionylcholinesterase (PChE), and carboxylesterase (CbE). These enzymes are found in various tissues and are involved in detoxification processes in fish. The sensitivity of BChE, PChE, and CbE to different pesticides and other compounds can vary not only between fish species but also between different tissues within the same species. Moreover, their precise physiological roles remain unclear. Among cholinesterases, AChE is the most commonly used biochemical biomarker of neurotoxicity. AChE activity is usually measured in the brain and muscles, and also in the whole body (in the case of embryos and larvae), or in other organs such as liver and gills [120]. Brain or muscle AChE inhibition significantly correlated with reductions in spontaneous swimming and feeding activity in fish, and threshold concentrations for sublethal neurochemical and behavioral endpoints were similar [121,122].

3.1. Acetylcholinesterase as a Neurotoxicity Biomarker

Acetylcholinesterase (AChE), a serine carboxylesterase, is an important component of cholinergic synapses that terminates impulse transmission by rapid hydrolysis of the neurotransmitter acetylcholine (ACh). AChE is a very efficient catalyst that accelerates the substrate hydrolysis by more than eight orders of magnitude [123]. An AChE catalytic action includes a two-stage reaction: acylation and deacylation [124]. Although the primary function of AChE is to terminate neural transmission, it was also found that AChE plays a role in neural development [125]. AChE occurs in all tissues and is most abundant in the brain and muscles. AChE inhibition increases ACh concentration and neurotransmitter action [126], causing cognitive and behavioral disturbances. AChE activity is a sensitive biomarker of fish exposure to toxic agents [127]. However, its use requires an understanding of natural fluctuations. Seasonal variation in AChE activity has been observed in Cnesterodon decemmaculatus, with the highest values recorded in summer and a marked decrease in winter, likely associated with water temperature differences [128]. Seasonal changes in AChE activity and sensitivity to pesticides (glyphosate and chlorpyrifos) were also reported by other researchers for the same fish species [129].
Table 3 summarizes the effects of various toxic agents on AChE activity (100 papers containing 119 data points; in some studies, the toxicity of more than one compound was evaluated). Most data (45%) concern pesticides (35%—insecticides, 8%—herbicides, 2%—fungicides). Other groups of data concern pharmaceuticals and disinfectants (12%), elements (10%), microplastics (7%), and nanoparticles (6%). The remaining 18% of data describe the effects of other compounds such as antifouling agents, cosmetic components, aromatic hydrocarbons, cyanotoxins, plastic components, and others. Most experiments (44%) were conducted on embryos, larvae, or adults of Danio rerio.
Analysis of the methods of AChE activity measurements (in 100 papers) revealed that most authors (66%) used the method developed and described by Ellman et al. [130], 23% used commercial biochemical kits, 4% used other methods, and 7% did not specify the method used. Spectrophotometric cuvettes (16%) were often replaced by microplates (27%), and 57% of authors did not specify the reading method (but commercial kit users presumably used microplates; thus, microplates were probably used in most studies). However, it has been shown that copper, zinc, cadmium, and mercury affect the results of AChE measurements in vitro, likely due to their reactions with the products of Ellman’s reaction [131]. This poses a challenge to obtaining accurate data, particularly when assessing the in vitro neurotoxicity of these metals.
Table 3. The effects of toxic agents on brain (or whole body—in embryos and larvae) acetylcholinesterase (AChE) activity in fish. The original values are given in parentheses and expressed in the following units: ° mg/L * µg/L # ng/L ×× mg/kg feed ××× g/kg feed $ µg/kg sediment “” mg/L algal extract @ particles/L.
Table 3. The effects of toxic agents on brain (or whole body—in embryos and larvae) acetylcholinesterase (AChE) activity in fish. The original values are given in parentheses and expressed in the following units: ° mg/L * µg/L # ng/L ×× mg/kg feed ××× g/kg feed $ µg/kg sediment “” mg/L algal extract @ particles/L.
GCSFish SpeciesToxic AgentConcentration [µmol]Exp. dur. [d]AChE ActivityReference
MD. reriomicroplastic(10,000 @)5Xue et al. [132]
MD. rerio embryosmicroplastic (0.1–10 °)4Suman et al. [133]
MD. rerio embryosmicroplastic (0.1–3 °)5Martin-Folgar et al. [134]
MO. javanicusmicroplastic(0.5–5 °)21Usman et al. [135]
MO. mossambicuspolypropylene microplastic(100 ××)
(500 ××)
(1000 ××)
14

Jeyavani et al. [136]
MO. niloticusmicroplastic(100 °)21Yang et al. [137]
M
E
D. reriomicroplastic(2 °)30Santos et al. [138]
copper (as CuSO4•5H2O)0.157 (25 *)
M
E
D. labraxmicroplastic (0.26 ° or 0.69 °)4Barboza et al. [72]
mercury (as HgCl2)0.037–0.059 (0.010 ° or 0.016 °)
EA. testudineuschromium VI (as CrO3)27.503 or 55.006 (2.75 or 5.5 °)72Kumar et al. [139]
EC. decemmaculatusarsenic (as NaAsO2)3.849–38.488
(0.5–5 °)
4González Núñez et al. [140]
E
E
D. reriochromium III (as CrCl3•6H2O)
chromium VI (as (K2Cr2O7)
6.315 (1 °)
3.400 (1 °)
5
Xu et al. [141]
ED. rerioaluminum203.840 (5.5 °)15Alves et al. [142]
ED. rerio embryosmercury (as HgCl2)36.830 (10 °)

368.297 (100 °)
1
2–4
1
2–4



Henriques et al. [143]
ED. rerio embryos + larvaeantimony (as K2Sb2C8H4O12
• 3 H2O)
299.460–1197.838
(200–800 °)
2Xia et al. [144]
EH. molitrix larvaemercury (as HgCl2)0.004–0.037
(1–10 *)
14Wang et al. [145]
EO. niloticusaluminum (as Al2(SO4)30.003–0.009
(1 or 3 *)
14Oliveira et al. [146]
E
N
O. niloticustitanium (as TiO2)
titanium (as TiO2 NPs)
0.626 or 1.252 (0.05 or 0.1 °)7–14
Abegoda-Liyanage and Pathiratne [147]
ND. rerioselenium NPs6.331 or 126.629 (0.5 or 10 °)4Fan et al. [148]
ND. reriosilver NPs9.271 (1 °)
0.028–0.046 (3–5 *)
4
Marinho et al. [149]
N
N
N
N
O. mykissgraphene nanoflakes
graphene oxide
reduced graphene oxide
silicon carbide nanofibers
(4 °)36Jakubowska-Lehrmann et al. [150]
FD. reriopaclobutrazol34.038 (10 °)4–14Guo et al. [151]
FD. reriothifuzamide0.360 (0.19 °)
3.598 or 5.397 (1.9 or 2.85 °)
6Yang et al. [152]
FD. rerio embryosmancozeb0.0009 (0.5 *)
0.009 (5 *)
0.092 (50 *)
4

Vieira et al. [153]
HD. rerio embryosRoundup®
glyphosate
1.479 (0.25 °)2Ames et al. [154]
HD. rerio larvaeRoundup®0.028 (4.8 *)5Pompermaier et al. [155]
HD. rerio larvaehaloxyfop-p-methyl0.532–1.065
(0.2–0.4 °)
4Liu et al. [156]
HO. niloticuspendimethalin1.848 (0.52 °)28Hamed and El-Sayed [157]
HP. lineatusRoundup®5.915 or 29.574
(1 or 5 °)
4Modesto and Martinez [2]
H
I
C. carpioglyphosate
chlorpyrifos
20.701 (3.5 °)
0.071 (25 *)
21
Zhang et al. [158]
H
I
I
T. niloticadiuron
Nemacur®
malathion
4.290 (1 °)
0.330–6.593 (0.1–2 °)
0.303–6.054 (0.1–2 °)
1

El-Nahhal [159]
H
I
D. rerioDMA® 806 BR (Fipronil)
Regent® 800 WG (2,4-D)
0.134 (63.5 *)
(447 *)
4
Viana et al. [160]
IC. auratus gibeliotrichlorfon1942.2–7768.8
(0.5–2 ×××)
0.5–4Lu et al. [161]
IC. carpio
C. idella
A. nobilis
diafuran14.267–48.802
(1–3 °)
4Golombieski et al. [162]
IC. carpiochlorpyrifos0.066 or 0.131 (23 or 46 *)14Pala et al. [163]
IC. carpioλ cyhalothrin0.0003 or 0.0006 (0.14 or 0.28 *)15–45Chatterjee et al. [164]
IC. macropomummalathion22.097 (7.3 °)4de Souza et al. [90]
IC. macropomumtrichlorfon1.010 or 1.670 (0.26 or 0.43 °)1–4Duncan et al. [165]
I
I
C. punctatustriazophos
deltamethrin
0.011 or 0.022 (3.4 or 6.8 *)
0.007 or 0.0014 (0.36 or 0.72 *)
4
Singh et al. [166]
IC. umblachlorpyrifos156.880 (55 °)

313.760 (110 °)
1
4
1
4



Kirici [111]
IClarias batrachusthiamethoxam23.756 or 47.513 (6.93 or 13.86 °)45Mukherjee et al. [167]
I
I
D. reriochlorpyrifos
cyfluthrin
0.003 (1.16 *)
0.016 or 0.033 (7.06 or 14.12 *)
5
Zhang et al. [168]
ID. reriodinotefuran0.989 (0.2 °)
4.946 (1 °)
28
Ran et al. [169]
ID. rerioimidacloprid0.0006 (0.15 *)
0.059 or 0.176 (15 or 45 *)
4
Guerra et al. [170]
I
I
D. rerioimidacloprid
thiamethoxam
0.0002–0.078 (0.05–20 *)
0.0002–0.069 (0.05–20 *)
14–35
Zhang et al. [171]
ID. reriosulfoxaflor3.138–12.659 (0.87–3.51 °)4Benli and Celik [172]
ID. reriomethomyl3.082–143.641 (0.5–23.3 °)6Jablonski et al. [173]
ID. rerio embryoschlorphoxim7.513–22.540 (2.5–7.5 °)4Xiong et al. [174]
ID. rerio larvaefenpropathrin0.046–0.183 (0.016–0.064 °)4Yu et al. [175]
ID. rerio larvaeisoprocarb5.175–12.937 (1–2.5 °)6Wang et al. [176]
IG. affinischlorpyrifos0.847 (0. 297 °)4Kavitha and Rao [177]
IG. affiniscypermethrin4.8 × 10−7 or 1.5 × 10−5
(0.2 or 6.25 #)
7Touaylia et al. [178]
IG. affiniscarbofuran0.863 or 0.701 (0.191 or 0.255 °)15–40Rouachdia et al. [179]
IH. fossilischlorpyrifos0.257 or 0.548 (0.09 or 0.192°)7–30Mishra et al. [180]
I

I
J. multidentatacypermethrin

chlorpyrifos
9.610 × 10−5 or 9.610 × 10−4
(0.04 or 0.4 *)
0.001 or 0.011 (0.4 or 4 *)
4

Bonansea et al. [122]
IO. latipesdiazinon0.033 or 0.066 (10 or 20 *)122Flynn et al. [181]
IO. mykisschlorpyrifos6.413 or 12.830
(2.25 or 4.5 °)
0.021 (7.25 *)
0.021 (7.25 *)
1–4

1–2
3–4



Topal et al. [182]
IO. mykisschlorpyrifos0.006 (2 *)
0.006 (2 *)
0.011 (4 *)
0.017 (6 *)
7
14–21
7–21
7–21



Mehtabidah et al. [183]
IO. mykissphosmet0.016 (5 *)
0.016 (5 *)
0.079 or 0.158 (25 or 50 *)
1–2
3–4
1–4


Muhammed and Dogan [184]
IO. mykiss larvaechlorpyrifos0.0009 (0.3 *)
0.009 (3 *)
21
Weeks Santos et al. [185]
IO. niloticuschlorpyrifos14.251–42.753
(5–15 *)
30Oruç [3]
I

I

I
O. niloticusmalathion

chlorpyrifos

λ-cyhalothrin
4.313 (1.425 °)

0.357 (0.125 °)

0.009 (0.0039 °)
1
2
1
2
1
2





Amin et al. [186]
IO. niloticuscarbofuran1.112 (0.246 °)30Hamed et al. [187]
IP. lineatusfipronil12.581 (5.5 °)
0.188 (82 $)
15
Santillan Deiú et al. [188]
IR. quelentrichlorfon42.728 (11 °)21Baldissera et al. [189]
PdCorydoras paleatustriclosan0.653 (189 *)2Sager et al. [190]
PdD. reriofluoxetine0.016–0.052 (5–16 #)4Orozco-Hernández et al. [191]
PdD. reriofluoxetine0.003–0.0323 (0.1–10 *)21Correia et al. [192]
PdD. rerio embryos + larvaecloramine T70.286 (16 °)
140.573 (32 °)
281.146 (64 °)
562.291 (128 °)
4


Rivero-Wendt et al. [193]
PdD. rerio embryos + larvae2,5-dichloro-1,4-benuinone1.130 (0.2 °)
2.260 or 3.390 (0.4 or 0.6 °)
4
Chen et al. [194]
PdDanio reriometformin0.009–0.310 (1–40 *)120Elizalde-Velázquez et al. [195]
PdDanio reriosertraline3.266 (1 °)
32.655 or 326.552 (10 or 100 °)
28
Yang et al. [196]
PdDanio rerionortriptyline0.003–1.898 (0.88–500 *)7Oliveira et al. [197]
PdDanio rerio embryosmoxidectin0.002–0.008 (1.5–5 *)4Muniz et al. [198]
PdDanio rerio embryos + larvaesertraline3.266–326.552 (1–100 °)10Yang et al. [152]
PdGambusia affinisgestodene1.42 × 10−5 (4.4 #)
0.0012 (378.7 #)
60
Tan et al. [199]
PdOreochromis mossambicustriclosan0.452–3.613 (0.131–1.046 °)4Deepika et al. [200]
PdOreochromis niloticussynthetic progesterone0.636–2.544 (0.2–0.8 °)4Rocha et al. [201]
PdRhamdia quelenciprofloxacin0.003 (1 *)
0.030 or 0.302 (10 or 100 *)
28
Carvalho et al. [202]
OA. testudineusnaphthalene32.769–39.011 (4.2–5.0 °)3Nayak and Patnaik [203]
O
O
C. carpioammonia NH3
nitrite NO2-
1802.701 (30.7 °)
3341.304 (153.7 °)
4Molayemraftar et al. [204]
O
O
O
C. gariepinusbenzene
toluene
xylene
9.76 × 10−6 (0.762 #)
0.0003 (26.614 #)
0.0011 (89.403 #)
30

Sayed et al. [68]
OC. gariepinusburnt tyre ash(0.56–2.24 °)28Iheanacho et al. [205]
OC. mrigalaphenol24.652–73.956
(2.32 or 6.96 °)
7–28Muthukumaravel et al. [206]
OD. reriotributylin3.24 × 10−5 (10 #)
0.0003–0.001 (100–300 #)
42
Li and Li [207]
OD. reriobisphenol A0.001–0.007 (0.22–1.5 *)4Heredia-Garcia et al. [208]
OD. reriobisphenol AF0.149 or 1.487 (0.05 or 0.5 °)4Rao et al. [209]
OD. reriomethylparaben0.007 or 0.072 (1 or 11 *)30Thakkar et al. [210]
OD. rerioethanol108.535 (5 °)7–28Agostini et al. [211]
OD. rerio embryosbisphenol A49.936 (11.4 °)1Murugan et al. [212]
OD. rerio embryosmethylparaben0.0007 or 0.007 (0.1 or 1 *)6Raja et al. [213]
OD. rerio embryosoctocrylene0.014 (5 *)
0.138 or 1.383 (50 or 500 *)
4
Gayathri et al. [214]
OD. rerio embryosbenzophenone-30.001 or 0.010 (1 or 10 *)3Sandoval-Gío et al. [215]
OD. rerio embryos + larvaeammonia NH33.523–49.912 (0.06–0.85 °)7Mariz et al. [216]
O

O
D. rerio larvaehexabromobenzene

pentabromobenzene
0.054 (30 *)
0.181–0.544 (100–300 *)
0.063–0.212 (30–100 *)
0.635 (300 *)
6


Chen et al. [217]
OG. affinisbisphenol A20.763 or 33.904 (4.74 or 7.74 °)15–60Belhamra et al. [218]
OG. affinisdecabromodiphenyl ether0.026 or 0.052 (25 or 50 *)2Pérez-Iglesias et al. [219]
OO. mossambicusammonia NH358.720 (1 °)28–56Gopi et al. [220]
OO. mossambicusdichloromethane8595–9301 (730–790 °)4Nirmala et al. [221]
OO. niloticusbenzylparaben2.19 × 10−5–0.022 (0.005–5 *)56Lin et al. [222]
OO. niloticusguanitoxin(125 or 250 “”)4Passos et al. [223]
GCS—group of chemical substance; E—metals and other elements; F—fungicides; H—herbicides; I—insecticides; M—microplastics; N—nanoparticles; O—other substances; Pd—pharmaceuticals and disinfectants; Exp. dur.—exposure duration; d—days; n/a—not applicable; NPs—nanoparticles; A. testudineusAnabas testudineus, A. nobilisAristychthys nobilis, C. umblaCapoeta umbla, C. auratus gibelioCarassius auratus gibelio, C. punctatusChanna punctatus, C. mrigalaCirrhinus mrigala, C. gariepinusClarias gariepinus, C. decemmaculatusCnesterodon decemmaculatus, C. macropomumColossoma macropomum, C. idellaCtenopharyngodon idella, C. carpioCyprinus carpio, D. rerioDanio rerio, D. labraxDicentrarchus labrax, G. affinisGambusia affinis, H. fossilisHeteropneustes fossilis, H. molitrixHypophthalmichthys molitrix, J. multidentataJenynsia multidentata, O. mykissOncorhynchus mykiss, O. mossambicusOreochromis mossambicus, O. niloticusOreochromis niloticus, O. javanicusOryzias javanicus, O. latipesOryzias latipes, P. lineatusProchilodus lineatus, R. quelenRhamdia quelen, T. niloticaTilapia nilotica.

3.2. The Effects of Toxic Agents on Acetylcholinesterase Activity

Analysis of the results shown in Table 3 revealed that in most cases (64%), AChE inhibition was reported; in 25% of cases, there was no change; and only in 11% of cases did an increase in AChE activity occur. In cases where no significant change observed, most data (44%) concerned lower concentrations of agents, while at higher concentrations, changes occurred; in 35% of cases, no change was a single result for one concentration tested or observed at all levels of a studied agent, and in 18% of cases, after shorter times of exposure. In contrast, at longer times, changes were observed, and only in 3% of cases (one case) did a nonlinear reaction occur, and no change was reported at the intermediate concentration of an agent. These data showed apparent concentration- and time-related effects of chemicals on AChE activity and a high responsiveness and sensitivity of this enzyme to various toxic agents. This confirms that AChE is a valuable neurotoxicity biomarker.
The results of many studies indicate that various aquatic pollutants may modulate AChE activity in fish, e.g., organophosphorus and carbamate insecticides are well-known AChE inhibitors that act by specifically binding to the active site of AChE and blocking the access of the physiological substrate [4,126,224,225], as well as organophosphate esters, used as plasticizers and flame retardants [226]. However, chemicals other than carbamates and organophosphates have also been documented to alter acetylcholinesterase activities (Table 3). In vitro inhibition of Cyprinus carpio AChE by 35 insecticides and their derivatives was tested [227]. It was found that various chemical forms of active compounds (e.g., oxon vs. thiono or diethyl vs. dimethyl) exhibited different inhibitory potencies. Also, combinations of different insecticides showed an additive inhibitory effect [227]. An in vitro study of the impact of metal ions on Diodon hystrix brain AChE activity [228] revealed inhibition order: Cr6+ < Co2+ < Ag2+ < Cu2+ < Pb2+ < As5+ < Cd2+ < Zn2+ < Ni2+ < Hg2+; and proved that AChE activity is a valuable biomarker for evaluating metal toxicity to fish.
AChE inhibitors include irreversible and reversible ones [126]. Reversible inhibitors, competitive or noncompetitive, mostly have therapeutic applications, while toxic effects are associated with irreversible AChE activity modulators [126].
The data of various studies showing the effects of aquatic pollutants on the fish brain or whole-body AChE activity (Table 3) reveal that most examined agents, including microplastics, pharmaceuticals, pesticides, metals, etc., usually inhibited the enzyme. Still, the effects were concentration- and time-related. AChE activation also sometimes occurred, or non-linear alternate bidirectional changes were observed at various concentrations or different times of exposure to the same agent, making interpretation of the neurotoxic effect difficult. In most cases of microplastic exposures, inhibition of AChE was observed; however, at low concentrations, enzyme activation sometimes occurred [135,138]. Also, most fish exposure to pharmaceuticals resulted in AChE inhibition, except for some antidepressants (sertraline) that induced no changes or activated the enzyme. The different effects observed in fish are related to the duration of exposure and the concentration of the pharmaceutical [152,196]. Most pesticides, such as insecticides, herbicides, fungicides, and antifouling agents, caused AChE inhibition in fish, often concentration- and/or time-related. Many commonly used triazine, carbamate, organophosphate, neonicotinoid, methylurea, or phenylurea pesticides are AChE inhibitors due to their binding to the AChE active site [229]. For some pesticides that usually inhibited AChE, activation was reported in single cases [155,186,208], which might have been related to the very low concentrations used. Inhibition of AChE activity by other chemicals was also reported for: disinfectants, bisphenol A, cosmetic compounds, organic solvents, metals and other elements, nitrogenous metabolites, and other aquatic pollutants. It is worth mentioning that, in the case of fish exposure to nanoparticles, AChE activity was rarely inhibited (Table 3), suggesting that environmentally relevant concentrations of these pollutants exhibit low neurotoxicity. According to the review by Olivares-Rubio and Espinosa-Aguirre [230], polycyclic aromatic hydrocarbons (e.g., benzo[a]pyrene, pyrene, and anthracene) inhibited AChE activity. Still, PAHs with a low molecular weight did not induce changes in AChE activity.
Acetylcholinesterase activity is sometimes measured in various tissues, and the results are not always the same as for the brain. They also differ among the organs examined. Different values and time- and concentration-related splenic and cardiac AChE activity patterns were observed in Oncorhynchus mykiss treated with clothianidin (a neonicotinoid insecticide) [231]. No alterations in brain AChE activities were reported following TiO2 and nano-TiO2 exposures, but an increase occurred in the gill and liver [147]. Different patterns of gill and muscle AChE activity changes over time of exposure to three pesticides, compared with the brain, were also reported [186]. No changes in brain AChE activity were observed following low and high cypermethrin or chlorpyrifos exposures, while muscle AChE activities significantly decreased after exposures to high concentrations of both pesticides [122]. AChE activity in the brain and muscles of three cyprinid fish species exposed to diafuran similarly decreased [162]. A considerable and significant decrease in AChE activity in the muscle of trichlorfon-exposed Oreochromis niloticus was also reported [232]. Similar changes in the brain and muscle AChE activities were observed in Prochilodus lineatus exposed to Roundup [2] and in Danio rerio treated with sulfoxaflor [172]. The decrease in AChE activities measured in the muscle, liver, and gill of Oreochromis mossambicus exposed to dichloromethane followed the decline in the brain [221]. However, the control value of AChE activity and the difference between the values for the control and exposed fish were the highest in the brain and muscle. The highest AChE activity was reported in the brain and muscle of Anabas testudineus. Enzyme inhibition following exposure to naphthalene also occurred in the gill and liver [203]. Chlorpyrifos reduced Danio rerio muscle AChE activity in a concentration-dependent manner. In contrast, no such relationship was observed for copper—a significant decrease in AChE activity occurred only at the lowest Cu concentration (6.3 µg/L) [233]. Similar activities and inhibition of brain and muscle AChE were reported in Tor putitora collected from various sampling sites with different pollution levels [234]. Some results showed an even higher sensitivity of muscle AChE, compared with the brain, to nano-Ag intoxication. These data indicate that AChE activity in muscle is probably an equally reliable biomarker as brain or whole-body AChE activity [149]. On the other hand, brain and muscle AChE may sometimes show opposite responses to toxic agents. In Leiarius marmoratus and Pseudoplatystoma reticulatum hybrids (pintado da Amazônia), AChE activity decreased in muscle but increased in the brain compared with the control group after Roundup exposure [235]. AChE activity in various tissues of Danio rerio was inhibited almost simultaneously when the fish were exposed to high concentrations of toxic agents (Cd2+ or deltamethrin). In contrast, at lower concentrations, inhibition occurred with a delay relative to the brain: brain > gill > muscle > liver [236]. These data suggest that muscle AChE activity should be interpreted with caution. Moreover, caution is also necessary because, for some toxic agents, AChE may not serve as a reliable biomarker of neurotoxicity. For example, a study conducted on Danio rerio revealed no changes in brain AChE activity during treatment with a 0.5% ethanol solution, while a significant decrease in acetylcholine level and choline acetyltransferase activity was observed [211].
Inhibition of AChE activity is usually associated with oxidative stress. Experimentally induced oxidative stress—caused by exposure of Danio rerio embryos to H2O2—resulted in a considerable and significant decrease in AChE activity [237]. However, the role of oxidative stress in modulating AChE activity remains unclear. According to various studies, oxidative stress may be involved in both the decrease [238,239] and increase [240] of AChE activity.

3.3. Intergenerational Effects of Toxic Agents on Acetylcholinesterase Activity

Although limited data are available, they indicate that AChE activity can serve as a biomarker of reproductive and intergenerational toxic effects, likely due to the parental transfer of neurotoxic compounds via gametes to the offspring. Larvae of Danio rerio obtained from adults exposed to a herbicide formulation containing 2,4-dichlorophenoxyacetic acid (2,4-D) exhibited elevated AChE activity [155]. In contrast, inhibition of AChE activity was observed in the offspring of Danio rerio adults exposed to rhodamine B dye [241]. It was also observed [242] that parental Danio rerio exposure to microcystin caused a reduction in dopamine, dihydroxyphenylacetic acid (DOPAC), and serotonin accompanied by hypoactivity and decreased AChE activity in the larvae. Microcystin was detected in the gonads of adults. It was probably transferred to the offspring, which affected the expression of genes involved in the neurotransmitter functions and neuronal development. It has also been reported that following parental exposure to benzo[a]pyrene, a significant reduction in AChE activity was observed in the larvae, along with decreased swimming velocity [116]. Genes involved in neuron growth and brain development were downregulated in the F1 larvae, while brain function in the exposed F0 adults remained unaffected. These findings suggest an epigenetic effect. A similar intergenerational epigenetic effect of chlorpyrifos-oxon on AChE activity has been observed [243]. Epigenetic mechanisms include acetylation or phosphorylation of histones, DNA methylation, and interactions of non-coding RNA during transcription. These may affect gene and protein expression [244] and suggest that neurotoxic compounds can be transferred via the gonads to the gametes and the offspring, causing developmental disturbances by changing gene expression. The effects observed in the next generation depend on the type of substance and the duration of exposure. Understanding the transgenerational impacts of environmental pollutants may help explain changes in species dynamics. Therefore, monitoring the effects of such pollutants is crucial as they may influence future generations and pose a threat to species survival.

3.4. Other Neurotoxicity Biochemical Biomarkers

In fish toxicology, other biochemical biomarkers of neurotoxicity are also occasionally used, such as the activities of BChE, PChE, and CbE, as well as levels of GABA (γ-aminobutyric acid); however, available data remain limited. Although GABA plays a key role as the primary inhibitory neurotransmitter in the central nervous system of fish, its use as a biomarker of neurotoxicity is associated with several significant limitations. GABA levels can be influenced not only by neurotoxic substances but also by general environmental stress, metabolic changes, temperature fluctuations, oxidative stress, and other forms of physiological disturbance [245,246]. Therefore, such changes may be nonspecific and do not always clearly indicate damage to the nervous system [245,246]. Monoamines such as dopamine, norepinephrine, and serotonin are valuable biochemical markers for assessing subtle neurotoxic effects in fish. However, like GABA, their diagnostic value increases when analyzed in conjunction with behavioral responses, enzymatic activities, and transcriptomic parameters. In Cyprinus carpio exposed to the dithiopyridine herbicide, concentrations of the neurotransmitters dopamine, norepinephrine, and acetylcholine were evaluated, revealing a decrease in catecholamine levels. In contrast, acetylcholine (ACh) concentration increased, accompanied by AChE inhibition [117]. In zebrafish exposed to methamphetamine, a concentration- and time-dependent decrease in brain levels of dopamine, norepinephrine, and serotonin was observed [118]. According to a meta-analysis by Santana et al. [247], fish cholinesterases (ChEs) are commonly used as biomarkers of environmental contamination due to their high sensitivity to a broad range of toxicants, including organophosphate and carbamate pesticides. The authors concluded that BChE response was more variable than AChE regardless of tissue, and no difference between their average activities was detected. Other authors emphasize the need to conduct species- and tissue-specific studies to characterize esterases before selecting them for use in monitoring programs [248]. The BChE enzyme is mainly associated with plasma or nonspecific tissue compartments, and in Solea senegalensis it shows minimal activity in relevant tissues such as muscle [249]. This limits its utility as a broad or sensitive biomarker across species and tissues. It was reported that malathion inhibited AChE and BChE activities in Senegalese sole (Solea senegalensis) in a dose- and time-dependent manner. Still, BChE activity was approximately two orders of magnitude lower than that of AChE [249].

4. Limitations of the Presented Studies

In this article, we have aimed to present the results of significant, well-designed, and carefully conducted scientific studies. However, it should be noted that these studies also have certain limitations related to the adopted conventions and methodologies.
Scientists conducting research on fish living in their natural habitats must contend with the dynamic changes occurring in the environment. Even slight variations in water temperature or oxygenation can strongly influence fish behavior as well as their motor and metabolic activity. The concentration of toxic substances in the environment can change significantly within just a few hours. Moreover, these substances exist in various chemical forms and engage in numerous interactions with both chemical and physical factors—factors that can also interact with one another. The half-lives of different xenobiotics in the environment vary and are influenced by physicochemical conditions.
The results of ecophysiological studies conducted in the natural environment provide a very accurate reflection of the physiological condition of fish living in waters with varying levels of pollution. However, it is not possible to determine the relationship between the dose or concentration of xenobiotics in the environment and their physiological effects based on the results of such studies.
The experimental regime used in laboratory studies allows for very precise determination of the physiological effects caused by specific doses of xenobiotics. However, a major limitation of this type of research is that experimental conditions often differ significantly from those found in natural environments. In many such studies, the doses of xenobiotics used are much higher than those typically encountered by fish in natural water bodies. Moreover, laboratory experiments often lack many of the environmental factors present in natural waters that can either neutralize toxic substances or enhance their toxicity.
Additionally, individual experiments often differ in key conditions that are fundamental to fish physiology, such as ammonium (NH3) concentration, nitrate (NO3) and nitrite (NO2) level, total and carbonate hardness, salinity, dissolved oxygen concentration, pH, temperature, and so on. Furthermore, in some publications, authors do not provide precise information about the physicochemical conditions under which the experiments were conducted.
The dose/physiological effect relationship is primarily influenced by the chosen experimental model, the method of chemical administration, and the duration of exposure. Different fish species exhibit varying sensitivities to xenobiotics, and, in general, substances administered by injection tend to produce a stronger physiological effect than those absorbed from water or ingested with food. Table 2 and Table 3 provide summaries that allow for a more detailed analysis of this issue.
To illustrate the mechanisms of neurotoxicity, we have cited the results of several experiments conducted under in vitro conditions. In vitro techniques typically involve cell cultures that do not account for interactions with bioregulators present in the organism or with other cells that together form tissues and organs. While the findings of such studies are scientifically valuable, they should be interpreted with great caution when extrapolating to conditions within the organism—and even more so when extrapolating to those in the natural environment.

5. Conclusions

Most aquatic pollutants cause oxidative stress in fish, the symptoms of which are elevated levels of oxidation products and changes in activities and concentrations of the components of the endogenous antioxidant system. Oxidative stress is a primary mechanism of tissue damage and physiological disorders. AChE activity is a sensitive and valuable but nonspecific biomarker of neurotoxicity in fish. It can be affected by many aquatic pollutants that mainly cause enzyme inhibition, and the responses are concentration- and time-related. This results in disturbed acetylcholine hydrolysis, the consequences of which are disturbed sensory, cognitive, and motor functions. Many aquatic pollutants show oxidative and neurotoxic potential, including developmental toxicity, which is a considerable threat not only to fish but also to their consumers and users of contaminated water: wildlife, domestic animals, and humans. Therefore, the inclusion of oxidative stress parameters and AChE activity measurements in routine natural water quality biomonitoring and toxicological studies is recommended. However, such assessments in toxicological experiments should be conducted under standardized experimental conditions, including defined physicochemical water parameters and appropriate selection of model organisms and tested tissues.

6. Recommendations

The analysis we conducted while preparing this review article led us to formulate a set of recommendations which, we hope, may serve as guidance for other researchers. Our recommendations are as follows:
(1)
In toxicity studies of various chemicals, it is essential—following ethical considerations and recommendations—to include biochemical parameters, such as indicators of oxidative stress and AChE activity.
(2)
In toxicological experiments, the potential influence of the physicochemical parameters of water on the results of biochemical analyses should be taken into account. It is important that researchers measure and report individual water quality parameters, such as the concentrations of ammonia, nitrites, and nitrates.
(3)
The vast majority of toxicological experiments focus on the effects of a single substance on fish of a given species. There is a need for studies aimed at determining the effects of co-exposure to multiple toxicants (e.g., pesticides and heavy metals).
(4)
There are few studies that include more than one fish species. Comparative studies involving various fish species simultaneously are needed.
(5)
It is recommended to conduct toxicological studies taking into account various development stages of fish (embryos, larvae, fry, sexually mature individuals).
(6)
Detailed information on the mechanisms of action and physiological roles of cholinesterases other than AChE in fish is lacking. Further research is needed to address this knowledge gap.

Author Contributions

Conceptualization, G.F., B.B. and M.W.; methodology, G.F. and M.W.; software, G.F. and M.W.; validation, G.F. and M.W.; formal analysis, G.F., B.B. and M.W.; investigation, G.F., Z.G., B.B. and M.W.; resources, G.F., Z.G., B.B. and M.W.; data curation, G.F., Z.G., B.B. and M.W.; writing—original draft preparation, G.F., Z.G., B.B. and M.W.; writing—review and editing, G.F., Z.G., B.B. and M.W.; visualization, G.F., Z.G., B.B. and M.W.; supervision, G.F., B.B. and M.W.; project administration, G.F., B.B. and M.W.; funding acquisition, G.F. and M.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The dataset supporting the reported results can be found at scientific databases, e.g., Web of Science or PubMed.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
2,4-D2,4-dichlorophenoxyacetic acid
8-OHdG8-hydroxy-2′-deoxyguanosine
8-OHGua8-hydroxyguanine
8-oxodG8-oxo-7,8-dihydro-2′-deoxyguanosine
A. nobilisAristychthys nobilis
A. testudineusAnabas testudineus
AChAcetylcholine
AChEAcetylcholinesterase
AhAromatic hydrocarbons
BChEButyrylcholinesterase
C. auratus gibelioCarassius auratus gibelio
C. carpioCyprinus carpio
C. decemmaculatusCnesterodon decemmaculatus
C. gariepinusClarias gariepinus
C. idellaCtenopharyngodon idella
C. macropomumColossoma macropomum
C. mrigalaCirrhinus mrigala
C. punctatusChanna punctatus
C. umblaCapoeta umbla
CATCatalase
CbECarboxylesterase
ChEsCholinesterases
Cu,ZnSODCopper-zinc superoxide dismutase
CYPsCytochrome P450 enzymes
dDays
D. labraxDicentrarchus labrax
D. rerioDanio rerio
DNADeoxyribonucleic acid
DOPACDihydroxyphenylacetic acid
EElements
ERODEthoxyresorufin-O-deethylase
Exp. dur.Exposure duration
FFungicides
G. affinisGambusia affinis
GABAγ-aminobutyric acid
GCSGroup of chemical substance
GPxGlutathione peroxidase
GRGlutathione reductase
GSHReduced glutathione
GSSGOxidized glutathione
GSTGlutathione S-transferase
HHerbicides
H. fossilisHeteropneustes fossilis
H. molitrixHypophthalmichthys molitrix
IInsecticides
J. multidentataJenynsia multidentata
LPOLipid peroxidation
mMinutes
MMicroplastics
MDAMalondialdehyde
MFOsMixed-function oxygenases
MnSODManganese superoxide dismutase
MTsMetallothioneins
NNanoparticles
n/aNot applicable
NMDA receptorN-methyl-D-aspartate receptor
NPsNanoparticles
NRF2Nuclear factor erythroid 2-related factor 2
OOther substances
O. javanicusOryzias javanicus
O. latipesOryzias latipes
O. mossambicusOreochromis mossambicus
O. mykissOncorhynchus mykiss
O. niloticusOreochromis niloticus
P. lineatusProchilodus lineatus
PChEPropionylcholinesterase
PODPeroxidase
R. quelenRhamdia quelen
ROSReactive oxygen species
SODSuperoxide dismutase
T. niloticaTilapia nilotica
TAOTotal antioxidant
TBARSThiobarbituric acid reactive substances
TPXTotal peroxides

References

  1. Kroon, F.; Streten, C.; Harries, S.A. Protocol for identifying suitable biomarkers to assess fish health: A systematic review. PLoS ONE 2017, 12, e0174762. [Google Scholar] [CrossRef]
  2. Modesto, K.A.; Martinez, C.B.R. Effects of Roundup Transorb on fish: Hematology, antioxidant defenses and acetylcholinesterase activity. Chemosphere 2010, 81, 781–787. [Google Scholar] [CrossRef] [PubMed]
  3. Oruç, E.Ö. Oxidative stress, steroid hormone concentrations and acetylcholinesterase activity in Oreochromis niloticus exposed to chlorpyrifos. Pest. Biochem. Physiol. 2010, 96, 160–166. [Google Scholar] [CrossRef]
  4. Rodriguez-Fuentes, G.; Rubio-Escalante, F.J.; Noreña-Barroso, E.; Escalante-Herrera, K.S.; Schlenk, D. Impacts of oxidative stress on acetylcholinesterase transcription, and activity in embryos of zebrafish (Danio rerio) following Chlorpyrifos exposure. Comp. Biochem. Physiol. C 2015, 172–173, 19–25. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, J.-W.; Kim, J.-H.; Lee, D.-C.; Lim, H.-J.; Kang, J.-C. Toxic effects on oxidative stress, neurotoxicity, stress, and immune responses in juvenile olive flounder, Paralichthys olivaceus, exposed to waterborne hexavalent chromium. Biology 2022, 11, 766. [Google Scholar] [CrossRef]
  6. Yoshikawa, T.; Naito, Y. What is oxidative stress. J. Jap. Med. Ass. 2002, 45, 271–276. [Google Scholar]
  7. Aon, M.A.; Cortassa, S.; O’Rourke, B. Redox-optimized ROS balance: A unifying hypothesis. Biochim. Biophys. Acta 2010, 1797, 865–877. [Google Scholar] [CrossRef]
  8. Lichtenberg, D.; Pinchuk, I. Oxidative stress, the term and the concept. Biochem. Biophys. Res. Comm. 2015, 461, 441–444. [Google Scholar] [CrossRef]
  9. Stoliar, O.B.; Lushchak, V.I. Environmental pollution and oxidative stress in fish. In Oxidative Stress—Environmental Induction and Dietary Antioxidants; Lushchak, V.I., Ed.; IntechOpen: London, UK, 2012. [Google Scholar]
  10. Romano, N.; Renugdas, N.; Fischer, H.; Shrivastava, J.; Baruah, K.; Egnew, N.; Sinha, A.K. Differential modulation of oxidative stress, antioxidant defense, histomorphology, ion-regulation and growth marker gene expression in goldfish (Carassius auratus) following exposure to different dose of virgin microplastics. Comp. Biochem. Physiol. C 2020, 238, 108862. [Google Scholar] [CrossRef]
  11. Falfushynska, H.I.; Stoliar, O.B. Function of metallothioneins in carp Cyprinus carpio from two field sites in Western Ukraine. Ecotoxicol. Environ. Saf. 2009, 72, 1425–1432. [Google Scholar] [CrossRef]
  12. Loro, V.L.; Jorge, M.B.; da Silva, K.R.; Wood, C.M. Oxidative stress parameters and antioxidant response to sublethal waterborne zinc in a euryhaline teleost Fundulus heteroclitus: Protective effects of salinity. Aquat. Toxicol. 2012, 110–111, 187–193. [Google Scholar] [CrossRef] [PubMed]
  13. Alvim, T.M.; Reis Martinez, C.B. Genotoxic and oxidative damage in the freshwater teleost Prochilodus lineatus exposed to the insecticides lambda-cyhalothrin and imidacloprid alone and in combination. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2019, 842, 85–93. [Google Scholar] [CrossRef] [PubMed]
  14. Paital, B.; Guru, D.; Mohapatra, P.; Panda, B.; Parida, N.; Rath, S.; Kumar, V.; Saxena, P.S.; Srivastava, A. Ecotoxic impact assessment of graphene oxide on lipid peroxidation at mitochondrial level and redox modulation in fresh water fish Anabas testudineus. Chemosphere 2019, 224, 796–804. [Google Scholar] [CrossRef] [PubMed]
  15. Tanaka, R.; Miyamoto, H.; Inoue, S.I.; Shigeta, K.; Kondo, M.; Ito, T.; Kodama, H.; Miyamoto, H.; Matsushita, T. Thermophile-fermented compost as a fish feed additive modulates lipid peroxidation and free amino acid contents in the muscle of the carp, Cyprinus carpio. J. Biosci. Bioengin. 2016, 121, 530–535. [Google Scholar] [CrossRef]
  16. Baker, B.P.; Van Wie, I.; Braun, E.; Jimenez, A.G. Thermal stability vs. variability: Insights in oxidative stress from a eurytolerant fish. Comp. Biochem. Physiol. A 2020, 249, 110767. [Google Scholar] [CrossRef]
  17. Liu, J.; Dong, C.; Zhai, Z.; Tang, L.; Wang, L. Glyphosate-induced lipid metabolism disorder contributes to hepatotoxicity in juvenile common carp. Environ. Pollut. 2021, 269, 116186. [Google Scholar] [CrossRef]
  18. Sánchez-Nuño, S.; Sanahuja, I.; Fernández-Alacid, L.; Ordóñez-Grande, B.; Carbonell, T.; Ibarz, A. Oxidative attack during temperature fluctuation challenge compromises liver protein homeostasis of a temperate fish model. Comp. Biochem. Physiol. B 2019, 236, 110311. [Google Scholar] [CrossRef]
  19. Safari Asl, R.; Shariatmadari, F.; Sharafi, M.; Torshizi, M.A.K.; Shahverdi, A. Dietary fish oil supplemented with vitamin E improves quality indicators of rooster cold-stored semen through reducing lipid peroxidation. Cryobiology 2018, 84, 15–19. [Google Scholar] [CrossRef]
  20. Tan, X.; Sun, Z.; Liu, Q.; Ye, H.; Zou, C.; Ye, C.; Wang, A.; Lin, H. Effects of dietary ginkgo biloba leaf extract on growth performance, plasma biochemical parameters, fish composition, immune responses, liver histology, and immune and apoptosis-related genes expression of hybrid grouper (Epinephelus lanceolatus♂ × Epinephelus fuscoguttatus♀) fed high lipid diets. Fish Shellfish Immunol. 2018, 72, 399–409. [Google Scholar] [CrossRef]
  21. Topal, A.; Çomakli, S.; Özkaraca, M.; Baran, A.; Köktürk, M.; Parlak, V.; Sağlam, Y.S.; Atamanalp, M.; Ceyhun, S.B. Immunofluorescence evaluation of 4-hydroxynonenal and 8-hydroxy-2-deoxyguanosine activation in zebrafish (Daino rerio) larvae brain exposed (microinjected) to propyl gallate. Chemosphere 2017, 183, 252–256. [Google Scholar] [CrossRef]
  22. Pulster, E.L.; Fogelson, S.; Carr, B.E.; Mrowicki, J.; Murawski, S.A. Hepatobiliary, PAHs and prevalence of pathological changes in Red Snapper. Aquat. Toxicol. 2021, 230, 105714. [Google Scholar] [CrossRef]
  23. Raibeemol, K.P.; Chitra, K.C. Induction of immunological, hormonal and histological alterations after sublethal exposure of chlorpyrifos in the freshwater fish, Pseudetroplus maculatus (Bloch, 1795). Fish Shellfish Immunol. 2020, 102, 1–12. [Google Scholar] [CrossRef]
  24. Zhong, Y.; Shen, L.; Ye, X.; Zhou, D.; He, Y.; Zhang, H. Mechanism of immunosuppression in zebrafish (Danio rerio) spleen induced by environmentally relevant concentrations of perfluorooctanoic acid. Chemosphere 2020, 249, 126200. [Google Scholar] [CrossRef] [PubMed]
  25. Gómez Manrique, W.; Pereira Figueiredo, M.A.; Charlie-Silva, I.; Belo, M.A.A.; Corsi Dib, C. Spleen melanomacrophage centers response of Nile tilapia during Aeromanas hydrophila and Mycobacterium marinum infections. Fish Shellfish Immunol. 2019, 95, 514–518. [Google Scholar] [CrossRef] [PubMed]
  26. Ottinger, C.A.; Smith, C.R.; Blazer, V.S. In vitro immune function in laboratory-reared age-0 smallmouth bass (Micropterus dolomieu) relative to diet. Fish Shellfish Immunol. 2019, 95, 1–10. [Google Scholar] [CrossRef] [PubMed]
  27. Mahboub, H.H.; Khedr, M.H.E.; Elshopakey, G.E.; Shakweer, M.S.; Mohamed, D.I.; Ismail, T.A.; Ismail, S.H.; Rahman, A.N.A. Impact of silver nanoparticles exposure on neuro-behavior, hematology, and oxidative stress biomarkers of African catfish (Clarias gariepinus). Aquaculture 2021, 544, 737082. [Google Scholar] [CrossRef]
  28. Jia, R.; Du, J.; Cao, L.; Feng, W.; He, Q.; Xu, P.; Yin, G. Application of transcriptome analysis to understand the adverse effects of hydrogen peroxide exposure on brain function in common carp (Cyprinus carpio). Environ. Pollut. 2021, 286, 117240. [Google Scholar] [CrossRef]
  29. Mohamed, A.A.; El-Houseiny, W.; EL-Murr, A.E.; Ebraheim, L.L.M.; Ahmed, A.I.; El-Hakim, Y.M.A. Effect of hexavalent chromium exposure on the liver and kidney tissues related to the expression of CYP450 and GST genes of Oreochromis niloticus fish: Role of curcumin supplemented diet. Ecotoxicol. Environ. Saf. 2020, 188, 109890. [Google Scholar] [CrossRef]
  30. Mohamed, A.A.; Rahman, A.N.A.A.; Mohammed, H.H.; Ebraheim, L.L.M.; Abo-ElMaaty, A.M.A.; Ali, S.A.; Elhady, W.M. Neurobehavioral, apoptotic, and DNA damaging effects of sub-chronic profenofos exposure on the brain tissue of Cyprinus carpio L.: Antagonistic role of Geranium essential oil. Aquat. Toxicol. 2020, 224, 105493. [Google Scholar] [CrossRef]
  31. Topal, A.; Alak, G.; Altun, S.; Erol, H.S.; Atamanalp, M. Evaluation of 8-hydroxy-2-deoxyguanosine and NFkB activation, oxidative stress response, acetylcholinesterase activity, and histopathological changes in rainbow trout brain exposed to linuron. Environ. Toxicol. Pharmacol. 2017, 49, 14–20. [Google Scholar] [CrossRef]
  32. Almeida, É.C.; Passos, L.S.; Vieira, C.E.D.; Acayaba, R.D.; Montagner, C.C.; Pinto, E.; Martinez, C.B.D.R.; Fonseca, A.L. Can the insecticide imidacloprid affect the health of the Neotropical freshwater fish Astyanax altiparanae (Teleostei: Characidae)? Environ. Toxicol. Pharmacol. 2021, 85, 103634. [Google Scholar] [CrossRef]
  33. Zhang, L.; Zheng, X.C.; Huang, Y.Y.; Ge, Y.P.; Sun, M.; Chen, W.L.; Liu, W.B.; Li, X.F. Carbonyl cyanide 3-chlorophenylhydrazone induced the imbalance of mitochondrial homeostasis in the liver of Megalobrama amblycephala: A dynamic study. Comp. Biochem. Physiol. C 2021, 244, 109003. [Google Scholar] [CrossRef] [PubMed]
  34. Blank do Amaral, A.M.; Kuhn de Mour, L.; de Pellegrin, D.; Guerra, L.J.; Cerezer, F.O.; Saibt, N.; Prestes, O.D.; Zanella, R.; Loro, V.L.; Clasen, B. Seasonal factors driving biochemical biomarkers in two fish species from a subtropical reservoir in southern Brazil: An integrated approach. Environ. Pollut. 2020, 266, 115168. [Google Scholar] [CrossRef] [PubMed]
  35. Iturburu, F.G.; Bertrand, L.; Mendieta, J.R.; Amé, M.V.; Menone, M.L. An integrated biomarker response study explains more than the sum of the parts: Oxidative stress in the fish Australoheros facetus exposed to imidacloprid. Ecol. Indic. 2018, 93, 351–357. [Google Scholar] [CrossRef]
  36. Feng, L.; Gan, L.; Jiang, W.D.; Wu, P.; Liu, Y.; Jiang, J.; Tang, L.; Kuang, S.Y.; Tang, W.N.; Zhang, Y.A.; et al. Gill structural integrity changes in fish deficient or excessive in dietary isoleucine: Towards the modulation of tight junction protein, inflammation, apoptosis and antioxidant defense via NF-κB, TOR and Nrf2 signaling pathways. Fish Shellfish Immunol. 2017, 63, 127–138. [Google Scholar] [CrossRef]
  37. Shahab, M.; Rosati, R.; Meyer, D.N.; Shields, J.N.; Crofts, E.; Baker, T.R.; Jamesdaniel, S. Cisplatin-induced hair cell loss in zebrafish neuromasts is accompanied by protein nitration and Lmo4 degradation. Toxicol. Appl. Pharmacol. 2021, 410, 115342. [Google Scholar] [CrossRef]
  38. Lee, J.W.; Deng, D.F.; Lee, J.; Kim, K.; Jung, H.J.; Choe, Y.; Park, S.H.; Yoon, M. The adverse effects of selenomethionine on skeletal muscle, liver, and brain in the steelhead trout (Oncorhynchus mykiss). Environ. Toxicol. Pharmacol. 2020, 80, 103451. [Google Scholar] [CrossRef]
  39. Huang, D.J.; Zhang, Y.M.; Song, G.; Long, J.; Liu, J.H.; Ji, W.H. Contaminants-induced oxidative damage on the carp Cyprinus carpio collected from the Upper Yellow River, China. Environ. Monit. Ass. 2007, 128, 483–488. [Google Scholar] [CrossRef]
  40. Falfushynska, H.; Horyn, O.; Fedoruk, O.; Khoma, V.; Rzymski, P. Difference in biochemical markers in the gibel carp (Carassius auratus gibelio) upstream and downstream of the hydropower plant. Environ. Pollut. 2019, 255, 113213. [Google Scholar] [CrossRef]
  41. Haider, K.; Haider, M.R.; Neha, K.; Yar, M.S. Free radical scavengers: An overview on heterocyclic advances and medicinal prospects. Eur. J. Med. Chem. 2020, 204, 112607. [Google Scholar] [CrossRef]
  42. Delfino, R.J.; Staimer, N.; Vaziri, N.D. Air pollution and circulating biomarkers of oxidative stress. Air Qual. Atm. Health 2011, 4, 37–52. [Google Scholar] [CrossRef] [PubMed]
  43. Nordberg, M. Metallothioneins: Historical review and state of knowledge. Talanta 1998, 46, 243–254. [Google Scholar] [CrossRef] [PubMed]
  44. Banday, U.Z.; Swaleh, S.B.; Usmani, N. Heavy metal toxicity has an immunomodulatory effect on metallothionein and glutathione peroxidase gene expression in Cyprinus carpio inhabiting a wetland lake and a culture pond. Chemosphere 2020, 251, 126311. [Google Scholar] [CrossRef] [PubMed]
  45. Beg, M.U.; Al-Jandal, N.; Al-Subiai, S.; Karam, Q.; Husain, S.; Butt, S.A.; Ali, A.; Al-Hasan, E.; Al-Dufaileej, S.; Al-Husaini, M. Metallothionein, oxidative stress and trace metals in gills and liver of demersal and pelagic fish species from Kuwaits’ marine area. Mar. Pollut. Bull. 2015, 100, 662–672. [Google Scholar] [CrossRef]
  46. Pandey, S.; Parvez, S.; Sayeed, I.; Haque, R.; Bin-Hafeez, B.; Raisuddin, S. Biomarkers of oxidative stress: A comparative study of river Yamuna fish Wallago attu (Bl. & Schn.). Sci. Total Environ. 2003, 309, 105–115. [Google Scholar] [CrossRef]
  47. Bacanskas, L.R.; Whitaker, J.; Di Giulio, R.T. Oxidative stress in two populations of killifish (Fundulus heteroclitus) with differing contaminant exposure histories. Mar. Environ. Res. 2004, 58, 597–601. [Google Scholar] [CrossRef]
  48. Falfushynska, H.; Gnatyshyna, L.; Priydun, C.; Stoliar, O.; Nam, Y.K. Variability of responses in the crucian carp carassius from two Ukrainian ponds determined by multi-marker approach. Ecotoxicol. Environ. Saf. 2010, 73, 1896–1906. [Google Scholar] [CrossRef]
  49. Kurhaluk, N. Formation of an antioxidant profile in the sea trout (Salmo trutta m. trutta L.) from the Slupia River. Zoology 2019, 133, 54–65. [Google Scholar] [CrossRef]
  50. Sinha, A.K.; Zinta, G.; AbdElgawad, H.; Asard, H.; Blust, R.; De Boeck, G. High environmental ammonia elicits differential oxidative stress and antioxidant responses in five different organs of a model estuarine teleost (Dicentrarchus labrax). Comp. Biochem. Physiol. C 2015, 174–175, 21–31. [Google Scholar] [CrossRef]
  51. Johannsson, O.E.; Giacomin, M.; Sadauskas-Henrique, H.; Campos, D.F.; Braz-Mota, S.; Heinrichs-Caldas, W.D.; Baptista, R.; Wood, C.M.; Almeida-Val, V.M.F.; Val, A.L. Does hypoxia or different rates of re-oxygenation after hypoxia induce an oxidative stress response in Cyphocharax abramoides (Kner 1858), a Characid fish of the Rio Negro? Comp. Biochem. Physiol. A 2018, 224, 53–67. [Google Scholar] [CrossRef]
  52. Liu, M.J.; Guo, H.Y.; Liu, B.; Zhu, K.C.; Guo, L.; Liu, B.S.; Zhang, N.; Yang, J.W.; Jiang, S.G.; Zhang, D.C. Gill oxidative damage caused by acute ammonia stress was reduced through the HIF-1α/NF-κb signaling pathway in golden pompano (Trachinotus ovatus). Ecotoxicol. Environ. Saf. 2021, 222, 112504. [Google Scholar] [CrossRef]
  53. Cheng, C.H.; Yang, F.F.; Ling, R.Z.; Liao, S.A.; Miao, Y.T.; Ye, C.X.; Wang, A.L. Effects of ammonia exposure on apoptosis, oxidative stress and immune response in pufferfish (Takifugu obscurus). Aquat. Toxicol. 2015, 164, 61–71. [Google Scholar] [CrossRef]
  54. Wang, S.; Meng, F.; Liu, Y.; Xia, S.; Wang, R. Exogenous inositol ameliorates the effects of acute ammonia toxicity on intestinal oxidative status, immune response, apoptosis, and tight junction barriers of great blue-spotted mudskippers (Boleophthalmus pectinirostris). Comp. Biochem. Physiol. C 2021, 240, 108911. [Google Scholar] [CrossRef] [PubMed]
  55. Kennel, K.B.; Greten, F.R. Immune cell-produced ROS and their impact on tumor growth and metastasis. Redox Biol. 2021, 42, 101891. [Google Scholar] [CrossRef] [PubMed]
  56. Ching, B.; Chew, S.F.; Wong, W.P.; Ip, Y.K. Environmental ammonia exposure induces oxidative stress in gills and brain of Boleophthalmus boddarti (mudskipper). Aquat. Toxicol. 2009, 95, 203–212. [Google Scholar] [CrossRef] [PubMed]
  57. Alak, G.; Ucar, A.; Parlak, V.; Yeltekin, A.Ç.; Taş, I.H.; Ölmez, D.; Kocaman, E.M.; Yılgın, M.; Atamanalp, M.; Yanık, T. Assessment of 8-hydroxy-2-deoxyguanosine activity, gene expression and antioxidant enzyme activity on rainbow trout (Oncorhynchus mykiss) tissues exposed to biopesticide. Comp. Biochem. Physiol. C 2017, 203, 51–58. [Google Scholar] [CrossRef]
  58. Oliva, M.; Gravato, C.; Guilhermino, L.; Galindo-Riaño, M.D.; Perales, J.A. EROD activity and cytochrome P4501A induction in liver and gills of Senegal sole Solea senegalensis from a polluted Huelva Estuary (SW Spain). Comp. Biochem. Physiol. C 2014, 166, 134–144. [Google Scholar] [CrossRef]
  59. Yang, C.; Lim, W.; Song, G. Mediation of oxidative stress toxicity induced by pyrethroid pesticides in fish. Comp. Biochem. Physiol. C 2020, 234, 108758. [Google Scholar] [CrossRef]
  60. Vega-López, A.; Galar-Martínez, M.; Jiménez-Orozco, F.A.; García-Latorre, E.; Domínguez-López, M.L. Gender related differences in the oxidative stress response to PCB exposure in an endangered goodeid fish (Girardinichthys viviparus). Comp. Biochem. Physiol. A 2007, 146, 672–678. [Google Scholar] [CrossRef]
  61. Slaninova, A.; Smutna, M.; Modra, H.; Svobodova, Z. A review: Oxidative stress in fish induced by pesticides. Neuroendocrinol. Let. 2009, 30, 2–12. [Google Scholar]
  62. Sun, Y.; Tang, L.; Liu, Y.; Hu, C.; Zhou, B.; Lam, P.K.S.; Lam, J.C.W.; Chen, L. Activation of aryl hydrocarbon receptor by dioxin directly shifts gut microbiota in zebrafish. Environ. Pollut. 2019, 255, 113357. [Google Scholar] [CrossRef]
  63. Choi, Y.; Jeon, J.; Kim, S.D. Identification of biotransformation products of organophosphate ester from various aquatic species by suspect and non-target screening approach. Water Res. 2021, 200, 117201. [Google Scholar] [CrossRef]
  64. Na, N.; Guo, H.; Zhang, S.; Li, Z.; Yin, L. In vitro and in vivo acute toxicity of fenpyroximate to flounder Paralichthys olivaceus and its gill cell line. Aquat. Toxicol. 2009, 92, 76–85. [Google Scholar] [CrossRef]
  65. Isik, I.; Celik, I. Acute effects of methylparathion and diazinon as inducers for oxidative stress on certain biomarkers in various tissues of rainbow trout (Oncorhynchus mykiss). Pest. Biochem. Physiol. 2008, 92, 38–42. [Google Scholar] [CrossRef]
  66. Nayak, S.; Dash, S.N.; Pati, S.S.; Priyadarshini, P.; Patnaik, L. Lipid peroxidation and antioxidant levels in Anabas testudineus (Bloch) under naphthalene (PAH) stress. Aquacult. Res. 2021, 52, 5739–5749. [Google Scholar] [CrossRef]
  67. Ji, Y.; Lu, G.H.; Wang, C.; Zhang, J. Biochemical responses of freshwater fish Carassius auratus to polycyclic aromatic hydrocarbons and pesticides. Water Sci. Eng. 2012, 5, 145–154. [Google Scholar] [CrossRef]
  68. Sayed, A.E.-D.H.; Soliman, H.A.M.; Idriss, S.K.; Abdel Ghaffar, S.K.; Hussein, A.A.A. Oxidative stress and immunopathological alterations of Clarias gariepinus exposed to monocyclic aromatic hydrocarbons (BTX). Water Air Soil Pollut. 2023, 234, 354. [Google Scholar] [CrossRef]
  69. Sadauskas-Henrique, H.; Duarte, R.M.; Gagnon, M.M.; Val, V.M.F.A. Validation of a suite of biomarkers of fish health in the tropical bioindicator species, tambaqui (Colossoma macropomum). Ecol. Indic. 2017, 73, 443–451. [Google Scholar] [CrossRef]
  70. Danion, M.; Floch, S.L.; Lamour, F.; Quentel, C. EROD activity and antioxidant defenses of sea bass (Dicentrarchus labrax) after an in vivo chronic hydrocarbon pollution followed by a post-exposure period. Environ. Sci. Pollut. Res. 2014, 21, 13769–13778. [Google Scholar] [CrossRef]
  71. Aguilar, L.; Dzul-Caamal, R.; Rendón-von Osten, J.; da Cruz, A.L. Effects of polycyclic aromatic hydrocarbons in Gambusia yucatana, an Endemic Fish from Yucatán Peninsula, Mexico. Polycycl. Aromat. Compd. 2022, 42, 907–924. [Google Scholar] [CrossRef]
  72. Barboza, L.G.A.; Vieira, L.R.; Branco, V.; Figueiredo, N.; Carvalho, F.; Carvalho, C.; Guilhermino, L. Microplastics cause neurotoxicity, oxidative damage and energy-related changes and interact with the bioaccumulation of mercury in the European seabass, Dicentrarchus labrax (Linnaeus, 1758). Aquat. Toxicol. 2018, 195, 49–57. [Google Scholar] [CrossRef]
  73. Gobi, N.; Vaseeharan, B.; Rekha, R.; Vijayakumar, S.; Faggio, C. Bioaccumulation, cytotoxicity and oxidative stress of the acute exposure selenium in Oreochromis mossambicus. Ecotoxicol. Environ. Saf. 2018, 162, 147–159. [Google Scholar] [CrossRef]
  74. Husak, V.V.; Mosiichuk, N.M.; Storey, J.M.; Storey, K.B.; Lushchak, V.I. Acute exposure to the penconazole-containing fungicide Topas partially augments antioxidant potential in goldfish tissues. Comp. Biochem. Physiol. C 2017, 193, 1–8. [Google Scholar] [CrossRef]
  75. Palanikumar, L.; Kumaraguru, A.K.; Ramakritinan, C.M.; Anand, M. Toxicity, biochemical and clastogenic response of chlorpyrifos and carbendazim in milkfish Chanos chanos. Int. J. Environ. Sci. Technol. 2014, 11, 765–774. [Google Scholar] [CrossRef]
  76. Han, Y.; Liu, T.; Wang, J.; Wang, J.; Zhang, C.; Zhu, L. Genotoxicity and oxidative stress induced by the fungicide azoxystrobin in zebrafish (Danio rerio) livers. Pest. Biochem. Physiol. 2016, 133, 13–19. [Google Scholar] [CrossRef] [PubMed]
  77. Li, Z.H.; Zlabek, V.; Velíšek, J.; Grabic, R.; Machova, J.; Kolařová, J.; Li, P.; Randak, T. Antioxidant responses and plasma biochemical characteristics in the freshwater rainbow trout, Oncorhynchus mykiss, after acute exposure to the fungicide propiconazole. Czech J. Anim. Sci. 2011, 56, 61–69. [Google Scholar] [CrossRef]
  78. Destro, A.L.F.; Silva, S.B.; Gregorio, K.P.; de Oliveira, J.M.; Lozi, A.A.; Zuanon, J.A.S.; Salaro, A.L.; da Matta, S.L.P.; Gonçalves, R.V.; Freitas, M.B. Effects of subchronic exposure to environmentally relevant concentrations of the herbicide atrazine in the Neotropical fish Astyanax altiparanae. Ecotoxicol. Environ. Saf. 2021, 208, 111601. [Google Scholar] [CrossRef] [PubMed]
  79. Verma, S.K.; Soni, R.; Gupta, P. Herbicide pretilachlor induces oxidative stress in freshwater fish Clarias batrachus. Biol. Bull. 2023, 50, 449–456. [Google Scholar] [CrossRef]
  80. El-Houseiny, W.; Anter, R.G.; Arisha, A.H.; Mansour, A.T.; Safhi, F.A.; Alwutayd, K.M.; Elshopakey, G.E.; Abd El-Hakim, Y.M.; Mohamed, E.M.M. Growth retardation, oxidative stress, immunosuppression, and inflammatory disturbances induced by herbicide exposure of catfish, Clarias gariepinus, and the alleviation effect of dietary wormwood, Artemisia cina. Fishes 2023, 8, 297. [Google Scholar] [CrossRef]
  81. Braz-Mota, S.; Sadauskas-Henrique, H.; Duarte, R.M.; Val, A.L.; Almeida-Val, V.M. Roundup® exposure promotes gills and liver impairments, DNA damage and inhibition of brain cholinergic activity in the Amazon teleost fish Colossoma macropomum. Chemosphere 2015, 135, 53–60. [Google Scholar] [CrossRef]
  82. Neglur, S.B.; David, M.; Sanakal, R.D. Studies on Fenaxoprop-P-Ethyl Induced Antioxidant Response in Cyprinus carpio L. Res. J. Agricult. Sci. 2021, 12, 2185–2192. [Google Scholar]
  83. Jin, Y.; Zhang, X.; Shu, L.; Chen, L.; Sun, L.; Qian, H.; Liu, W.; Fu, Z. Oxidative stress response and gene expression with atrazine exposure in adult female zebrafish (Danio rerio). Chemosphere 2010, 78, 846–852. [Google Scholar] [CrossRef]
  84. Ghaffar, A.; Hussain, R.; Ahmad, N.; Ghafoor, R.; Akram, M.W.; Khan, I.; Khan, A. Evaluation of hemato-biochemical, antioxidant enzymes as biochemical biomarkers and genotoxic potential of glyphosate in freshwater fish (Labeo rohita). Chem. Ecol. 2021, 37, 646–667. [Google Scholar] [CrossRef]
  85. Abdelmagid, A.D.; Said, A.M.; Gawad, E.A.A.; Shalaby, S.A.; Dawood, M.A. Propolis nanoparticles relieved the impacts of glyphosate-induced oxidative stress and immunosuppression in Nile tilapia. Environ. Sci. Pollut. Res. 2022, 29, 19778–19789. [Google Scholar] [CrossRef]
  86. Acar, Ü.; İnanan, B.E.; Navruz, F.Z.; Yılmaz, S. Alterations in blood parameters, DNA damage, oxidative stress and antioxidant enzymes and immune-related genes expression in Nile tilapia (Oreochromis niloticus) exposed to glyphosate-based herbicide. Comp. Biochem. Physiol. C 2021, 249, 109147. [Google Scholar] [CrossRef]
  87. Gomes, J.D.L.C.; do Amaral, A.M.B.; Storck, T.R.; Moraes, B.S.; Loro, V.L.; Clasen, B. Can vitamin C supplementation improve the antioxidant capacity of Rhamdia quelen fish exposed to atrazine? Arch. Environ. Contam. Toxicol. 2022, 82, 551–557. [Google Scholar] [CrossRef]
  88. Dinu, D.; Marinescu, D.; Munteanu, M.C.; Staicu, A.C.; Costache, M.; Dinischiotu, A. Modulatory effects of deltamethrin on antioxidant defense mechanisms and lipid peroxidation in Carassius auratus gibelio liver and intestine. Arch. Environ. Contam. Toxicol. 2010, 58, 757–764. [Google Scholar] [CrossRef]
  89. Bharti, S.; Rasool, F. Analysis of the biochemical and histopathological impact of a mild dose of commercial malathion on Channa punctatus (Bloch) fish. Toxicol. Rep. 2021, 8, 443–455. [Google Scholar] [CrossRef]
  90. Souza, S.S.; Machado, R.N.; da Costa, J.C.; Campos, D.F.; da Silva, G.S.; de Almeida-Val, V.M.F. Severe damages caused by Malathion exposure in Colossoma macropomum. Ecotoxicol. Environ. Saf. 2020, 205, 111340. [Google Scholar] [CrossRef]
  91. Yonar, S.M. Toxic effects of malathion in carp, Cyprinus carpio carpio: Protective role of lycopene. Ecotoxicol. Environ. Saf. 2013, 97, 223–229. [Google Scholar] [CrossRef]
  92. Yonar, S.M.; Ural, M.Ş.; Silici, S.; Yonar, M.E. Malathion-induced changes in the haematological profile, the immune response, and the oxidative/antioxidant status of Cyprinus carpio carpio: Protective role of propolis. Ecotoxicol. Environ. Saf. 2014, 102, 202–209. [Google Scholar] [CrossRef]
  93. Narra, M.R.; Rajender, K.; Reddy, R.R.; Murty, U.S.; Begum, G. Insecticides induced stress response and recuperation in fish: Biomarkers in blood and tissues related to oxidative damage. Chemosphere 2017, 168, 350–357. [Google Scholar] [CrossRef]
  94. Abdel Rahman, A.N.; Mansour, D.A.; Abd El-Rahman, G.I.; Elseddawy, N.M.; Zaglool, A.W.; Khamis, T.; Mahmoud, S.F.; Mahboub, H.H. Imidacloprid toxicity in Clarias gariepinus: Protective role of dietary Hyphaene thebaica against biochemical and histopathological disruption, oxidative stress, immune genes expressions, and Aeromonas sobria infection. Aquaculture 2022, 555, 738170. [Google Scholar] [CrossRef]
  95. Menezes, C.; Leitemperger, J.; Murussi, C.; de Souza Viera, M.; Adaime, M.B.; Zanella, R.; Loro, V.L. Effect of diphenyl diselenide diet supplementation on oxidative stress biomarkers in two species of freshwater fish exposed to the insecticide fipronil. Fish Physiol. Biochem. 2016, 42, 1357–1368. [Google Scholar] [CrossRef]
  96. Abdel Rahman, A.N.; Mohamed, A.A.R.; Mohammed, H.H.; Elseddawy, N.M.; Salem, G.A.; El-Ghareeb, W.R. The ameliorative role of geranium (Pelargonium graveolens) essential oil against hepato-renal toxicity, immunosuppression, and oxidative stress of profenofos in common carp, Cyprinus carpio (L.). Aquaculture 2020, 517, 734777. [Google Scholar] [CrossRef]
  97. Hemalatha, D.; Amala, A.; Rangasamy, B.; Nataraj, B.; Ramesh, M. Sublethal toxicity of quinalphos on oxidative stress and antioxidant responses in a freshwater fish Cyprinus carpio. Environ. Toxicol. 2016, 31, 1399–1406. [Google Scholar] [CrossRef]
  98. Bacchetta, C.; Rossi, A.; Ale, A.; Campana, M.; Parma, M.J.; Cazenave, J. Combined toxicological effects of pesticides: A fish multi-biomarker approach. Ecol. Indic. 2014, 36, 532–538. [Google Scholar] [CrossRef]
  99. Cui, J.; Zhang, Y.; Liu, L.; Zhang, Q.; Xu, S.; Guo, M.Y. Polystyrene microplastics induced inflammation with activating the TLR2 signal by excessive accumulation of ROS in hepatopancreas of carp (Cyprinus carpio). Ecotoxicol. Environ. Saf. 2023, 251, 114539. [Google Scholar] [CrossRef]
  100. Hamed, M.; Soliman, H.A.; Osman, A.G.; Sayed, A.E.D.H. Antioxidants and molecular damage in Nile Tilapia (Oreochromis niloticus) after exposure to microplastics. Environ. Sci. Pollut. Res. 2020, 27, 14581–14588. [Google Scholar] [CrossRef]
  101. Xing, X.; Li, M.; Yuan, L.; Song, M.; Ren, Q.; Shi, G.; Meng, F.; Wang, R. The protective effects of taurine on acute ammonia toxicity in grass carp Ctenopharynodon idellus. Fish Shellfish Immunol. 2016, 56, 517–522. [Google Scholar] [CrossRef]
  102. Faheem, M.; Lone, K.P. Oxidative stress and histopathologic biomarkers of exposure to bisphenol-A in the freshwater fish, Ctenopharyngodon idella. Braz. J. Pharm. Sci. 2017, 53, e17003. [Google Scholar] [CrossRef]
  103. Afzal, G.; Ahmad, H.I.; Hussain, R.; Jamal, A.; Kiran, S.; Hussain, T.; Saeed, S. Bisphenol A induces histopathological, hematobiochemical alterations, oxidative stress, and genotoxicity in common carp (Cyprinus carpio L.). Oxid. Med. Cell. Longev. 2022, 2022, 5450421. [Google Scholar] [CrossRef]
  104. Banaee, M.; Badr, A.A.; Multisanti, C.R.; Haghi, B.N.; Faggio, C. The toxicity effects of the individual and combined exposure of methyl tert-butyl ether (MTBE) and tire rubber powder (RP) on Nile tilapia fish (Oreochromis niloticus). Comp. Biochem. Physiol. C 2023, 274, 109759. [Google Scholar] [CrossRef]
  105. Zhang, M.; Li, M.; Wang, R.; Qian, Y. Effects of acute ammonia toxicity on oxidative stress, immune response and apoptosis of juvenile yellow catfish Pelteobagrus fulvidraco and the mitigation of exogenous taurine. Fish Shellfish Immunol. 2018, 79, 313–320. [Google Scholar] [CrossRef]
  106. Maradonna, F.; Nozzi, V.; Dalla Valle, L.; Traversi, I.; Gioacchini, G.; Benato, F.; Colletti, E.; Gallo, P.; Di Marco Pisciottano, I.; Mita, D.G.; et al. A developmental hepatotoxicity study of dietary bisphenol A in Sparus aurata juveniles. Comp. Biochem. Physiol. C 2014, 166, 1–13. [Google Scholar] [CrossRef]
  107. Revathy, V.; Chitra, K.C. Effects of diisononyl phthalate on the antioxidant status in gill, liver and muscle tissues of the fish, Oreochromis mossambicus. Asian J. Adv. Basic Sci. 2018, 6, 37–48. [Google Scholar]
  108. Bojarski, B.; Jakubiak, M.; Bień, M.; Batoryna, M.; Formicki, G.; Socha, M.; Drąg-Kozak, E.; Tombarkiewicz, B. Assessment of gentamicin effect on oxidoreductive balance and microstructure of trunk kidney in Prussian carp (Carassius gibelio). Ann. Wars. Univ. Life Sci. SGGW Anim. Sci. 2019, 58, 115–123. [Google Scholar] [CrossRef]
  109. Stengel, D.; Wahby, S.; Braunbeck, T. In search of an understandable set of endpoints for the routine monitoring of neurotoxicity in vertebrates: Sensory perception and nerve transmission in zebrafish (Danio rerio) embryos. Environ. Sci. Pollut. Res. 2018, 25, 4066–4084. [Google Scholar] [CrossRef]
  110. Carvalho, P.S.; Fonseca-Rodrigues, D.; Pacheco, M.; Almeida, A.; Pinto-Ribeiro, F.; Pereira, P. Comparative neurotoxicity of dietary methylmercury and waterborne inorganic mercury in fish: Evidence of optic tectum vulnerability through morphometric and histopathological assessments. Aquat. Toxicol. 2023, 261, 106557. [Google Scholar] [CrossRef]
  111. Kirici, M. Assessment of 8-hydroxy-2-deoxyguanosine activity, apoptosis, acetylcholinesterase, and antioxidant enzyme activity in Capoeta umbla brain exposed to chlorpyrifos. Int. J. Ocean. Hydrobiol. 2022, 51, 167–177. [Google Scholar] [CrossRef]
  112. Puga, S.; Pereira, P.; Pinto-Ribeiro, F.; O’Driscoll, N.J.; Mann, E.; Barata, M.; Pousão-Ferreira, P.; Canário, J.; Almeida, A.; Pacheco, M. Unveiling the neurotoxicity of methylmercury in fish (Diplodus sargus) through a regional morphometric analysis of brain and swimming behavior assessment. Aquat. Toxicol. 2016, 180, 320–333. [Google Scholar] [CrossRef]
  113. Sanchez-Aceves, L.M.; Pérez-Alvarez, I.; Belén Onofre-Camarena, D.; Gutiérrez-Noya, V.M.; Rosales-Pérez, K.E.; Orozco-Hernández, J.M.; Hernández-Navarro, M.D.; Islas Flores, H.; Gómez-Olivan, L.M. Prolonged exposure to the synthetic glucocorticoid dexamethasone induces brain damage via oxidative stress and apoptotic response in adult Danio rerio. Chemosphere 2024, 364, 143012. [Google Scholar] [CrossRef]
  114. Chang, X.; Shen, Y.; Yang, M.; Yun, L.; Liu, Z.; Feng, S.; Yang, G.; Meng, X.; Su, X. Antipsychotic drug-induced behavioral abnormalities in common carp: The potential involvement of the gut microbiota-brain axis. J. Hazard. Mat. 2024, 472, 134444. [Google Scholar] [CrossRef]
  115. Oger, M.J.L.; Vermeulen, O.; Lambert, J.; Madanu, T.L.; Kestemont, P.; Cornet, V. Down to size: Exploring the influence of plastic particle Dimensions on physiological and nervous responses in early-stage zebrafish. Environ. Pollut. 2024, 351, 124094. [Google Scholar] [CrossRef] [PubMed]
  116. Wan, T.; Au, D.W.-T.; Mo, J.; Chen, L.; Cheung, K.-M.; Kong, R.Y.-C.; Seemann, F. Assessment of parental benzo[a]pyrene exposure induced cross-generational neurotoxicity and changes in offspring sperm DNA methylome in medaka fish. Environ. Epigenetics 2022, 8, dvac013. [Google Scholar] [CrossRef] [PubMed]
  117. Morsy, G.M. Bioaccumulation and neurotoxicity of dithiopyridine herbicide in the brain of freshwater fish, Cyprinus carpio. Toxicol. Indust. Health 2015, 31, 1116–1127. [Google Scholar] [CrossRef]
  118. Bedrossiantz, J.; Bellot, M.; Dominguez-García, P.; Faria, M.; Prats, E.; Gómez-Canela, C.; López-Arnau, R.; Escubedo, E.; Raldúa, D. A zebrafish model of neurotoxicity by binge-like methamphetamine exposure. Front. Pharmacol. 2021, 12, 770319. [Google Scholar] [CrossRef]
  119. Vieira, M.; Nunes, B. Cholinesterases of marine fish: Characterization and sensitivity towards specific chemicals. Environ. Sci. Pollut. Res. 2021, 28, 48595–48609. [Google Scholar] [CrossRef]
  120. Martínez-Morcillo, S.; Pérez-López, M.; Míguez, M.P.; Valcárcel, Y.; Soler, F. Comparative study of esterase activities in different tissues of marine fish species Trachurus trachurus, Merluccius merluccius and Trisopterus luscus. Sci. Total Environ. 2019, 679, 12–22. [Google Scholar] [CrossRef]
  121. Sandahl, J.F.; Baldwin, D.H.; Jenkins, J.J.; Scholz, N.L. Comparative thresholds for acetylcholinesterase inhibition and behavioral impairment in Coho salmon exposed to chlorpyrifos. Environ. Toxicol. Chem. 2005, 24, 136–145. [Google Scholar] [CrossRef]
  122. Bonansea, R.I.; Wunderlin, D.A.; Amé, M.V. Behavioral swimming effects and acetylcholinesterase activity changes in Jenynsia multidentata exposed to chlorpyrifos and cypermethrin individually and in mixtures. Ecotoxicol. Environ. Saf. 2016, 129, 311–319. [Google Scholar] [CrossRef]
  123. Tõugu, V. Acetylcholinesterase: Mechanism of catalysis and inhibition. Curr. Med. Chem. Cent. Nerv. Syst. Agents 2001, 1, 155–170. [Google Scholar] [CrossRef]
  124. Zhou, Y.; Wang, S.; Zhang, Y. Catalytic reaction mechanism of acetylcholinesterase determined by Born-Oppenheimer ab initio QM/MM molecular dynamics simulations. J. Phys. Chem. B 2010, 114, 8817–8825. [Google Scholar] [CrossRef] [PubMed]
  125. Bigbee, J.W.; Sharma, K.W.; Gupta, J.J.; Dupree, J.L. Morphogenic role for acetylcholinesterase in axonal outgrowth during neural development. Environ. Health Perspect. 1999, 107 (Suppl. S1), 81–87. [Google Scholar] [CrossRef] [PubMed]
  126. Colovic, M.B.; Krstic, D.Z.; Lazarevic-Pasti, T.D.; Bondzic, A.M.; Vasic, V.M. Acetylcholinesterase inhibitors: Pharmacology and toxicology. Curr. Neuropharmacol. 2013, 11, 315–335. [Google Scholar] [CrossRef] [PubMed]
  127. De la Torre, F.R.; Ferrari, L.; Salibian, A. Freshwater pollution biomarker: Response of brain acetylcholinesterase activity in two fish species. Comp. Biochem. Physiol. C 2002, 131, 271–280. [Google Scholar] [CrossRef]
  128. Menéndez-Helman, R.J.; Ferreyroa, G.V.; dos Santos Afonso, M.; Salibián, A. Circannual rhythms of acetylcholinesterase (AChE) activity in the freshwater fish Cnesterodon decemmaculatus. Ecotoxicol. Environ. Saf. 2015, 111, 236–241. [Google Scholar] [CrossRef]
  129. Bernal-Rey, D.L.; Cantera, C.G.; dos Santos Afonso, M.; Menéndez-Helman, R.J. Seasonal variations in the dose-response relationship of acetylcholinesterase activity in freshwater fish exposed to chlorpyrifos and glyphosate. Ecotoxicol. Environ. Saf. 2020, 187, 109673. [Google Scholar] [CrossRef]
  130. Ellman, G.L.; Courtney, K.D.; Andres, V.J.R.; Featherstone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  131. Frasco, M.F.; Fournier, D.; Carvalho, F.; Guilhermino, L. Do metals inhibit acetylcholinesterase (AChE)? Implementation of assay conditions for the use of AChE activity as a biomarker of metal toxicity. Biomarkers 2005, 10, 360–375. [Google Scholar] [CrossRef]
  132. Xue, Y.-H.; Jia, T.; Yang, N.; Sun, Z.-X.; Xu, Z.-Y.; Wen, X.-L.; Feng, L.-S. Transcriptome alterations in zebrafish gill after exposure to different sizes of microplastics. J. Environ. Sci. Health A 2022, 57, 347–356. [Google Scholar] [CrossRef] [PubMed]
  133. Suman, A.; Mahapatra, A.; Gupta, P.; Ray, S.S.; Singh, R.K. Polystyrene microplastics modulated bdnf expression triggering neurotoxicity via apoptotic pathway in zebrafish embryos. Comp. Biochem. Physiol. C 2023, 271, 109699. [Google Scholar] [CrossRef] [PubMed]
  134. Martin-Folgar, R.; Torres-Ruiz, M.; de Alba, M.; Cañas-Portilla, A.I.; González, M.C.; Morales, M. Molecular effects of polystyrene nanoplastics toxicity in zebrafish embryos (Danio rerio). Chemosphere 2023, 312, 137077. [Google Scholar] [CrossRef] [PubMed]
  135. Usman, S.; Razis, A.F.A.; Shaari, K.; Amal, M.N.A.; Saad, M.Z.; Isa, N.M.; Nazarudin, M.F. Polystyrene microplastics exposure: An insight into multiple organ histological alterations, oxidative stress and neurotoxicity in Javanese medaka fish (Oryzias javanicus Bleeker, 1854). Int. J. Environ. Res. Pub. Health 2021, 18, 9449. [Google Scholar] [CrossRef]
  136. Jeyavani, J.; Sibiya, A.; Stalin, T.; Vigneshkumar, G.; Al-Ghanim, K.A.; Riaz, M.N.; Govindarajan, M.; Vaseeharan, B. Biochemical, genotoxic and histological implications of polypropylene microplastics on freshwater fish Oreochromis mossambicus: An aquatic eco-toxicological assessment. Toxics 2023, 11, 282. [Google Scholar] [CrossRef]
  137. Yang, J.; Karbalaei, S.; Hussein, S.M.; Ahmad, A.F.; Walker, T.R.; Salimi, K. Biochemical effects of polypropylene microplastics on red tilapia (Oreochromis niloticus) after individual and combined exposure with boron. Environ. Sci. Eur. 2023, 35, 71. [Google Scholar] [CrossRef]
  138. Santos, D.; Luzio, A.; Felix, L.; Cabecinha, E.; Bellas, J.; Monteiro, S.M. Microplastics and copper induce apoptosis, alter neurocircuits, and cause behavioral changes in zebrafish (Danio rerio) brain. Ecotoxicol. Environ. Saf. 2022, 242, 113926. [Google Scholar] [CrossRef]
  139. Kumar, N.; Bhushan, S.; Patole, P.B.; Gite, A. Multi-biomarker approach to assess chromium, pH and temperature toxicity in fish. Comp. Biochem. Physiol. C 2022, 254, 109264. [Google Scholar] [CrossRef]
  140. González Núñez, A.A.; Ferro, J.P.; Campos, L.B.; Eissa, B.L.; Mastrángelo, M.M.; Ferrari, L.; Ossana, N.A. Evaluation of the acute effects of arsenic on adults of the neotropical native fish Cnesterodon decemmaculatus using a set of biomarkers. Environ. Toxicol. Chem. 2022, 41, 1246–1259. [Google Scholar] [CrossRef]
  141. Xu, Y.; Wang, L.; Zhu, J.; Jiang, P.; Zhang, Z.; Li, L.; Wu, Q. Chromium induced neurotoxicity by altering metabolism in zebrafish larvae. Ecotoxicol. Environ. Saf. 2021, 228, 112983. [Google Scholar] [CrossRef]
  142. Alves, C.; Tamagno, W.A.; Vanin, A.P.; Pompermaier, A.; Gil Barcellos, L.J. Cannabis sativa based oils against aluminum induced neurotoxicity. Sci. Rep. 2023, 13, 813. [Google Scholar] [CrossRef]
  143. Henriques, M.C.; Carvalho, I.; Santos, C.; Herdeiro, M.T.; Fardilha, M.; Pavlaki, M.D.; Loureiro, S. Unveiling the molecular mechanisms and developmental consequences of mercury (Hg) toxicity in zebrafish embryo-larvae: A comprehensive approach. Neurotoxicol. Teratol. 2023, 100, 107302. [Google Scholar] [CrossRef] [PubMed]
  144. Xia, S.; Zhu, X.; Yan, Y.; Zhang, Y.; Chen, G.; Lei, D.; Wang, G. Developmental neurotoxicity of antimony (Sb) in the early life stages of zebrafish. Ecotoxicol. Environ. Saf. 2021, 218, 112308. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, Y.-J.; Chen, C.-Z.; Li, P.; Liu, L.; Chai, Y.; Li, Z.-H. Chronic toxic effects of waterborne mercury on silver carp (Hypophthalmichthys molitrix) larvae. Water 2022, 14, 1774. [Google Scholar] [CrossRef]
  146. Oliveira, V.M.; Assis, C.R.D.; Silva Costa, C.M.; Silva, R.P.F.; Santos, J.F.; Carvalho, L.B., Jr.; Bezerra, R.S. Aluminium sulfate exposure: A set of effects on hydrolases from brain, muscle and digestive tract of juvenile Nile tilapia (Oreochromis niloticus). Comp. Biochem. Physiol. C 2017, 191, 101–108. [Google Scholar] [CrossRef]
  147. Abegoda-Liyanage, C.S.; Pathiratne, A. Comparison of toxicity of nano and bulk titanium dioxide on Nile tilapia (Oreochromis niloticus): Acetylcholinesterase activity modulation and DNA damage. Bull. Environ. Contam. Toxicol. 2023, 110, 101. [Google Scholar] [CrossRef]
  148. Fan, S.; Yang, Y.; Sun, L.; Yu, B.; Dai, C.; Qu, Y. Different toxicity to liver and gill of zebrafish by selenium nanoparticles derived from bio/chemical methods. Environ. Sci. Pollut. Res. 2022, 29, 61512–61521. [Google Scholar] [CrossRef]
  149. Marinho, C.S.; Matias, M.V.F.; Toledo, E.K.M.; Smaniotto, S.; Ximenes-da-Silva, A.; Tonholo, J.; Santos, E.L.; Machado, S.S.; Zanta, C.L.P.S. Toxicity of silver nanoparticles on different tissues in adult Danio rerio. Fish Physiol. Biochem. 2021, 47, 239–249. [Google Scholar] [CrossRef]
  150. Jakubowska-Lehrmann, M.; Dąbrowska, A.; Białowąs, M.; Makaras, T.; Hallmann, A.; Urban-Malinga, B. The impact of various carbon nanomaterials on the morphological, behavioural, and biochemical parameters of rainbow trout in the early life stages. Aquat. Toxicol. 2023, 259, 106550. [Google Scholar] [CrossRef]
  151. Guo, D.; Luo, L.; Kong, Y.; Kuang, Z.; Wen, S.; Zhao, M.; Zhang, W.; Fan, J. Enantioselective neurotoxicity and oxidative stress effects of paclobutrazol in zebrafish (Danio rerio). Pest. Biochem. Physiol. 2022, 185, 105136. [Google Scholar] [CrossRef]
  152. Yang, H.; Chen, H.; Liang, X.; Zhao, Y.; Martyniuk, C.J. Molecular and behavioral responses of zebrafish embryos/larvae after sertraline exposure. Ecotoxicol. Environ. Saf. 2021, 208, 111700. [Google Scholar] [CrossRef]
  153. Vieira, R.; Venâncio, C.A.S.; Félix, L.M. Toxic effects of a mancozeb-containing commercial formulation at environmental relevant concentrations on zebrafish embryonic development. Environ. Sci. Pollut. Res. 2020, 27, 21174–21187. [Google Scholar] [CrossRef]
  154. Ames, J.; Stringini Severo, E.; Guilherme da Costa-Silva, D.; Rosso Storck, T.; Blank do Amaral, A.M.; Azambuja Miragem, A.; Broock Rosemberg, D.; Loro, V.L. Glyphosate-based herbicide (GBH) causes damage in embryo-larval stages of zebrafish (Danio rerio). Neurotoxicol. Teratol. 2023, 95, 107147. [Google Scholar] [CrossRef]
  155. Pompermaier, A.; Tamagno, W.A.; Alves, C.; Barcellos, L.J.G. Persistent and transgenerational effects of pesticide residues in zebrafish. Comp. Biochem. Physiol. C 2022, 262, 109461. [Google Scholar] [CrossRef] [PubMed]
  156. Liu, Y.; Guo, J.; Liu, W.; Yang, F.; Deng, Y.; Meng, Y.; Cheng, B.; Fu, J.; Zhang, J.; Liao, X.; et al. Effects of haloxyfop-p-methyl on the developmental toxicity, neurotoxicity, and immunotoxicity in zebrafish. Fish Shellfish Immunol. 2023, 132, 108466. [Google Scholar] [CrossRef] [PubMed]
  157. Hamed, H.S.; El-Sayed, Y.S. Antioxidant activities of Moringa oleifera leaf extract against pendimethalin-induced oxidative stress and genotoxicity in Nile tilapia, Oreochromis niloticus (L.). Fish Physiol. Biochem. A 2019, 45, 71–82. [Google Scholar] [CrossRef] [PubMed]
  158. Zhang, D.; Ding, W.; Liu, W.; Li, L.; Zhu, G.; Ma, J. Single and combined effects of chlorpyrifos and glyphosate on the brain of common carp: Based on biochemical and molecular perspective. Int. J. Mol. Sci. 2023, 24, 12934. [Google Scholar] [CrossRef]
  159. El-Nahhal, Y. Toxicity of some aquatic pollutants to fish. Environ. Monitor. Ass. 2018, 190, 449. [Google Scholar] [CrossRef]
  160. Viana, N.P.; Menezes da Silva, L.C.; Portruneli, N.; Soares, M.P.; Cardoso, I.L.; Bonansea, R.I.; Goulart, B.V.; Montagner, C.C.; Espíndola, E.L.G.; Wunderlin, D.A.; et al. Bioconcentration and toxicological impacts of fipronil and 2,4-D commercial formulations (single and in mixture) in the tropical fish, Danio rerio. Environ. Sci. Pollut. Res. 2022, 29, 11685–11698. [Google Scholar] [CrossRef]
  161. Lu, J.; Zhang, M.; Lu, L. Tissue metabolism, hematotoxicity, and hepatotoxicity of trichlorfon in Carassius auratus gibelio after a single oral administration. Front. Physiol. 2018, 9, 551. [Google Scholar] [CrossRef]
  162. Golombieski, J.I.; Marchesan, E.; Rabaioli Camargo, E.; Salbego, J.; Schmitt Baumart, J.; Loro, V.L.; de Oliveira Machado, S.L.; Zanella, R.; Baldisserotto, B. Acetylcholinesterase enzyme activity in carp brain and muscle after acute exposure to diafuran. Sci. Agric. 2008, 65, 340–345. [Google Scholar] [CrossRef]
  163. Pala, A.; Serdar, O.; Yonar, S.M.; Yonar, M.E. Ameliorative effect of Fennel (Foeniculum vulgare) essential oil on chlorpyrifos toxicity in Cyprinus carpio. Environ. Sci. Pollut. Res. 2021, 28, 890–897. [Google Scholar] [CrossRef] [PubMed]
  164. Chatterjee, A.; Bhattacharya, R.; Chatterjee, S.; Saha, N.C. λ cyhalothrin induced toxicity and potential attenuation of hematological, biochemical, enzymological and stress biomarkers in Cyprinus carpio L. at environmentally relevant concentrations: A multiple biomarker approach. Comp. Biochem. Physiol. C 2021, 250, 109164. [Google Scholar] [CrossRef] [PubMed]
  165. Duncan, W.P.; Silva Idalino, J.J.; da Silva, A.G.; Moda, R.F.; Menezes da Silva, H.C.; Matoso, D.A.; Silva Gomes, A.L. Acute toxicity of the pesticide trichlorfon and inhibition of acetylcholinesterase in Colossoma macropomum (Characiformes: Serrasalmidae). Aquacult. Int. 2020, 28, 815–830. [Google Scholar] [CrossRef]
  166. Singh, S.; Tiwari, R.K.; Pandey, R.S. Evaluation of acute toxicity of triazophos and deltamethrin and their inhibitory effect on AChE activity in Channa punctatus. Toxicol. Rep. 2018, 5, 85–89. [Google Scholar] [CrossRef]
  167. Mukherjee, D.; Saha, S.; Chukwuka, A.V.; Ghosh, B.; Dhara, K.; Saha, N.C.; Pal, P.; Faggio, C. Antioxidant enzyme activity and pathophysiological responses in the freshwater walking catfish, Clarias batrachus Linn under sub-chronic and chronic exposures to the neonicotinoid, Thiamethoxam®. Sci. Total Environ. 2022, 836, 155716. [Google Scholar] [CrossRef]
  168. Zhang, W.; Fan, R.; Luo, S.; Liu, Y.; Jin, Y.; Li, Y.; Li, B.; Chen, Y.; Jia, L.; Yuan, X. Combined effects of chlorpyrifos and cyfluthrin on neurobehavior and neurotransmitter levels in larval zebrafish. J. Appl. Toxicol. 2022, 42, 1662–1670. [Google Scholar] [CrossRef]
  169. Ran, L.; Yang, Y.; Zhou, X.; Jiang, X.; Hu, D.; Lu, P. The enantioselective toxicity and oxidative stress of dinotefuran on zebrafish (Danio rerio). Ecotoxicol. Environ. Saf. 2021, 226, 112809. [Google Scholar] [CrossRef]
  170. Guerra, L.J.; Blank do Amaral, A.M.; de Quadros, V.A.; da Luz Fiuza, T.; Rosemberg, D.B.; Prestes, O.D.; Zanella, R.; Clasen, B.; Loro, V.L. Biochemical and behavioral responses in zebrafish exposed to imidacloprid oxidative damage and antioxidant responses. Arch. Environ. Contam. Toxicol. 2021, 81, 255–264. [Google Scholar] [CrossRef]
  171. Zhang, J.-G.; Ma, D.-D.; Xiong, Q.; Qiu, S.-Q.; Huang, G.-Y.; Shi, W.-J.; Ying, G.G. Imidacloprid and thiamethoxam affect synaptic transmission in zebrafish. Ecotoxicol. Environ. Saf. 2021, 227, 112917. [Google Scholar] [CrossRef]
  172. Benli, P.P.; Celik, M. In vivo effects of neonicotinoid-sulfoximine insecticide sulfoxaflor on acetylcholinesterase activity in the tissues of zebrafish (Danio rerio). Toxics 2021, 9, 73. [Google Scholar] [CrossRef]
  173. Jablonski, C.A.; Brandão Pereira, T.C.; De Souza Teodoro, L.; Altenhofen, S.; Rübensam, G.; Bonan, C.D.; Bogo, M.R. Acute toxicity of methomyl commercial formulation induces morphological and behavioral changes in larval zebrafish (Danio rerio). Neurotoxicol. Teratol. 2022, 89, 107058. [Google Scholar] [CrossRef]
  174. Xiong, Y.; Wang, C.; Dong, M.; Li, M.; Hu, C.; Xu, X. Chlorphoxim induces neurotoxicity in zebrafish embryo through activation of oxidative stress. Environ. Toxicol. 2022, 38, 566–578. [Google Scholar] [CrossRef]
  175. Yu, T.; Xu, X.; Mao, H.; Han, X.; Liu, Y.; Zhang, H.; Lai, J.; Gu, J.; Xia, M.; Hu, C.; et al. Fenpropathrin exposure induces neurotoxicity in zebrafish embryos. Fish Physiol. Biochem. 2022, 48, 1539–1554. [Google Scholar] [CrossRef]
  176. Wang, S.; Han, X.; Yu, T.; Liu, Y.; Zhang, H.; Mao, H.; Hu, C.; Xu, X. Isoprocarb causes neurotoxicity of zebrafish embryos through oxidative stress-induced apoptosis. Ecotoxicol. Environ. Saf. 2022, 242, 113870. [Google Scholar] [CrossRef]
  177. Kavitha, P.; Rao, J.V. Toxic effects of chlorpyrifos on antioxidant enzymes and target enzyme acetylcholinesterase interaction in mosquito fish, Gambusia affinis. Environ. Toxicol. Pharmacol. 2008, 26, 192–198. [Google Scholar] [CrossRef]
  178. Touaylia, S.; Khazri, A.; Mezni, A.; Mezni, A. Assessment of biochemical biomarkers of mosquitofish (Gambusia affinis (Baird et Girard, 1853)) on exposure to the insecticide cypermethrin. Int. J. Environ. Stud. 2022, 81, 1122–1131. [Google Scholar] [CrossRef]
  179. Rouachdia, R.; Trea, F.; Tichati, L.; Ouali, K. Assessment of biochemical markers and behavior response of non-target freshwater teleost Gambusia affinis to carbofuran toxicity. Appl. Ecol. Environ. Res. 2023, 21, 545–559. [Google Scholar] [CrossRef]
  180. Mishra, A.K.; Gopesh, A.; Singh, K.P. Effects of chlorpyrifos toxicity on brain, pseudobranchial neurosecretory system and swimming performance of a catfish, Heteropneustes fossilis. Drug Chem. Toxicol. 2023, 47, 67–80. [Google Scholar] [CrossRef] [PubMed]
  181. Flynn, K.; Johnson, R.; Lothenbach, D.; Swintek, J.; Whiteman, F.; Etterson, M. The effects of combinations of limited ration and diazinon exposure on acetylcholinesterase activity, growth and reproduction in Oryzias latipes, the Japanese medaka. J. Appl. Toxicol. 2020, 40, 535–547. [Google Scholar] [CrossRef]
  182. Topal, A.; Şişecioğlu, M.; Atamanalp, M.; Işık, A.; Yılmaz, B. The in vitro and in vivo effects of chlorpyrifos on acetylcholinesterase activity of rainbow trout brain. J. Appl. Anim. Res. 2016, 44, 243–247. [Google Scholar] [CrossRef]
  183. Mehtabidah, A.; Majid, M.; Hussain, I.; Kali, S.; Naz, T.; Niazi, M.B.K.; Khan, M.R.A.; Zafar, M.I. Chlorpyrifos mediated oxidative damage and histopathological alterations in freshwater fish Oncorhynchus mykiss in Northern Pakistan. Aquacult. Res. 2020, 51, 4583–4594. [Google Scholar] [CrossRef]
  184. Muhammed, M.; Dogan, D. Toxicity and biochemical responses induced by phosmet in rainbow trout (Oncorhynchus mykiss). Toxicol. Res. 2021, 10, 983–991. [Google Scholar] [CrossRef] [PubMed]
  185. Weeks Santos, W.; Gonzalez, P.; Cormier, B.; Mazzella, N.; Moreira, A.; Clérandeau, C.; Morin, B.; Cachot, J. Subchronic exposure to environmental concentrations of chlorpyrifos affects swimming activity of rainbow trout larvae. Environ. Toxicol. Chem. 2021, 40, 3092–3102. [Google Scholar] [CrossRef] [PubMed]
  186. Amin, M.; Yousuf, M.; Attaullah, M.; Ahmad, N.; Nor Azra, M.; Lateef, M.; Dad Buneri, I.; Zekker, I.; El-Saber Batiha, G.; Aboelenin, S.M.; et al. Cholinesterase activity as a potential biomarker for neurotoxicity induced by pesticides in vivo exposed Oreochromis niloticus (Nile tilapia): Assessment tool for organophosphates and synthetic pyrethroids. Environ. Technol. 2023, 44, 2148–2156. [Google Scholar] [CrossRef] [PubMed]
  187. Hamed, H.S.; Ismal, S.M.; Faggio, C. Effect of allicin on antioxidant defense system, and immune response after carbofuran exposure in Nile tilapia, Oreochromis niloticus. Comp. Biochem. Physiol. C 2021, 240, 108919. [Google Scholar] [CrossRef]
  188. Santillan Deiú, A.; de la Torre, F.R.; Ondarza, P.M.; Miglioranza, K.S.B. Multibiomarker responses and bioaccumulation of fipronil in Prochilodus lineatus exposed to spiked sediments: Oxidative stress and antioxidant defenses. Pest. Biochem. Physiol. 2021, 177, 104876. [Google Scholar] [CrossRef]
  189. Baldissera, M.D.; Souza, C.F.; Zanell, R.; Prestes, O.D.; Meinhart, A.D.; Da Silva, A.S.; Baldisserotto, B. Behavioral impairment and neurotoxic responses of silver catfish Rhamdia quelen exposed to organophosphate pesticide trichlorfon: Protective effects of diet containing rutin. Com. Biochem. Physiol. C 2021, 239, 108871. [Google Scholar] [CrossRef]
  190. Sager, E.; Scarcia, P.; Marino, D.; Mac Loughlin, T.; Rossi, A.; de La Torre, F. Oxidative stress responses after exposure to triclosan sublethal concentrations: An integrated biomarker approach with a native (Corydoras paleatus) and a model fish species (Danio rerio). J. Toxicol. Environ. Health A 2022, 85, 291–306. [Google Scholar] [CrossRef]
  191. Orozco-Hernández, J.M.; Gómez-Oliván, L.M.; Elizalde-Velázquez, G.A.; Rosales-Pérez, K.E.; Cardoso-Vera, J.D.; Heredia-García, G.; Islas-Flores, H.; García-Medina, S.; Galar-Martínez, M. Fluoxetine-induced neurotoxicity at environmentally relevant concentrations in adult zebrafish Danio rerio. NeuroToxicology 2022, 90, 121–129. [Google Scholar] [CrossRef]
  192. Correia, D.; Domingues, I.; Faria, M.; Oliveira, M. Chronic effects of fluoxetine on Danio rerio: A biochemical and behavioral perspective. Appl. Sci. 2022, 12, 2256. [Google Scholar] [CrossRef]
  193. Rivero-Wendt, C.L.G.; Fernandes, L.G.; dos Santos, A.N.; Brito, I.L.; dos Santos Jaques, J.A.; dos Santos dos Anjos, E.; Fernandes, C.E. Effects of Chloramine T on zebrafish embryos malformations associated with cardiotoxicity and neurotoxicity. J. Toxicol. Environ. Health A 2023, 86, 372–381. [Google Scholar] [CrossRef] [PubMed]
  194. Chen, Y.; Liu, C.; Xiao, L.; Gao, G.; He, L.; Zhao, K.; Shang, X. 2, 5-dichloro-1, 4-benuinone exposure to zebrafish embryos/larvae causes neurodevelopmental toxicity. Ecotoxicol. Environ. Saf. 2022, 243, 114007. [Google Scholar] [CrossRef] [PubMed]
  195. Elizalde-Velázquez, G.A.; Gómez-Oliván, L.M.; García-Medina, S.; Rosales-Pérez, K.E.; Orozco-Hernández, J.M.; Islas-Flores, H.; Galar-Martínez, M.; Hernández-Navarro, M.D. Chronic exposure to realistic concentrations of metformin prompts a neurotoxic response in Danio rerio adults. Sci. Total Environ. 2022, 849, 157888. [Google Scholar] [CrossRef] [PubMed]
  196. Yang, H.; Gu, X.; Chen, H.; Zeng, Q.; Mao, Z.; Jin, M.; Li, H.; Ge, Y.; Zha, Y.; Martyniuk, C.J. Transcriptome profiling reveals toxicity mechanisms following sertraline exposure in the brain of juvenile zebrafish (Danio rerio). Ecotoxicol. Environ. Saf. 2022, 242, 113936. [Google Scholar] [CrossRef]
  197. Oliveira, A.C.; Fascineli, M.L.; Andrade, T.S.; Sousa-Moura, D.; Domingues, I.; Camargo, N.S.; Oliveira, R.; Grisolia, C.K.; Villacis, R.A.R. Exposure to tricyclic antidepressant nortriptyline affects early-life stages of zebrafish (Danio rerio). Ecotoxicol. Environ. Saf. 2021, 210, 111868. [Google Scholar] [CrossRef]
  198. Muniz, M.S.; Halbach, K.; Araruna, I.C.A.; Martins, R.X.; Seiwert, B.; Lechtenfeld, O.; Reemtsma, T.; Farias, D. Moxidectin toxicity to zebrafish embryos: Bioaccumulation and biomarker responses. Environ. Pollut. 2021, 283, 117096. [Google Scholar] [CrossRef]
  199. Tan, Y.; Liang, C.; Guo, Y.; Zou, H.; Guo, Y.; Ye, J.; Hou, L.; Wang, X. Thyroid endocrine disruption and neurotoxicity of gestodene in adult female mosquitofish (Gambusia affinis). Chemosphere 2023, 313, 37594. [Google Scholar] [CrossRef]
  200. Deepika, D.; Padmavathy, P.; Srinivasan, A.; Sugumar, G.; Jawahar, P. Short term effects of antimicrobial agent triclosan on Oreochromis mossambicus (Peters, 1852): Biochemical and genetic alterations. Ind. J. Anim. Res. 2023, 57, 788–794. [Google Scholar] [CrossRef]
  201. Rocha, C.S.; Puchale, R.Z.; Barcarolli, I.F. Avaliação toxicológica da progesterona em biomarcadores de tilápia do Nilo (Oreochromis niloticus). Rev. Bras. Meio Amb. 2022, 10, 26–40. [Google Scholar]
  202. Carvalho, L.A.S.J.; Oya-Silva, L.F.; Perussolo, M.C.; Guaita, G.O.; Brito, J.C.M.; Evans, A.A.; Prodocimo, M.M.; Cestari, M.M.; Braga, T.T.; Silva de Assis, H.C. Experimentally exposed toxic effects of long-term exposure to environmentally relevant concentrations of CIP in males and females of the silver catfish Rhamdia quelen. Chemosphere 2023, 336, 139216. [Google Scholar] [CrossRef]
  203. Nayak, S.; Patnaik, L. Acetylcholinesterase, as a potential biomarker of naphtalene toxicity in different tissues of a freshwater teleost, Anabas testudineus. Environ. Eng. Landsc. Manag. 2021, 29, 403–409. [Google Scholar] [CrossRef]
  204. Molayemraftar, T.; Peyghan, R.; Jalali, M.R.; Shahriari, A. Single and combined effects of ammonia and nitrite on common carp, Cyprinus carpio: Toxicity, hematological parameters, antioxidant defenses, acetylcholinesterase, and acid phosphatase activities. Aquaculture 2022, 548, 737676. [Google Scholar] [CrossRef]
  205. Iheanacho, S.C.; Ekpenyong, J.; Nwose, R.; Adeolu, A.I.; Offu, P.; Amadi-Eke, A.; Iheanacho, A.C.; Ogunji, J. Effects of burnt tire-ash on Na+/K+, Ca2+-ATPase, serum immunoglobulin and brain acetylcholinesterase activities in Clarias gariepinus (Burchell, 1822). Drug Chem. Toxicol. 2023, 46, 503–509. [Google Scholar] [CrossRef] [PubMed]
  206. Muthukumaravel, K.; Vasanthi, N.; Kanagavalli, V.; Santhanabharathi, B.; Pradhoshini, K.P.; Alam, L.; Faggio, C. Potential biomarker of phenol toxicity in freshwater fish C. mrigala: Serum cortisol, enzyme acetylcholine esterase and survival organ gill. Comp. Biochem. Physiol. C 2023, 263, 109492. [Google Scholar] [CrossRef] [PubMed]
  207. Li, P.; Li, Z.-H. Neurotoxicity and physiological stress in brain of zebrafish chronically exposed to tributyltin. J. Toxicol. Environ. Health A 2021, 84, 20–30. [Google Scholar] [CrossRef]
  208. Heredia-García, G.; Elizalde-Velázquez, G.A.; Gómez-Oliván, L.M.; Islas-Flores, H.; García-Medina, S.; Galar-Martínez, M.; Dublán-García, O. Realistic concentrations of Bisphenol-A trigger a neurotoxic response in the brain of zebrafish: Oxidative stress, behavioral impairment, acetylcholinesterase inhibition, and gene expression disruption. Chemosphere 2023, 330, 138729. [Google Scholar] [CrossRef]
  209. Rao, J.V.; Begum, G.; Pallela, R.; Usman, P.K.; Rao, R.N. Changes in behavior and brain acetylcholinesterase activity in mosquito fish, Gambusia affinis in response to the sub-lethal exposure to chlorpyrifos. Int. J. Environ. Res. Public Health 2023, 2, 478–483. [Google Scholar] [CrossRef]
  210. Thakkar, S.; Seetharaman, B.; Ramasamy, V. Impact of chronic sub-lethal methylparaben exposure on cardiac hypoxia and alterations in neuroendocrine factors in zebrafish model. Mol. Biol. Rep. 2022, 49, 331–340. [Google Scholar] [CrossRef]
  211. Agostini, J.F.; Zuehl Dal Toé, H.C.; Medeiros Vieira, K.; Baldin, S.L.; Fernandes Costa, N.L.; Uribe Cruz, C.; Longo, L.; Machado, M.M.; da Silveira, T.R.; Schuck, P.F.; et al. Cholinergic system and oxidative stress changes in the brain of a zebrafish model chronically exposed to ethanol. Neurotoxicol. Res. 2018, 33, 749–758. [Google Scholar] [CrossRef]
  212. Murugan, R.; Arokiyaraj, B.H.S.; Arockiaraj, J. Deacetyl epoxyazadiradione ameliorates BPA-induced neurotoxicity by mitigating ROS and inflammatory markers in N9 cells and zebrafish larvae. Comp. Biochem. Physiol. C 2023, 271, 109692. [Google Scholar] [CrossRef]
  213. Raja, G.L.; Subhashree, K.D.; Lite, C.; Santosh, W.; Barathi, S. Transient exposure of methylparaben to zebrafish (Danio rerio) embryos altered cortisol level, acetylcholinesterase activity and induced anxiety-like behaviour. Gen. Comp. Endocrinol. 2019, 279, 53–59. [Google Scholar] [CrossRef]
  214. Gayathri, M.; Sutha, J.; Mohanthi, S.; Ramesh, M.; Poopal, R.-K. Ecotoxicological evaluation of the UV-filter octocrylene (OC) in embryonic zebrafish (Danio rerio): Developmental, biochemical and cellular biomarkers. Comp. Biochem. Physiol. C 2023, 271, 109688. [Google Scholar] [CrossRef]
  215. Sandoval Gío, J.J.; Noreña Barroso, E.; Escalante Herrera, K.; Rodríguez Fuentes, G. Effect of benzophenone-3 to acetylcholinesterase and antioxidant system in zebrafish (Danio rerio) embryos. Bull. Environ. Contam. Toxicol. 2021, 107, 814–819. [Google Scholar] [CrossRef]
  216. Mariz, C.F.; de Melo Alves, M.K.; dos Santos, S.M.V.; Alves, R.N.; Carvalho, P.S.M. Lethal and sublethal toxicity of un-ionized ammonia to early life stages of Danio rerio. Zebrafish 2023, 20, 67–76. [Google Scholar] [CrossRef] [PubMed]
  217. Chen, X.; Guo, W.; Lei, L.; Guo, Y.; Yang, L.; Han, J.; Zhou, B. Bioconcentration and developmental neurotoxicity of novel brominated flame retardants, hexabromobenzene and pentabromobenzene in zebrafish. Environ. Pollut. 2021, 268, 115895. [Google Scholar] [CrossRef] [PubMed]
  218. Belhamra, R.; Tichati, L.; Trea, F.; Ouali, K. Effect of subacute treatment with bisphenol A on oxidative stress biomarkers and lipid peroxidation in Gambusia affinis mosquitofish. Toxicol. Environ. Health Sci. 2023, 15, 6. [Google Scholar] [CrossRef]
  219. Pérez-Iglesias, J.M.; González, P.; Calderón, M.R.; Natale, G.S.; Almeida, C.S. Comprehensive evaluation of the toxicity of the flame retardant (decabromodiphenyl ether) in a bioindicator fish (Gambusia affinis). Environ. Sci. Pollut. Res. 2022, 29, 50845–50855. [Google Scholar] [CrossRef]
  220. Gopi, N.; Iswarya, A.; Vijayakumar, S.; Jayanthi, S.; Nord, A.S.M.; Velusamye, P.; Vaseeharan, B. Protective effects of dietary supplementation of probiotic Bacillus licheniformis Dahb1 against ammonia induced immunotoxicity and oxidative stress in Oreochromis mossambicus. Comp. Biochem. Physiol. C 2022, 259, 109379. [Google Scholar] [CrossRef]
  221. Nirmala, G.N.; Sharma, A.; Ragunathan, V. Antagonistic effect of dichloromethane on Oreochromis mossambicus and immune stimulation activity of Aloe vera. Front. Environ. Sci. 2022, 10, 913065. [Google Scholar] [CrossRef]
  222. Lin, H.; Jia, Y.; Han, F.; Xia, C.; Zhao, Q.; Zhang, J.; Li, E. Toxic effects of waterborne benzylparaben on the growth, antioxidant capacity and lipid metabolism of Nile tilapia (Oreochromis niloticus). Aquat. Toxicol. 2022, 248, 106197. [Google Scholar] [CrossRef]
  223. Passos, L.S.; Gomes, L.C.; Pereira, T.M.; Sadauskas-Henrique, H.; Dal Pont, G.; Ostrensky, A.; Pinto, E. Response of Oreochromis niloticus (Teleostei: Cichlidae) exposed to a guanitoxin-producing cyanobacterial strain using multiple biomarkers. Sci. Total Environ. 2022, 835, 155471. [Google Scholar] [CrossRef]
  224. Fulton, M.H.; Key, P.B. Acetylcholinesterase inhibition in estuarine fish and invertebrates as an indicator of organophosphorus insecticide exposure and effects. Environ. Toxicol. Chem. 2001, 20, 37–45. [Google Scholar] [CrossRef]
  225. Arora, S.; Kumar, A. Mixture toxicity assessment of selected insecticides to silver perch fingerling, Bidyanus bidyanus. Ecotoxicol. Environ. Saf. 2021, 226, 112790. [Google Scholar] [CrossRef]
  226. Patisaul, H.B.; Behl, M.; Birnbaum, L.S.; Blum, A.; Diamond, M.L.; Fernández, S.R.; Hogberg, H.T.; Kwiatkowski, C.F.; Page, J.D.; Soehl, A.; et al. Beyond cholinesterase inhibition: Developmental neurotoxicity of organophosphate ester flame retardants and plasticizers. Environ. Health Perspect. 2021, 129, 105001. [Google Scholar] [CrossRef]
  227. Sato, R.; Mitani, K.; Matsumoto, T.; Takahashi, S.; Yamada, R.-H.; Kera, Y. Effects of insecticides in vitro on acetylcholinesterase purified from body muscle of Koi carp (Cyprinus carpio). Jpn. J. Environ. Toxicol. 2007, 10, 31–38. [Google Scholar]
  228. Nordin, N.; Abdulla, R.; Ahmad, S.A.; Sabullah, M.K. Acetylcholinesterase (AChE) of Diodon hystrix brain as an alternative biomolecule in heavy metals biosensing. J. Appl. Sci. Eng. 2022, 25, 573–580. [Google Scholar] [CrossRef]
  229. Mladenovic, M.B.; Arsic, B.B.; Ragno, R.; Stankovic, N.; Mihovic, N.; Regan, A.; Milicevic, J.S.; Micic, R. The targeted pesticides as acetylcholinesterase inhibitors: Comprehensive cross-organism molecular modelling studies performed to anticipate the pharmacology of harmfulness to humans in vitro. Molecules 2018, 23, 2192. [Google Scholar] [CrossRef] [PubMed]
  230. Olivares-Rubio, H.F.; Espinosa-Aguirre, J.J. Acetylcholinesterase activity in fish species exposed to crude oil hydrocarbons: A review and new perspectives. Chemosphere 2021, 264, 128401. [Google Scholar] [CrossRef] [PubMed]
  231. Fakhereddin, T.; Doğan, D. Pro-oxidant potency of clothianidin in rainbow trout. Arch. Ind. Hyg. Toxicol. 2021, 72, 164–172. [Google Scholar] [CrossRef] [PubMed]
  232. Guimaraes, A.T.B.; Silva de Assisb, H.C.; Boeger, W. The effect of trichlorfon on acetylcholinesterase activity and histopathology of cultivated fish Oreochromis niloticus. Ecotoxicol. Environ. Saf. 2007, 68, 57–62. [Google Scholar] [CrossRef]
  233. Tilton, F.A.; Bammler, T.K.; Gallagher, E.P. Swimming impairment and acetylcholinesterase inhibition in zebrafish exposed to copper or chlorpyrifos separately, or as mixtures. Comp. Biochem. Physiol. C 2011, 153, 9–16. [Google Scholar] [CrossRef]
  234. Ullah, S.; Li, Z.; Hassan, S.; Ahmad, S.; Guo, X.; Wanghe, K.; Nabi, G. Heavy metals bioaccumulation and subsequent multiple biomarkers based appraisal of toxicity in the critically endangered Tor putitora. Ecotoxicol. Environ. Saf. 2021, 228, 113032. [Google Scholar] [CrossRef]
  235. dos Santos Teixeira, J.M.; da Silva Lima, V.; de Moura, F.R.; da Costa Marisco, P.; Sinhorin, A.P.; Gindri Sinhorin, V.D. Acute toxicity and effects of Roundup Original® on pintado da Amazônia. Environ. Sci. Pollut. Res. 2018, 25, 25383–25389. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, T.; Yang, M.; Pan, H.; Li, S.; Ren, B.; Ren, Z.; Xing, N.; Qi, L.; Ren, Q.; Xu, S.; et al. Does time difference of the acetylcholinesterase (AChE) inhibition in different tissues exist? A case study of zebra fish (Danio rerio) exposed to cadmium chloride and deltamethrin. Chemosphere 2017, 168, 908–916. [Google Scholar] [CrossRef] [PubMed]
  237. Issac, P.K.; Guru, A.; Velayutham, M.; Pachaiappan, R.; Arasu, M.V.; Al-Dhabi, N.A.; Choi, K.C.; Harikrishnan, R.; Arockiaraj, J. Oxidative stress induced antioxidant and neurotoxicity demonstrated in vivo zebrafish embryo or larval model and their normalization due to morin showing therapeutic implications. Life Sci. 2021, 283, 119864. [Google Scholar] [CrossRef] [PubMed]
  238. Jha, R.; Rizvi, S.I. Age-dependent decline in erythrocyte acetylcholinesterase activity: Correlation with oxidative stress. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 2009, 153, 195–198. [Google Scholar] [CrossRef]
  239. Liu, H.; Wu, J.; Yao, J.-Y.; Wang, H.; Li, S.-T. The role of oxidative stress in decreased acetylcholinesterase activity at the neuromuscular junction of the diaphragm during sepsis. Oxidative Med. Cell. Longev. 2017, 2017, 9718615. [Google Scholar] [CrossRef]
  240. Melo, J.B.; Agostinho, P.; Oliveira, C.R. Involvement of oxidative stress in the enhancement of acetylcholinesterase activity induced by amyloid beta-peptide. Neurosci. Res. 2003, 45, 117–127. [Google Scholar] [CrossRef]
  241. Snega Priya, P.; Pratiksha Nandhini, P.; Vaishnavi, S.; Pavithra, V.; Almutairi, M.H.; Almutairi, B.O.; Arokiyaraj, S.; Pachaiappan, R.; Arockiaraj, J. Rhodamine B, an organic environmental pollutant induces reproductive toxicity in parental and teratogenicity in F1 generation in vivo. Comp. Biochem. Physiol. C 2024, 280, 109898. [Google Scholar] [CrossRef]
  242. Wu, Q.; Yan, W.; Cheng, H.; Liu, C.; Hung, T.-C.; Guo, X.; Li, G. Parental transfer of microcystin-LR induced transgenerational effects of developmental neurotoxicity in zebrafish offspring. Environ. Pollut. 2017, 231, 471–478. [Google Scholar] [CrossRef]
  243. Schmitt, C.; Peterson, E.; Willis, A.; Kumar, N.; McManus, M.; Subbiah, S.; Crago, J. Transgenerational effects of developmental exposure to chlorpyrifos-oxon in zebrafish (Danio rerio). Toxicol. Appl. Pharmacol. 2020, 408, 115275. [Google Scholar] [CrossRef] [PubMed]
  244. Brander, S.M.; White, J.W.; De Courten, B.M.; Major, K.; Hutton, S.J.; Connon, R.E.; Mehinto, A. Accounting for transgenerational effects of toxicant exposure in population models alters the predicted long-term population status. Environ. Epigenetics 2022, 8, dvac023. [Google Scholar] [CrossRef] [PubMed]
  245. Schmidt, M.; Windisch, H.S.; Ludwichowski, K.-U.; Seegert, S.L.L.; Pörtner, H.-O.; Storch, D.; Bock, C. Differences in neurochemical profiles of two gadid species under ocean warming and acidification. Front. Zool. 2017, 14, 49. [Google Scholar] [CrossRef]
  246. Regan, M.D.; Turko, A.J.; Heras, J.; Andersen, M.K.; Lefevre, S.; Wang, T.; Bayley, M.; Brauner, C.J.; Huong, D.T.T.; Phuong, N.T.; et al. Ambient CO2, fish behaviour and altered GABAergic neurotransmission: Exploring the mechanism of CO2-altered behaviour by taking a hypercapnia dweller down to low CO2 levels. J. Exp. Biol. 2016, 219, 109–118. [Google Scholar] [CrossRef] [PubMed]
  247. Santana, M.S.; Sandrini-Neto, L.; Di Domenico, M.; Mela Prodocimo, M. Pesticide effects on fish cholinesterase variability and mean activity: A meta-analytic review. Sci. Total Environ. 2021, 757, 143829. [Google Scholar] [CrossRef]
  248. Solé, M.; Vega, S.; Varo, I. Characterization of type “B” esterases and hepatic CYP450 isoenzimes in Senegalese sole for their further application in monitoring studies. Ecotoxicol. Environ. Saf. 2012, 78, 72–79. [Google Scholar] [CrossRef]
  249. Ortiz-Delgado, J.B.; Funes, V.; Albendín, G.; Scala, E.; Sarasquete, C. Toxicity of malathion during Senegalese sole, Solea senegalensis larval development and metamorphosis: Histopathological disorders and effects on type B esterases and CYP1A enzymatic systems. Environ. Toxicol. 2021, 36, 1894–1910. [Google Scholar] [CrossRef]
Table 1. Products of oxidative damage of biological molecules that are used as biomarkers of oxidative stress in fish.
Table 1. Products of oxidative damage of biological molecules that are used as biomarkers of oxidative stress in fish.
BiomarkerProcessReferences
lipid peroxidesoxidative damage to lipidsTanaka et al. [15]
hydroxyl lipidsoxidative damage to lipidsBaker et al. [16]; Tanaka et al. [15]
malondialdehydeoxidative damage to lipidsLiu et al. [17]; Paital et al. [14]; Sánchez-Nuño et al. [18]; Safari Asl et al. [19]; Tan et al. [20]
4-hydroxynonenaloxidative damage to lipidsTopal et al. [21]; Sánchez-Nuño et al. [18]
lipofuscinoxidative damage to lipidsPulster et al. [22]; Raibeemol and Chitra [23]; Zhong et al. [24]; Gómez Manrique et al. [25]; Ottinger et al. [26]
8-hydroxy-2′-deoxyguanosine,
8-oxo-7,8-dihydro-2′-deoxyguanosine
oxidative damage to DNA guanosineMahboub et al. [27]; Jia et al. [28]; Mohamed et al. [29,30]; Topal et al. [31]
8-hydroxyguanineoxidative damage to DNA guanineno data found
5,6-dihydroxy-5,6-dihydrothymineoxidative damage to DNA thymineno data found
advanced oxidation protein productsoxidation of proteinsSánchez-Nuño et al. [18]
protein carbonylsoxidation of proteinsAlmeida et al. [32]; Zhang et al. [33]; Blank do Amaral et al. [34]; Iturburu et al. [35]; Feng et al. [36]
Hydroxyleucineoxidation of protein leucineno data found
Nitrotyrosinenitration of protein tyrosineShahab et al. [37]; Lee et al. [38]
Table 2. The effects of toxic agents on oxidative stress parameters commonly determined in fish toxicology. The original values are given in parentheses and expressed in the following units: ° mg/L * µg/L ** µmol/g (injection) ~ µL/L ^ µmol/kg (injection) ^^ mg/kg # ng/L ## ppm ### mg/kg (injection).
Table 2. The effects of toxic agents on oxidative stress parameters commonly determined in fish toxicology. The original values are given in parentheses and expressed in the following units: ° mg/L * µg/L ** µmol/g (injection) ~ µL/L ^ µmol/kg (injection) ^^ mg/kg # ng/L ## ppm ### mg/kg (injection).
Fish SpeciesToxic AgentExp. dur. [d]Concentration [µmol]No ChangeDecreaseIncreaseReference
AhAnabas testudineusnaphthalene332.789 (4.2 °)
34.329 (4.4 °)
35.890 (4.6 °)
37.450 (4.8 °)
39.010 (5 °)
CAT, GPx, GSH, TBARS
TBARS



GPx, GSH
GPx, GSH
GPx, GSH
GPx, GSH

CAT, TBARS
CAT, TBARS
CAT, TBARS
CAT, TBARS
Nayak et al. [66]
AhCarassius auratusbenzo(k)fluoranthene 1–150.399 (0.1 ^^)
0.240 (0.06 ^^)
1.189 (0.3 ^^)
5.870 (1.48 ^^)
29.367 (7.41 ^^)
GSH, GST, TBARS
GST, TBARS
TBARS
TBARS

GSH
GSH
GSH
GSH


GST
GST, TBARS
GST, TBARS
Ji et al. [67]
AhClarias gariepinusbenzene 309.76 × 10−6 (0.762 #)GSTSOD, TASMDASayed et al. [68]
AhClarias gariepinustoluene300.00033 (26.614 #)GST, SOD, TAS, MDASayed et al. [68]
AhClarias gariepinusxylene300.0008 (89.403 #)GST, SOD, TAS, MDASayed et al. [68]
AhColossoma macropomumbenzo[a]pyrene4(1 ^)

(10 ^)
(100 ^)
(1000 ^)
SOD, CAT, GPx, GST, LPO
SOD, CAT, GPx, GST
SOD, CAT, GPx, GST
CAT, GPx






LPO
SOD, LPO
SOD, GST, LPO
Sadauskas-Henrique et al. [69]
AhDicentrarchus labraxpolycyclic aromatic hydrocarbons21(835  ±  52 #)GSH, SOD, GPx, CATSODGSHDanion et al. [70]
AhGambusia yucatanapolycyclic aromatic hydrocarbons4(4.37 *)
(8.73 *)
(17.46 *)
(34.95 *)
SOD, GPx
SOD, GPx,
SOD, GPx
GPx
TBARS, CAT
TBARS, CAT
TBARS, CAT
TBARS, CAT
GST
GST
GST
GST, SOD
Aguilar et al. [71]
EDicentrarchus labraxmercury (as HgCl2)40.037 (0.010 °)
0.059 (0.016 °)


LPO
LPO
Barboza et al. [72]
EOreochromis mossambicusselenium (as Na2SeO3)40.029 (5 *)
0.058 (10 *)
0.145 (25 *)
0.289 (50 *)
0.578 (100 *)








LPO, GSH
LPO, GSH
LPO, GSH
LPO, GSH
LPO, GSH
Gobi et al. [73]
FCarassius auratusTopas 100 EC45.278 (1.5 °)

52.783 (15 °)
87.972 (25 °)
LOOH, SOD, CAT, GPx, GST, GR
LOOH, CAT, GST, GR
LOOH, GR


GST
GST


SOD, GPx, GST
SOD, CAT, GPx, GST, GR
Husak et al. [74]
FChanos chanoscarbendazim40.015 (2.85 *)
0.029 (5.45 *)
0.057 (10.97 *)
0.105 (20.17 *)
0.237 (45.31 *)
LPO, CAT, GST
LPO



CAT, GST
CAT, GST
CAT, GST
CAT, GST


LPO
LPO
LPO
Palanikumar et al. [75]
FDaniorerioazoxystrobin7–280.002 (1 *)
0.025 (10 *)

0.248 (100 *)
GST, MDA
CAT, GST, MDA

CAT, GST
SOD, CAT
SOD

SOD
ROS, SOD, GST, MDA
ROS, SOD, CAT, GST, MDA
ROS, SOD, CAT, GST, MDA
Han et al. [76]
FOncorhynchus mykisspropiconazole414.727 (5.04 °)TBARS, CAT, GRSOD, CAT, GPx, GRSOD, GPx, TBARSLi et al. [77]
HAstyanax altiparanaeatrazine (as Atrazina Atanor®)300.002 (0.5 *)
0.005 (1 *)
0.009 (2 *)
0.046 (10 *)
CAT, SOD, MDA
CAT, SOD, MDA
CAT, SOD, MDA
SOD, MDA
GST
GST
GST
GST



CAT
Destro et al. [78]
HClarias batrachuspretilachlor
(50% EC)
30–600.930 (0.29 °)
1.251 (0.39 °)
1.860 (0.58 °)
GR
GR
GR


TBARS, SOD, CAT
TBARS, SOD, CAT
TBARS, SOD, CAT
Verma et al. [79]
HClarias gariepinusoxyfluorfen603.207 (1.16 °)SOD, CAT, GPx, GSHMDAEl-Houseiny et al. [80]
HColossoma macropomumglyphosate (as Roundup)459.146 (10°)
88.719 (15 °)
SOD, GPx
SOD
GST
GST

GPx
Braz-Mota et al. [81]
HCyprinus carpiofenaxoprop-P-ethyl15–300.104 (37.5 *)GPx, GR, GSTMDANeglur et al. [82]
HDanio rerioatrazine140.005 (1 *)
0.046 (10 *)
0.464 (100 *)
4.636 (1000 *)
MDA, SOD, CAT
MDA
CAT
GSH
GSH
GSH
GSH

SOD, CAT
SOD, MDA
SOD, CAT, MDA
Jin et al. [83]
HLabeo rohitaglyphosate4–122.957 (0.5 °)

3.549 (0.6 °)

4.140 (0.7 °)
4.732 (0.8 °)
ROS, TBARS, GSH, CAT, SOD, POD
ROS, TBAR, GSH, CAT, SOD, POD





GSH, CAT, SOD, POD
GSH, CAT, SOD, POD




ROS, TBARS
ROS, TBARS
Ghaffar et al. [84]
HOreochromis
niloticus
glyphosate (as Roundup)14–283.549 (0.6 °)GSHMDAAbdelmagid et al. [85]
HOreochromis niloticusglyphosate-based herbicide14(5 °)
(10 °)
(20 °)
(30 °)
(40 °)






TBARS
TBARS
TBARS

CAT
CAT
CAT
CAT
Acar et al. [86]
HRhamdia quelenatrazine40.046 (10 *)GPx, MDACAT Gomes et al. [87]
ICarassius gibeliodeltamethrin1–140.004 (2 *)GPXSOD, CAT, GPX, GST, GR, GSH LPO, CAT, GPX, GST, GR, GSH Dinu et al. [88]
IChanos chanoschlorpyrifos (as Trickel)40.004 (1.38 *)
0.006 (2.15 *)
0.013 (4.53 *)
0.026 (9.27 *)
0.054 (18.97 *)
LPO, CAT, GST
LPO



CAT, GST
CAT, GST
CAT, GST
CAT, GST


LPO
LPO
LPO
Palanikumar et al. [75]
IChanna punctatusmalathion (commercial grade)4–121.211 (0.4 °)SOD, CAT, LPOBharti and Rasool [89]
IColossoma macropomummalathion (emulsion)422.097 (7.30 °)SOD, GPx, LPO GST, CAT, SOD, GPxSouza et al. [90]
ICyprinus carpiomalathion (commercial formulation)141.514 (0.5 °)
3.027 (1 °)

GPx, GSH
GPx, GSH
MDA, SOD, CAT
MDA, SOD, CAT
Yonar [91]
ICyprinus carpiomalathion (commercial formulation)101.514 (0.5 °)
3.027 (1 °)

GPx, GSH
GPx, GSH
MDA, SOD, CAT
MDA, SOD, CAT
Yonar et al. [92]
IClarias batrachuschlorpyrifos
(20% EC)
154.706 (1.65 °) SOD, CAT, GSH, LPO, GST, GPXSOD, CAT, GSH, LPO, GST, GPXNarra et al. [93]
IClarias batrachusmonocrotophos
(36% EC)
159.590 (2.14 °) SOD, CAT, GSH, LPO, GST, GPXSOD, CAT, GSH, LPO, GST, GPXNarra et al. [93]
IClarias gariepinusimidacloprid (as Sunclopride 35% SC)600.008 (2.03 *)SOD, GPxMDAAbdel Rahman et al. [94]
ICyprinus carpiofipronil (as Standak-BASF)80.001 (0.65 *)MDA, GSTMDA, GSTMenezes et al. [95]
ICyprinus carpioprofenofos600.013 (4.74 *)SOD, CAT, GSHMDAAbdel Rahman et al. [96]
ICyprinus carpioquinalphos
(25% EC)
200.0009 (1.09 ~)
0.0018 (2.18 ~)


SOD, CAT, MDA, GST
SOD, CAT, MDA, GST
Hemalatha et al. [97]
IPiaractus mesopotamicusendosulfan (as Zebra Ciagro®)40.003 (1.1 *)GPx, CAT, GST, GRBacchetta et al. [98]
IPiaractus mesopotamicuslambda-cyhalothrin (Cilambda®)40.016 (0.7 *)GR, GSTCAT, GPx, GR, GSTBacchetta et al. [98]
IRhamdia quelenfipronil (as Standak-BASF)80.002 (0.65 *)MDA, GSTGSTMDAMenezes et al. [95]
MCyprinus carpiopolystyrene
microplastics
21(1000 #)CAT, SOD, GPXROS, MDACui et al. [99]
MDicentrarchus labraxfluorescence red polymer microspheres4(0.26 °)
(0.69 °)


LPO
LPO
Barboza et al. [72]
MOreochromis niloticusmicroplastics (>100 nm in size)15(1 °)
(10 °)
(100 °)


TAC
TAC
TAC
SOD, CAT, TPX
SOD, CAT, TPX,
SOD, CAT, TPX
Hamed et al. [100]
OCtenopharyngodon idellaammonium acetaten/a9000 (9 **)CATSOD, GSH, MDAXing et al. [101]
OCtenopharyngodon idellabisphenol A140.014 (3.2 ##)CAT, GSHTBARS, GSTFaheem and Lone [102]
OCyprinus carpiobisphenol A3013.141 (3.0 °)
19.712 (4.5 °)
26.282 (6 °)
TBARS, ROS


SOD, CAT, POD, GSH
SOD, CAT, POD, GSH

TBARS, ROS
TBARS, ROS
Afzal et al. [103]
OOreochromis
niloticus
methyl tert-butyl ether270.028 (2.5 ~)
0.057 (5 ~)

TAO
GR, TAO
SOD, CAT, GPx, MDA
SOD, CAT, GPx, MDA
Banaee et al. [104]
OPelteobagrus fulvidracoammonium acetaten/a8000 (8 **)GPXTAOSOD, CAT, MDAZhang et al. [105]
OSparus auratabisphenol A2121.902 (5 ^^)
219.020 (50 ^^)
GST
CAT, GST

CAT
Maradonna et al. [106]
OOreochromis mossambicusdiisononyl phthalate1–40.717 (300 ##)SOD, CAT, GR, GPxSOD, CAT, GPx, GRSOD, CAT, GR, MDARevathy and Chitra [107]
PdCarassius gibeliogentamicin33.595 (5 ###)GSH, SOD, CAT, GPxBojarski et al. [108]
Exp. dur.—exposure duration; d—days; n/a—not applicable; CAT—catalase; GPx—glutathione peroxidase; GR—glutathione reductase; GST—glutathione S-transferase; GSH—reduced glutathione; LPO—lipid peroxidation; MDA—malondialdehyde; POD—peroxidase; SOD—superoxide dismutase; ROS—reactive oxygen species; TAO—total antioxidant; TBARS—thiobarbituric acid reactive substances; TPX—total peroxides; Ah—aromatic hydrocarbons; E—elements; F—fungicides; H—herbicides; I—insecticides; M—microplastics; O—other substances.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Formicki, G.; Goc, Z.; Bojarski, B.; Witeska, M. Oxidative Stress and Neurotoxicity Biomarkers in Fish Toxicology. Antioxidants 2025, 14, 939. https://doi.org/10.3390/antiox14080939

AMA Style

Formicki G, Goc Z, Bojarski B, Witeska M. Oxidative Stress and Neurotoxicity Biomarkers in Fish Toxicology. Antioxidants. 2025; 14(8):939. https://doi.org/10.3390/antiox14080939

Chicago/Turabian Style

Formicki, Grzegorz, Zofia Goc, Bartosz Bojarski, and Małgorzata Witeska. 2025. "Oxidative Stress and Neurotoxicity Biomarkers in Fish Toxicology" Antioxidants 14, no. 8: 939. https://doi.org/10.3390/antiox14080939

APA Style

Formicki, G., Goc, Z., Bojarski, B., & Witeska, M. (2025). Oxidative Stress and Neurotoxicity Biomarkers in Fish Toxicology. Antioxidants, 14(8), 939. https://doi.org/10.3390/antiox14080939

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop