Available online on 15.01.2026 at http://jddtonline.info
Journal of Drug Delivery and Therapeutics
Open Access to Pharmaceutical and Medical Research
Copyright © 2026 The Author(s): This is an open-access article distributed under the terms of the CC BY-NC 4.0 which permits unrestricted use, distribution, and reproduction in any medium for non-commercial use provided the original author and source are credited
Open Access Full Text Article Research Article
Oxidative stress and neuronal dysfunction induced by combined low-dose exposure to abamectin and cypermethrin
1,2 Essotolom Badjabaissi*, 3 Sabrina Chris Janiba Sanvee, 4 Yendubé T. Kantati, 1,4 Komlan Mawubédjro Dossou-Yovo, 4,5 Mindédé Assih, 1 Tchare Assiki, 6 Toukilnan Djiwa, 3 Batomayena Bakoma, 1 Aboudoulatif Diallo
1 Laboratory of Toxicology, Faculty of Health Sciences, University of Lomé, Lomé, Togo
2 Laboratory of Chemistry of Water, Food and Various Products, National Institute of Hygiene Lomé, Lomé, Togo
3 Laboratory of Chemistry and Natural Substances, Faculty of Health Sciences, University of Lomé, Lomé, Togo
4 Laboratory of Physiology/Pharmacology Laboratory, Research Unit in Pathophysiology, Bioactive Substances and Safety (PBSI), Faculty of Sciences, University of Lomé, Lomé, Togo
5 Laboratory of Pharmacology, Faculty of Health Sciences, University of Lomé, Lomé, Togo
6 Department of Pathology, Kara University of Kara Hospital, Kara, Togo
|
Article Info: _______________________________________________ Article History: Received 24 Oct 2025 Reviewed 08 Dec 2025 Accepted 02 Jan 2026 Published 15 Jan 2026 _______________________________________________ Cite this article as: Badjabaissi E, Sanvee SCJ, Kantati YT, Dossou-Yovo KM, Assih M, Assiki T, Djiwa T, Bakoma B, Diallo A, Oxidative stress and neuronal dysfunction induced by combined low-dose exposure to abamectin and cypermethrin, Journal of Drug Delivery and Therapeutics. 2026; 16(1):104-118 DOI: http://dx.doi.org/10.22270/jddt.v16i1.7534 ______________________________________________ For Correspondence: Essotolom Badjabaïssi, Laboratory of Toxicology, Faculty of Health Sciences, University of Lomé, Lomé, Togo |
Abstract _______________________________________________________________________________________________________________ Background : The increase in agricultural production has been accompanied by intensive pesticide use, leading to chronic environmental contamination. In Togo, residues exceeding regulatory limits have been detected in vegetable crops, particularly abamectin (ABM) and cypermethrin (CYP). ABM, derived from Streptomyces avermitilis, acts on GABA-gated chloride channels and crosses the blood–brain barrier, inducing oxidative stress and neurotoxic damage. CYP, a synthetic pyrethroid, targets voltage-gated sodium channels, resulting in abnormal neuronal discharges and excessive production of reactive oxygen species. While their acute effects are well known, the subchronic impacts of low-dose exposure, especially in combination, remain poorly understood. In this context, the present study aims to evaluate the subchronic neurotoxic effects of combined exposure to abamectin and cypermethrin, administered at realistic doses, in rats. Methods : This study evaluated the subchronic neurotoxicity of ABM and CYP, administered individually or in combination for 90 days to 40 Wistar rats divided into five groups: control, ABM (0.3 mg/kg/day), CYP (0.7 mg/kg/day), mixture M1 (ABM 0.3 + CYP 0.7 mg/kg/day), and mixture M2 (ABM 0.6 + CYP 1.4 mg/kg/day). Results : The results revealed that exposure, particularly to mixtures, decreased brain protein levels, significantly reduced glutathione, superoxide dismutase, and catalase, and markedly increased malondialdehyde levels compared to groups exposed to a single pesticide.. Glutathione S-transferase activity was less affected, but caspase-3 was activated in M1 and M2 groups, indicating apoptosis induction. Histological analyses in M1 and M2 groups showed pronounced hypocellularity associated with cellular hypertrophy and necrotic foci compared with controls. Conclusions : The observed synergistic effect highlights an increased toxicological risk, underscoring the need for further mechanistic studies and appropriate regulatory measures. Under environmental conditions where combined exposures are frequent, these results highlight the importance of considering the impact of mixtures rather than isolated compounds. Keywords: Abamectin, Cypermethrin, Oxidative stress, Pesticide mixture, low-dose |
INTRODUCTION
Global demographic expansion and the intensifying demand for agricultural productivity have driven a substantial escalation in pesticide use over the past decades. Insecticides, fungicides, and herbicides are now widely employed to maximize agricultural yields 1–3. This massive use, although effective in the short term, is accompanied by a continuous release of residues into the environment 4–6. These residues, present in soils, water, and crops, chronically expose human and animal populations to low doses of potentially toxic compounds 7–10.
In Togo, several studies have highlighted frequent, even inappropriate, pesticide use in agricultural practices 11, 12. Pesticide residues have notably been detected in locally grown vegetables, sometimes at levels exceeding the maximum limits set by the Codex Alimentarius. A recent study conducted by Badjabaissi et al. (2024) revealed the presence of multiple pesticide residues in market gardening crops. Among these pesticides are abamectin and cypermethrin, two substances particularly widespread in Togolese agriculture 13.
Abamectin, derived from the metabolism of Streptomyces avermitilis, and cypermethrin, a synthetic pyrethroid, are both recognized for their effectiveness against a wide spectrum of parasites. However, beyond their usefulness in agriculture, these molecules present concerning toxicological profiles. Numerous studies have demonstrated their impact on various organs 14–20, with a predilection for the central nervous system 21–25. Their mechanism of action relies mainly on the disruption of ion channels and the induction of oxidative stress, leading to neuronal alterations that are sometimes irreversible. Abamectin is one of the most widely used pesticides in the world due to its powerful insecticidal and vermicidal activity. It easily crosses the blood-brain barrier and activates GABA-dependent chloride channels, leading to inhibition of nerve influx and neurotoxic damage 26, 27. It is also known to affect non-target species, including fish, beneficial insects, and mammals 28, 29. Studies have shown that abamectin disrupts antioxidant activities and increases reactive oxygen species (ROS), impairing immune responses and causing neurotoxic and genotoxic damage 23, 30. Cypermethrin, on the other hand, is a synthetic pyrethroid commonly used in agriculture, veterinary medicine, and in the control of domestic insects/parasites 31. Although considered safe for domestic applications, some studies have reported adverse effects of cypermethrin on the brains of laboratory animals 32, 14. Cypermethrin acts mainly on voltage-dependent sodium channels, keeping them open longer than normal, which provokes repetitive neuronal discharges and excessive production of reactive oxygen species (ROS) 32, 33. These ROS are responsible for oxidative damage, including lipid peroxidation and cellular dysfunctions.
In laboratory animals, repeated oral exposures to these pesticides have led to the generation of reactive oxygen species (ROS) and nitrogen species, resulting in lipid peroxidation (LPO) 34, 35. These data suggest that the brain, due to its low antioxidant capacity, high oxygen consumption, and abundance in polyunsaturated fatty acids, represents a preferred target of pesticide toxicity 36, 37.
Although the acute neurotoxic effects of abamectin and cypermethrin are well documented, the consequences of prolonged low-dose exposures, often more representative of real exposure conditions, remain less studied. Moreover, the combined effects of these two pesticides, which may be synergistic or additive, have not been experimentally investigated. It is, however, well established that it is difficult to predict the toxicity of a mixture from the individual effects of its components 38, hence the importance of specific research on pesticide cocktails.
In this context, the present study aims to evaluate the subchronic neurotoxic effects of combined exposure to abamectin and cypermethrin, administered at realistic doses, in rats. The focus will be on the modulation of the brain antioxidant system, particularly on key oxidative stress enzymes, the apoptotic system, and the histology of the cerebral cortex, in order to better understand the mechanisms underlying this mixed neurotoxicity.
MATERIALS AND METHODS
1. Study framework and animal material
Male and female rats of the Wistar strain were provided by the animal facility of the Pharmaceutical Department of the Faculty of Health Sciences (FSS) of the University of Lome (Lome-Togo). A total of 20 male et 20 femele rats aged 5 to 6 weeks weighing approximately 150-180 g were used. They were fed and provided with water ad libitum under standard breeding conditions (temperature: 22±3°C, photoperiod 12/12H). The rats' urine and droppings collection trays were maintained once every three days until the end of the experiment. The work was carried out in the pharmacology and toxicology laboratory of the said department.
2. Chemicals
In this study, we used abamectin (Pestanal®, 31732) and cypermethrin (Pestanal®, 36128) both from Sigma-Aldrich. The products and reagents used for the evaluation of biological parameters were mainly from Sigma-Aldrich such as TRIS buffer (252859-100G), 5,5'-Dithiobis(2-Nitrobenzoic acid) (D8130-5G), glutathione (G4251-10G), pyrogallol (06931-50MG), 1-Chloro-2,4-dinitrobenzene (237329-50G) as well as the kits for the enzyme assay (Caspase-3, CASP3C-1KT).
3. Experimental
The rats were divided into five (05) groups, with eight (08) rats per group. Group T, the control group (T), received an acetone/distilled water solution (0.5%); group ABM, rats treated with abamectin (ABM) at a dose of 0.3 mg/kg/day; group CYP, rats treated with cypermethrin (CYP) at a dose of 0.7 mg/kg/day. Group M1, rats treated with ABM (0.3 mg/kg/day) and CYP (0.7 mg/kg/day) and Group M2, rats treated with ABM (0.6 mg/kg/day) and CYP (1.4 mg/kg/day).
They were subjected to a 10 day housing period in the animal facility of the Pharmaceutical Department of the Faculty of Health Sciences, University of Lomé. Two (02) pesticides (abamectin and cypermethrin) were used in this study, alone or in a mixture at consecutive doses of 0.3 mg/kg/day (1/30 of the LD50) and 0.7 mg/kg/day (1/250 of the LD50) for oral administration for 90 days. The choice of these doses was made on the basis of the results of pesticide dosage in vegetables reported by Badjabaissi et al. (2024) in Togo 13. This study revealed that abamectin levels exceeded the maximum residue limit (MRL) in 7 out of 10 samples, while cypermethrin had the highest concentrations among all the substances analyzed, also exceeding the MRL. The pesticides were dissolved in acetone (0.5%) and then supplemented with distilled water to reach the required volume. The composition was prepared weekly throughout the experimental period.
4. Study of cellular and molecular integrity of neurocytes
Estimation of relative brain weight and preparation of cytosolic samples
Brains were collected the day after the 90th day of treatment, after a 12-hour fasting period. They were quickly rinsed with cold phosphate-buffered saline (PBS, 0.1 M, pH 7.4) and gently dried with semi-absorbent paper at 4°C. Each brain was weighed, and the relative weight (RP) was calculated using the following formula : RP = 100 x (organ weight/rat weight). For each experimental group, two brains (one male and one female) were randomly selected and fixed in 10% formalin for histopathological analysis. The remaining six brains (three males and three females) were used for biochemical analyses. Each brain intended for biochemical analysis was homogenized in cold phosphate-buffered saline (PBS, 0.1 M, pH 7.4) using a glass homogenizer. One (1) gram of total brain was diluted in 2 mL of PBS, and the resulting homogenate was centrifuged at 3000 rpm for 15 minutes at 4°C. The supernatant (cytosolic fraction) was collected and used for the determination of biochemical parameters. The cytosolic fractions were used to assess:
Evaluation of biochemical parameter : Total protein determination at the tissue level
The total protein concentration in tissue homogenates was determined according to the Bradford method (1976), using bovine serum albumin (BSA) as the standard 39.
Evaluation of oxidative stress parameters
The assay for malondialdehyde (MDA) was performed following the method described by Patlolla et al. (2009) 40. Briefly, the reaction mixture contained 1-methyl-2-phenylindole (10.3 mM in acetonitrile, 650 µL), to which 250 µL of either MDA standard or tissue homogenate was added. The mixture was vortexed, and 150 µL of hydrochloric acid (12 N) and 10 µL of butylated hydroxytoluene (BHT, 0.1 M) were subsequently added. The samples were incubated at 45 °C for 1 hour and then centrifuged at 3500 rpm for 10 minutes. The absorbance of the supernatant was measured at 586 nm. The MDA standard curve was prepared using concentrations of 0, 0.625, 1.25, 2.5, 5, 10, and 20 nM. A blank solution consisting of 75% acetonitrile, 25% ferric chloride, and 200 µL of Tris buffer (pH 7.4) was used as reference.
The determination of reduced glutathione (GSH) levels was carried out using the modified method of Sedlak and Raymond (1968) 41. In this procedure, tissue homogenates or standard GSH solutions were mixed with Tris buffer (0.2 M, pH 8.2) and 5,5'-dithiobis-2-nitrobenzoic acid (DTNB, 0.01 M). The reaction mixture was brought to a final volume of 1 mL with absolute methanol, sealed, and homogenized for 30 minutes at room temperature. After centrifugation at 3000 rpm for 15 minutes, the absorbance of the supernatant was measured at 412 nm following a 5-minute incubation. GSH concentrations were calculated from a standard calibration curve (10-6 to 10-2 mg/mL) and expressed as nanomoles per milligram of protein (nmol GSH/mg protein).
Glutathione S-transferase (GST) activity was determined according to the method of Habig et al. (1974) 42. The reaction mixtures were prepared as follows: the blank contained 850 µL of phosphate-buffered saline (PBS, 0.1 M, pH 7.4), 55 µL of 1-chloro-2,4-dinitrobenzene (CDNB, 0.02 M), and 100 µL of reduced glutathione (GSH, 0.1 M). For the test samples, 830 µL of PBS, 50 µL of CDNB (0.02 M), 100 µL of GSH (0.1 M), and 20 µL of tissue homogenate were mixed. The formation of the conjugate, 1-S-glutathionyl-2,4-dinitrobenzene, was monitored spectrophotometrically at 340 nm. GST activity was expressed as nanomoles of CDNB conjugated per minute per milligram of protein (nmol CDNB/min/mg protein).
Catalase (CAT) activity was determined according to the method of Aebi (1984) 43. The reaction mixtures were prepared as follows: the blank contained 800 µL of phosphate-buffered saline (PBS, 0.1 M, pH 7.4) and 200 µL of hydrogen peroxide (H2O2, 0.5 M). For the test samples, 780 µL of PBS, 200 µL of H2O2 (0.5 M), and 20 µL of tissue homogenate were mixed. Enzymatic activity was measured spectrophotometrically by monitoring the decrease in absorbance at 240 nm, corresponding to the decomposition of H2O2. Absorbance readings were taken after a 15 second delay over a period of 60 seconds. Catalase activity was expressed as micromoles of H2O2 decomposed per minute per milligram of protein (µmol H2O2/min/mg protein).
Superoxide dismutase (SOD) activity was determined according to the method described by Marklund and Marklund (1974) 44 and modified by Hassan et al. (2014) 45. The assay was based on the competition between the autoxidation of pyrogallol and the dismutation of superoxide radicals by SOD. Tissue homogenates or standards were incubated with Tris-succinate buffer (0.05 M, pH 8.2) and pyrogallol (8 mM) at 25 °C for 20 minutes. The rate of pyrogallol autoxidation was monitored spectrophotometrically at 412 nm over a 3minute period, with absorbance readings taken every 30 seconds.
Caspase-3 activity was assessed in cytosolic extracts of brain tissue using a commercial colorimetric kit (Caspase-3 Activity Colorimetric Assay Kit, ref. APT131, Sigma®, Germany). The method was adapted for tissue analysis. The principle was based on the hydrolysis of the specific substrate Ac-DEVD-pNA, resulting in the release of the chromophore p-nitroaniline (pNA), whose absorbance at 405 nm is proportional to enzymatic activity. A standard curve was generated from pNA dilutions to establish the relationship between absorbance and concentration. Cytosolic extracts were incubated with the substrate at 37°C for 1 to 2 hours. After incubation, pNA absorbance was measured using a UV/Visible spectrophotometer. Caspase-3 activity was expressed in relative absorbance units (ΔDO) and compared between experimental groups. The increase in absorbance reflected increased caspase-3 activation, indicating an intensification of the apoptotic process in treated brain tissues.
5. Histological Tests
Histological examination was carried out in the cerebral cortex of rats from different batches. The number of tissue fragments, their appearance, size, and weight of each fragment were determined. Sections were then made, followed by systematic sampling. After dehydration in various ethyl alcohol baths of increasing strength (70%, 95%, and 100%), the samples were impregnated and embedded. Thin sections were then cut using a microtome and spread onto slides for hematoxylin-eosin staining. The slides were interpreted under a light microscope at various magnifications.
6. Statistical Analysis
The results were expressed as the mean of six replicates (mean ± standard deviation). Data visualization was performed using Microsoft Office Excel 2013 to generate graphs and histograms. Statistical analyses were conducted using GraphPad Prism version 8.4.3 (GraphPad Software, USA). The results were expressed as mean values ± standard error of the mean (M ± SEM). A p-value < 0.05 was considered statistically significant. Data were analyzed using one-way ANOVA, followed by Tukey’s multiple comparison test, according to the following significance levels : p > 0.05: not significant (ns) ; 0.05 > p > 0.01: significant (*) ; 0.01 > p > 0.001: highly significant (**) ; p < 0.001: very highly significant (***) ; p < 0.0001: extremely significant (****).
RESULTS
The results showed non-significant decreases in relative brain weight of pesticide-treated rats compared to the control group (Figure 1).
Figure 1 : Evolution of the relative weight (%) of the brains of treated rats after 90 days of treatment.
Abbreviations: T = Control; ABM = Abamectin (0.3 mg/kg); CYP = Cypermethrin (0.7 mg/kg); M1 = Abamectin (0.3 mg/kg) + Cypermethrin (0.7 mg/kg). M2 = Abamectin (0.6 mg/kg) + Cypermethrin (1.4 mg/kg). Each value represents Mean ± SEM. (n) number of animals per group = 8. (*) indicates a significant difference compared to the control group (T). Significance from p < 0.05.
Protein Assays
The results of cytosolic protein assessment in the whole brain are summarized in Figure 2. A significant decrease in protein levels was observed in the ABM (p < 0.01), M1 (p < 0.001) and M2 (p < 0.0001) groups, compared to the control group. Treated rats in the M1 group showed a significant decrease in protein levels compared to the CYP group.
Figure 2: Effect of pesticides on the variation of protein levels in the total brain of rats after 90 days of treatment.
Abbreviations: T = Control; ABM = Abamectin (0.3 mg/kg); CYP = Cypermethrin (0.7 mg/kg); M1 = Abamectin (0.3 mg/kg) + Cypermethrin (0.7 mg/kg). M2 = Abamectin (0.6 mg/kg) + Cypermethrin (1.4 mg/kg). Each value represents Mean ± SEM. (n) number of animals per group = 6. (*) indicates a significant difference compared to the control group (T); (#) compared to the CYP group. Significance from p < 0.05.
Effets des radicaux libres
Mixtures M1 (p < 0.01) and M2 (p < 0.0001) significantly increased MDA levels in the brain of rats compared to the control group. In this same figure, a highly significant increase in MDA levels in rats in group M2 (p < 0.0001) was reported compared to group M1. The addition of abamectin to cypermethrin (1.1 mg/kg/day) in rats (group M1) resulted in a significant increase (p < 0.01) in MDA levels compared to rats given only cypermethrin (group CYP). Furthermore, a significant increase was noted in group ABM (p < 0.05) compared to the control (Figure 3).
Figure 3: Effect of pesticides on the cytosolic variation of malondialdehyde levels in the whole brain of rats after 90 days of treatment.
Abbreviations: T = Control; ABM = Abamectin (0.3 mg/kg); CYP = Cypermethrin (0.7 mg/kg); M1 = Abamectin (0.3 mg/kg) + Cypermethrin (0.7 mg/kg). M2 = Abamectin (0.6 mg/kg) + Cypermethrin (1.4 mg/kg). Each value represents Mean ± SEM. (n) number of animals per group = 6. (*) indicates a significant difference compared to the control group (T); (#) compared to the CYP group and (z) compared to the M1 group. Significance from p < 0.05.
Effects on Non-Enzymatic Antioxidant System Parameters
The effects of abamectin and cypermethrin on GSH levels are shown in Figure 4. Exposure of rats to abamectin and cypermethrin resulted in a significant decrease in total brain glutathione (GSH) concentrations in groups ABM (p < 0.01); CYP (p < 0.05); M1 (p < 0.01); and M2 (p < 0.0001) compared to the control group (T). A significant decrease in glutathione levels was observed in rats from group M2 (p < 0.0001) compared to rats from group M1.
Figure 4: Effect of pesticides on cytosolic variation of glutathione levels in the whole brain of rats after 90 days of treatment.
Abbreviations: T = Control; ABM = Abamectin (0.3 mg/kg); CYP = Cypermethrin (0.7 mg/kg); M1 = Abamectin (0.3 mg/kg) + Cypermethrin (0.7 mg/kg). M2 = Abamectin (0.6 mg/kg) + Cypermethrin (1.4 mg/kg). Each value represents Mean ± SEM. (n) number of animals per group = 6. (*) indicates a significant difference compared to the control group, (z) compared to group M1. Significance from p < 0.05.
Effects on Enzymatic Antioxidant System Parameters
Exposure of rats to abamectin and cypermethrin did not result in a significant decrease in total brain glutathione S-transferase (GST) levels in treated groups compared to the control group (T) (Figure 5-A). A significant decrease in glutathione levels was observed in rats in group M1 compared to rats in group T (p < 0.001) and in group CYP (p < 0.01).
The effects of pesticides on rats were reflected in a significant decrease in SOD activity in groups M1 (p < 0.01) and M2 (p < 0.0001) compared to the control group. Compared to the groups treated with abamectin (ABM) and cypermethrin (CYP) alone, co-administration in group M1 resulted in a significant decrease in SOD activity in the total brain, p < 0.05 and p < 0.001, respectively (Figure 5-B).
The study showed a significant decrease (p < 0.05) in the level of catalase activity in the brain tissues of rats treated with abamectin, cypermethrin, and mixtures of both pesticides M1 and M2 (p < 0.0001) compared to rats in the control group. Treatment of rats with the abamectin and cypermethrin mixture in batch M1 resulted in a significant decrease (p < 0.05) in catalase activity, compared to rats treated with only abamectin (batch ABM) and cypermethrin (batch CYP). A significant decrease (p < 0.001) in catalase activity was also observed in batch M2 compared to batch M1 (Figure 5-C).
Figure 5: Effect of pesticides on the cytosolic variation of the parameters of the enzymatic antioxidant system in the whole brain of rats after 90 days of treatment. (A) Glutathione S-transferase levels; (B) Superoxide dismutase activity; (C) Catalase activity.
Abbreviations: T = Control; ABM = Abamectin (0.3 mg/kg); CYP = Cypermethrin (0.7 mg/kg); M1 = Abamectin (0.3 mg/kg) + Cypermethrin (0.7 mg/kg). M2 = Abamectin (0.6 mg/kg) + Cypermethrin (1.4 mg/kg). Each value represents Mean ± SEM. (n) number of animals per group = 6. (*) indicates a significant difference compared to the control group (T); (Σ) compared to the ABM group; (#) compared to the CYP group and (z) compared to the M1 group. Significance from p < 0.05.
d. Effects of pesticides on pro-apoptotic markers
- Caspase-3 activity
Statistical analysis of the results of the enzymatic assay of caspase-3 activity showed a significant increase in batches M1 (p < 0.05) and M2 (p < 0.0001) compared to the control batch. An increase in caspase-3 activity in batches M2 (p < 0.05) and M1 (p < 0.05) was obtained, respectively, compared to batches M1 and CYP. No statistically significant difference was observed in the other batches compared to the control (Figure 6).
Figure 6: Effect of pesticides on the variation of caspase 3 activity in the total brain of rats after 90 days of treatment.
Abbreviations: T = Control; ABM = Abamectin (0.3 mg/kg); CYP = Cypermethrin (0.7 mg/kg); M1 = Abamectin (0.3 mg/kg) + Cypermethrin (0.7 mg/kg). M2 = Abamectin (0.6 mg/kg) + Cypermethrin (1.4 mg/kg). Each value represents Mean ± SEM. (n) number of animals per group = 6. (*) indicates a significant difference compared to the control group (T); (#) compared to the CYP group and (z) compared to the M1 group. Significance from p < 0.05
e. Effect of pesticides on the histology of the cerebral cortex of rats
Histological analysis of sections of the cerebral cortex of treated rats revealed abnormalities compared to controls (Figure 7). A slight decrease in cellularity, associated with necrosis and vacuolization of the neuropil, was observed in the cerebral cortex of rats in the ABM group compared to controls. Cellularity within normal limits was observed in the CYP group. Histological sections of the cerebral cortex of rats in groups M1 and M2 showed marked hypocellularity with the presence of neuropil vacuolization compared to the control group (Figure 7, D & E). Neuronal necrosis was observed in group M1 (Figure 7, D).
Figure 2 : Histological sections in the rat brain after 90 days of treatment. A = Control; B = Abamectin; C = Cypermethrin; D = M1 (Abamectin + Cypermethrin); E = M2 ((Abamectin + Cypermethrin) x 2). n = neurons with normal structures. nv = neuropils with vacuoles. nc = necrosis. Magnification: x100.
DISCUSSION
Chronic exposure to low concentrations of abamectin, cypermethrin, or their combination did not result in any significant variation in relative brain weight between the treated groups and the control group. This stability could be explained by the fact that the administered doses were likely too low, or the exposure duration too short, to induce morphometric alterations.
Beyond the individual effects of each pesticide, the study also investigated the impact of their combination in order to assess potential interactions (additive or synergistic) that could amplify neurotoxicity. In this context, groups M1 and M2 showed a significant decrease in body weight compared with the control group. However, no significant effect on brain weight was observed following combined exposure. This finding suggests that while co-exposure to abamectin and cypermethrin may disrupt certain physiological parameters such as body weight, it may not, under the experimental conditions of this study, be sufficient to cause detectable morphometric alterations in the brain. Nevertheless, the absence of changes in brain weight does not rule out possible underlying functional or biochemical effects. Indeed, previous studies have shown that low-dose pesticide exposure can affect neurophysiology without necessarily inducing changes in brain mass 7, 46, 47. Thus, the impact of the mixture could manifest in a more subtle manner, particularly at the cellular or molecular level, which justifies the complementary analyses carried out in this study.
According to the results of our study, exposure of rats to pesticides alone (ABM) or in combination (M1 and M2) induced a disruption of the general metabolism of brain cells, particularly in protein profiles. This disruption is greater in batches exposed to the M1 mixture compared to ABM and CYP batches, suggesting an additive or synergistic toxic interaction. The measurement of total protein content is a classical indicator of stress in organisms 48, 49. Under severe environmental constraints (oxidative, thermal, or water stress, pollution, infections...), proteins undergo denaturation processes 50, leading to a reduction in their concentration due to structural degradation and inhibition of synthesis mechanisms 51. Furthermore, oxidative damage to proteins can compromise the integrity of cell membranes and disrupt key functions such as membrane fluidity, mitochondrial homeostasis, and regulation of oxidative stress 52.
The central nervous system (CNS), due to its intense activity, consumes a large proportion of the body’s total oxygen approximately 20-25%. This high oxygen demand is accompanied by a moderate production of reactive oxygen species (ROS), which remains well regulated by endogenous antioxidant systems 53–55. However, chronic exposure to certain pesticides may disrupt this balance by inducing significant oxidative stress, thereby placing the brain among the major targets of their toxicity 34, 56.
Overall, our results highlight an alteration of the cytosolic redox status in the brains of rats exposed to pesticides alone, suggesting a predominance of oxidative stress, characterized by a significant decrease in antioxidant potential. One of the alterations observed in our study was the decrease in GSH content in the cytosolic cells of rats treated with pesticides.
Glutathione (GSH) is an essential component that plays a central role in cellular defense against oxidative stress by scavenging free radicals 57. It functions both as a non-enzymatic antioxidant and as a critical cofactor of enzymes involved in xenobiotic detoxification, particularly by catalyzing conjugation reactions 58, 59. During its metabolic action, the sulfhydryl group of GSH is oxidized into disulfide compounds, and reduced glutathione (GSH) is conjugated for the elimination of xenobiotics 60. Sule et al. (2022) demonstrated that the depletion of intracellular sulfhydryl groups by pesticides is a prerequisite for ROS generation 34. Thus, the significant reduction in GSH content observed in rats exposed to abamectin and cypermethrin alone in this study, after chronic exposure, could lead to an increased susceptibility to free radical–induced damage.
In contrast to our findings, Bekkairi et al. (2024) 61, using a dose of 0.019 mg/kg for 21 days, and Radi et al. (2020) 25, using a dose of 2 mg/kg for 5 days, reported that abamectin increased GSH content. This discrepancy may be explained by the exposure duration. Acute or subacute exposure can trigger an initial adaptive response of the antioxidant system, resulting in elevated GSH levels to counteract oxidative aggression. Conversely, similar results to ours were reported by Sharma et al. (2014) after exposure to a higher dose of cypermethrin (3.83 mg/kg for 7 days) [62]. Our findings indicate that even at low doses, chronic exposure can induce continuous production of free radicals, leading to the progressive depletion of GSH and persistent impairment of the cellular redox defense system.
Beyond GSH, antioxidant enzymes such as GST, SOD, and CAT play crucial roles in the cellular antioxidant system. The conjugation of electrophilic substrates with the thiol group of GSH is catalyzed by glutathione S-transferases (GST), producing less harmful products for the organism 63. GST is also involved in neutralizing lipid peroxides, thereby limiting ROS-induced damage in various cells. Located mainly in the cytosol, these enzymes participate in diverse intracellular transport and biosynthetic processes 64, 65. Superoxide dismutase (SOD) and catalase (CAT) represent the main enzymatic defense mechanisms against the toxic effects of oxygen metabolism.
In the present study, GST and SOD activity was not significantly altered in rats exposed to abamectin or cypermethrin alone. This suggests that under the experimental conditions, these two antioxidant enzymes were able to maintain their function relatively stably, despite pesticide exposure. However, a downward trend was observed, possibly indicating the onset of redox imbalance, though not reaching statistical significance. In contrast, a significant inhibition of catalase (CAT) activity was observed in these groups. This enzyme plays a central role in the degradation of hydrogen peroxide (H₂O₂) into water (H₂O) and molecular oxygen (O₂), thereby preventing the accumulation of oxidant products harmful to cells 66–68. Our results suggest that these pesticides increase H₂O₂ in the cytosol and generate massive amounts of hydroxyl radicals °OH 25, 69, 70.
Because of the extremely reactive, unstable, and low-concentration nature of free radicals, indirect markers are generally measured 71. Malondialdehyde (MDA) levels reflect the extent of lipid peroxidation and represent one of the fundamental mechanisms of free radical–induced cellular damage 72. MDA is a product of oxidative degradation of unsaturated lipids in membranes 72, 73. Thus, under stress induced by xenobiotics such as pesticides, when antioxidant systems become ineffective, cellular damage can be assessed through MDA levels, making it a reliable biomarker of oxidative stress.
Previous studies have reported elevated MDA levels in the brains of rats treated with abamectin 23, 61 and cypermethrin 62,74,75 at relatively high doses. In our study, a significant increase in MDA levels was observed in the ABM group compared to controls. This may be due to insufficient anti-ROS capacity, leading to hydroxyl anion generation that attacks membrane phospholipids. Unlike abamectin, cypermethrin did not induce a significant change in MDA levels. In the ABM group, inhibition of antioxidant enzymes SOD and CAT, responsible for eliminating free radicals, led to superoxide accumulation. This accumulation promoted lipid peroxidation, as revealed in our study. Previous research has reported that lipid peroxidation accumulation is accompanied by DNA damage, which can disrupt gene expression and lead to cell death 76–78. This oxidative stress process also disrupts neuronal structure and reduces their capacity to produce intracellular ATP, essential for proper cellular functioning 79. Such cellular damage leads to degeneration in key brain regions such as the hippocampus and striatum.
After characterizing the individual effects of each pesticide on oxidative stress, mixture analysis allows assessment of potential interactions whether additive, synergistic, or antagonistic. A significant accumulation of superoxide, reflected by marked inhibition of SOD, CAT, and GST activities, was observed in the M1 group compared to rats exposed to a single pesticide. Lipid peroxidation was also more pronounced in the M1 group. The strong inhibition of GST activity in M1 rats indicates insufficient conjugation of electrophiles and inadequate detoxification of these species. This inhibition is particularly concerning as it is accompanied by a significant reduction in CAT and SOD activity in the mixture group. The decline in these free radical–eliminating enzymes confirms the oxidative stress state induced by these pesticides in brain tissue during co-exposure. The simultaneous decrease of these enzymes likely promotes further accumulation of superoxide and other ROS, exacerbating oxidative stress in the brain. These results suggest the failure of the brain’s antioxidant machinery to counteract ROS accumulation induced by these pesticides, likely due to the high oxidative metabolism in the brain 80.
Although other studies on abamectin and cypermethrin administered separately have reported similar effects at higher doses 30, 62, 81, 82, our study shows that comparable effects can occur at lower doses during chronic co-exposure to both pesticides. Since pesticides generate excess ROS, the compensatory effect of antioxidant enzymes may quickly become overwhelmed 34. The significantly higher MDA increase in the M1 group compared to the ABM and CYP groups suggests altered neuronal lipid membrane integrity 83 and rapid depletion of intracellular ATP, compromising neuronal viability and leading to cell degeneration in the hippocampus, striatum, and whole brain. Such disturbances could have major impacts on the physiology and behavior of rats intoxicated by these pesticides 55, 84, 85. Moreover, more significant changes in redox parameters were observed in the M2 group compared to the M1 group, suggesting a dose-dependent effect. These findings reveal that combined pesticide exposure exacerbates oxidative stress cumulatively or synergistically, leading to deeper impairment of antioxidant defense mechanisms.
The decrease in antioxidant enzyme activity was concomitant with a reduction in total protein content in the brains of treated rats. This reduction may be attributed to the inhibition of protein synthesis and reduced cell survival due to antioxidant enzyme deficiency observed in our study. In light of these imbalances, the possible involvement of apoptotic mechanisms warrants consideration.
Reactive oxygen species (ROS) trigger fatal mitochondrial energy (ATP) depletion, proteolytic enzyme induction, and DNA fragmentation, ultimately leading to apoptotic cell death 33, 86. Apoptosis is a programmed form of cell death, essential for cellular homeostasis, and is activated by various stimuli through caspases 87–89. The accumulation of abnormal protein aggregates can also induce apoptosis, a mechanism implicated in several neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease 90–93.
Oxidative stress induces the opening of mitochondrial pores, leading to the release of cytochrome c, activation of Apaf-1, and a cascade involving caspase-9 and caspase-3, the central proteins in apoptotic execution 94–96. In this study, we investigated for the first time the expression of caspase-3 genes in rats exposed to abamectin, cypermethrin, and their mixture. However, no significant increase in caspase-3 activity was observed with abamectin or cypermethrin alone. In contrast, our findings differ from those of Ibrahim et al. (2022), who reported increased caspase-3 activity in the fetal brains of pregnant rats exposed to abamectin alone 23. Moreover, previous studies have shown that brain apoptosis can result from oxidative stress induced by cypermethrin at high doses 70, 97. Thus, the discrepancy between our results and the literature could be explained by differences in experimental conditions (species, developmental stage, exposure duration, dose, or tissue analyzed).
Conversely, a significant increase in caspase-3 activity in the cytosol was observed in the mixture groups (M1 and M2), indicating enhanced activation of the apoptotic pathway. This activation may result from cumulative or synergistic toxic interactions between abamectin and cypermethrin, thereby amplifying pro-apoptotic mechanisms.
Indeed, individual exposure to abamectin or cypermethrin caused depletion of antioxidant enzymes such as catalase, superoxide dismutase (SOD), and glutathione (GSH), thereby weakening cellular antioxidant defenses. The combination of both substances could therefore intensify ROS production, leading to cytosolic stress disruption, DNA damage, and massive induction of neuronal apoptosis effects already reported at high doses for each pesticide when administered separately 30, 98, 99.
Although activation of this apoptotic mechanism indicates toxicity, it can also be interpreted as an adaptive response by the organism, aiming to eliminate severely damaged cells and thereby preventing deleterious genetic alterations that may lead to carcinogenesis 100, 101.
Histological analysis remains an essential step in understanding the effects of toxic substances on the central nervous system. Unlike biochemical or behavioral analyses, it allows direct observation of tissue organization and integrity 102, 103. In the case of the brain, histological sections provide precise insight into the state of cells, revealing neuronal loss, changes in cell density, or signs of degeneration. They thus establish a direct link between pesticide exposure and the observed structural alterations 104. The distribution of antioxidant defense mechanisms in the brain varies across cell types and anatomical regions 105. The cerebral cortex is particularly sensitive to oxidative stress due to its specific redox status, making it more vulnerable to ROS-induced damage 106.
In our study, histological examination of the cerebral cortex of rats chronically exposed to pesticides revealed a reduction in cell number compared with controls. In the ABM group, cortical sections showed necrosis and neuropil vacuolization compared with controls. These results suggest moderate neuronal damage, consistent with previous observations. Indeed, Bekkairi et al. (2024) reported in Algeria a reduction in brain size in rats treated with abamectin (0.0019 mg/kg for 21 days) compared with controls, due to cell destruction 61. Moreover, these authors described advanced autolytic changes and the presence of dark spots in several regions. Radi et al. (2019) reported pyknosis and black-stained pyramidal neurons associated with degenerative changes in the pyramidal layer of hippocampal sections in rats following abamectin administration (2 mg/kg for five days) 25. These data reinforce the idea that abamectin induces neurotoxicity marked by neuronal damage. Neuropil vacuolization, frequently associated with neurodegenerative diseases, reflects cellular degeneration and is a clear sign of brain injury.
In the CYP group, cortical cell density was within normal limits compared with controls. These results contrast with those of Ali et al. (2020), who reported shrunken and pale pyramidal cells with contracted cytoplasmic processes in the cerebral cortex of rats exposed to cypermethrin (1 mg/kg, three times per day for 21 days) 75. This discrepancy may be related to the relatively lower dose and shorter exposure duration in our study. It suggests that, under certain experimental conditions, cypermethrin alone may not induce major histological lesions. By contrast, histological sections of the cerebral cortex of rats in the M1 and M2 groups revealed marked hypocellularity and neuropil vacuolization compared with controls, indicating more pronounced alterations in cell density than those induced by individual pesticides. Cellular necrosis was also observed in the M1 group, suggesting neuronal loss due to direct cytotoxicity 107, 108.
The combination of these pesticides potentiates their deleterious effects, causing more severe brain damage than each pesticide administered alone. These histological observations corroborate the biochemical results in the M1 groups, showing increased oxidative stress, activation of apoptotic pathways, and neurobehavioral alterations compared to individual exposures. The combination of abamectin and cypermethrin thus appears to amplify neurotoxicity by simultaneously engaging apoptotic and necrotic mechanisms 109, 110. These results would support the hypothesis of additive or synergistic interactions between the two pesticides, making their combination more harmful than either pesticide alone. An increase in stress was noted in the M2 batch compared to the M1 batch, reflecting a dose-dependent effect. This toxicological potentiation more accurately reflects environmental and occupational exposure conditions, where individuals are rarely exposed to a single pesticide.
CONCLUSION
This study reveals that chronic low-dose exposure to abamectin or cypermethrin alone results in limited alterations in the rat brain, whereas their combination induces much more pronounced effects. Co-exposure disrupted protein metabolism and redox status, promoted lipid peroxidation, activated apoptotic pathways, and caused more severe histological damage. These findings suggest an additive or synergistic effect between the two pesticides, reflecting potentiation of their neurotoxicity. Under environmental conditions where combined exposures are frequent, these results highlight the importance of considering the impact of mixtures rather than isolated compounds. Further investigations, particularly on neurotransmitters and neurobehavioral functions, are necessary to better characterize the neurological risks associated with such co-exposures.
Funding Statement: No funding to declare.
Data Availability Statement: Data are available from the corresponding author upon request.
Acknowledgements: Not applicable
Conflict of Interest Statement: The authors declare that there are no conflicts of interest.
REFERENCES
1. Khan BA, Nadeem MA, Nawaz H, Amin MM, Abbasi GH, Nadeem M, et al. Pesticides: Impacts on Agriculture Productivity, Environment, and Management Strategies. In: Aftab T, éditeur. Emerging Contaminants and Plants [Internet]. Cham: Springer International Publishing; 2023 [cité 30 oct 2025]. p. 109‑34. (Emerging Contaminants and Associated Treatment Technologies). https://doi.org/10.1007/978-3-031-22269-6_5
2. Garraway JL. Insecticides, Fungicides and Herbicides. In: Biotechnology-the Science and the Business [Internet]. CRC Press; 2020 [cité 30 oct 2025]. p. 497‑514. Disponible sur: https://www.taylorfrancis.com/chapters/edit/10.1201/9781003078432-24/insecticides-fungicides-herbicides-james-garraway https://doi.org/10.1201/9781003078432-24
3. Gianessi LP. The increasing importance of herbicides in worldwide crop production. Pest Manag Sci. oct 2013;69(10):1099‑105. https://doi.org/10.1002/ps.3598 PMid:23794176
4. Yadav A, Tandon A, Seth B, Goyal S, Singh SJ, Tiwari SK, et al. Cypermethrin Impairs Hippocampal Neurogenesis and Cognitive Functions by Altering Neural Fate Decisions in the Rat Brain. Mol Neurobiol. janv 2021;58(1):263‑80. https://doi.org/10.1007/s12035-020-02108-9 PMid:32920670
5. Carvalho FP. Pesticides, environment, and food safety. Food Energy Secur. mai 2017;6(2):48‑60. https://doi.org/10.1002/fes3.108
6. Gavrilescu M. Fate of Pesticides in the Environment and its Bioremediation. Eng Life Sci. déc 2005;5(6):497‑526. https://doi.org/10.1002/elsc.200520098
7. Ghasemnejad-Berenji M, Nemati M, Pourheydar B, Gholizadeh S, Karimipour M, Mohebbi I, et al. Neurological effects of long-term exposure to low doses of pesticides mixtures in male rats: Biochemical, histological, and neurobehavioral evaluations. Chemosphere. 2021;264:128464. https://doi.org/10.1016/j.chemosphere.2020.128464 PMid:33049502
8. Eddleston M. Poisoning by pesticides. Medicine (Baltimore). 2020;48(3):214‑7. https://doi.org/10.1016/j.mpmed.2019.12.019
9. Singh NS, Sharma R, Parween T, Patanjali PK. Pesticide Contamination and Human Health Risk Factor. In: Oves M, Zain Khan M, M.I. Ismail I, éditeurs. Modern Age Environmental Problems and their Remediation [Internet]. Cham: Springer International Publishing; 2018 [cité 30 oct 2025]. p. 49‑68. https://doi.org/10.1007/978-3-319-64501-8_3
10. Grewal AS. Pesticide Residues in Food Grains, Vegetables, and Fruits: A Hazard to Human Health.(2017) J Med ChemToxicol 2 (1): 1-7. J Med Chem Toxicol [Internet]. 2017 [cité 30 oct 2025];2(1). Disponible sur: https://gcwgandhinagar.com/econtent/document/1587965466Pesticide-Residues-in-FoodGrains-Vegetables-and-Fruits-A-Hazard-to-Human-Health.pdf
11. Mensah AA, Mawussi G, Karou SD. Gestion des pesticides sur les perimetres maraichers de la Zone Cotiere du Togo. J Rech Sci L'Université Lomé. 15 nov 2021;23(2):11‑25.
12. Diallo A, Zotchi K, Lawson-evi P, Bakoma B, Badjabaissi E, Kwashie EG. Pesticides Use Practice by Market Gardeners in Lome (Togo). J Toxicol. 22 sept 2020;2020:1‑5. https://doi.org/10.1155/2020/8831873 PMid:33029138 PMCid:PMC7528109
13. Badjabaissi E, Aboudoulatif D, Assiki T, Dossou-Yovo K, Assih M, Sanvee S, et al. Analysis of Pesticide Residues in Lomé Vegetables: Implications for Health and Food Consumption. J Pharmacol Toxicol. 15 janv 2024;19:13‑26. https://doi.org/10.3923/jpt.2024.13.26
14. Hossam El Din HA, Abdallah AA, Abdel-Razik RK, Hamed NA, Elshatory A, Awad W, et al. Sex comparison of oxidative stress, mitochondrial dysfunction, and apoptosis triggers induced by single-dose Abamectin in albino rats. Pestic Biochem Physiol. 2024;201:105903. https://doi.org/10.1016/j.pestbp.2024.105903 PMid:38685225
15. Adiguzel C, Karaboduk H, Uzunhisarcikli M. Protective role of melatonin against abamectin-induced biochemical, immunohistochemical, and ultrastructural alterations in the testicular tissues of rats. Microsc Microanal. 2024;30(5):962‑77. https://doi.org/10.1093/mam/ozae080 PMid:39189879
16. Ismail T, Ahmad SN, Ahmad KR, Suleman S, Sitara S, Kanwal MA, et al. Cypermethrin (CYP) and lambda-cyhalothrin (λ-CYH) exposure induced histopathological defects of reproductive organs in pregnant mice. Fluoride. 2023;56:488‑502.
17. Kathim AS, Al-Aitte S. Cypermethrin insecticide induced histopathological alteration in testis and ovary of males and female mice. Tex J Agric Biol Sci. 2022;6:10‑7.
18. Kotb GA, Ziada RM, Farag AAG. Acute Abamectin exposure induces oxidative stress responses in liver of male albino rats. Egypt Acad J Biol Sci F Toxicol Pest Control. 2021;13(1):71‑81. https://doi.org/10.21608/eajbsf.2021.142524
19. Alalwani AD. Nephrotoxicity of cypermethrin in rats. Histopathological aspects. Histol Histopathol. 2020;35(12):1437‑48.
20. Moqbel FS, Al-Eryani MA, Abd Al Galil FM. Histopathological and biochemical effects of abamectin on kidney in male albino rats. J Entomol Zool Stud. 2017;5:245‑9.
21. Jaafer NS, Rabee AM. Effect of Cypermethrin on Hematological and Histological Parameters in Male Albino Mice | Ibn AL-Haitham Journal For Pure and Applied Sciences. Ibn AL-Haitham J Pure Appl Sci. 2025;38(3):68‑78. https://doi.org/10.30526/38.3.3481
22. Tavakkoli M, Dadkhah M, Saadati H, Afshari S, Mostafalou S. Neurobehavioral toxicity of cypermethrin in association with oxidative, inflammatory and neurotrophic changes in the hippocampus of rats. Int J Environ Health Res. 8 mai 2025;1‑12. https://doi.org/10.1080/09603123.2025.2503472 PMid:40338169
23. Ibrahim KA, Eleyan M, Khwanes SA, Mohamed RA, Ayesh BM. Alpha-mangostin attenuates the apoptotic pathway of abamectin in the fetal rats' brain by targeting pro-oxidant stimulus, catecholaminergic neurotransmitters, and transcriptional regulation of reelin and nestin. Drug Chem Toxicol. 2 nov 2022;45(6):2496‑508. https://doi.org/10.1080/01480545.2021.1960856 PMid:34338122
24. Ahmad L, Gul ST, Saleemi MK, Hussain R, Naqvi SNH, Du X xia, et al. The effect of different repeated doses of cypermethrin on the behavioral and histological alterations in the brain of rabbits (Oryctolagus cuniculi). 2021 [cité 30 oct 2025]; https://www.academia.edu/download/87290397/IJVS_2021_10_347_354.pdf https://doi.org/10.47278/journal.ijvs/2021.092 PMid:34776615
25. Radi AM, Mohammed ET, Abushouk AI, Aleya L, Abdel-Daim MM. The effects of abamectin on oxidative stress and gene expression in rat liver and brain tissues: Modulation by sesame oil and ascorbic acid. Sci Total Environ. janv 2020;701:134882. https://doi.org/10.1016/j.scitotenv.2019.134882 PMid:31739238
26. Woodward KN. GABAergic Neurotransmission and Toxicity 2: Macrocyclic Lactones. 2024 [cité 30 oct 2025]; Disponible sur: https://books.rsc.org/books/edited-volume/2189/chapter/7986729 https://doi.org/10.1039/9781839165795-00179
27. Gur C, Kandemir O, Kandemir FM. Investigation of the effects of hesperidin administration on abamectin‐induced testicular toxicity in rats through oxidative stress, endoplasmic reticulum stress, inflammation, apoptosis, autophagy, and JAK2 / STAT3 pathways. Environ Toxicol. mars 2022;37(3):401‑12. https://doi.org/10.1002/tox.23406 PMid:34748272
28. Hong Y, Huang Y, Yang X, Zhang J, Li L, Huang Q, et al. Abamectin at environmentally-realistic concentrations cause oxidative stress and genotoxic damage in juvenile fish (Schizothorax prenanti). Aquat Toxicol. 2020;225:105528. https://doi.org/10.1016/j.aquatox.2020.105528 PMid:32569996
29. Celik-Ozenci C, Tasatargil A, Tekcan M, Sati L, Gungor E, Isbir M, et al. Effects of abamectin exposure on male fertility in rats: potential role of oxidative stress-mediated poly (ADP-ribose) polymerase (PARP) activation. Regul Toxicol Pharmacol. 2011;61(3):310‑7. https://doi.org/10.1016/j.yrtph.2011.09.001 PMid:21945325
30. Liang Y, Dong B, Pang N, Hu J. ROS generation and DNA damage contribute to abamectin-induced cytotoxicity in mouse macrophage cells. Chemosphere. 1 nov 2019;234:328‑37. https://doi.org/10.1016/j.chemosphere.2019.06.031 PMid:31229705
31. BUG TG. Pests and pest control. Cent Excell Mar Biol Univ Karachi Karachi [Internet]. 2015 [cité 30 oct 2025];126. Disponible sur: https://www.zsp.com.pk/PRO2015.pdf#page=161
32. Kweon J, Lim W, Lee H, Kim J, Song G, Jeong W, et al. Cypermethrin triggers oxidative stress, apoptosis, and inflammation in bovine mammary glands by disruption of mitogen-activated protein kinase (MAPK) pathways and calcium homeostasis. Reprod Toxicol. 2025;132:108842. https://doi.org/10.1016/j.reprotox.2025.108842 PMid:39884399
33. Zhao Y, Ye X, Xiong Z, Ihsan A, Ares I, Martínez M, et al. Cancer metabolism: the role of ROS in DNA damage and induction of apoptosis in cancer cells. Metabolites. 2023;13(7):796. https://doi.org/10.3390/metabo13070796 PMid:37512503 PMCid:PMC10383295
34. Sule RO, Condon L, Gomes AV. A Common Feature of Pesticides: Oxidative Stress-The Role of Oxidative Stress in Pesticide‐Induced Toxicity. Zhou X, éditeur. Oxid Med Cell Longev. janv 2022;2022(1):5563759. https://doi.org/10.1155/2022/5563759 PMid:35096268 PMCid:PMC8791758
35. Jabłońska-Trypuć A. Pesticides as inducers of oxidative stress. React Oxyg Species. 2017;3(8):96‑110. https://doi.org/10.20455/ros.2017.823
36. Eskenazi B, Rosas LG, Marks AR, Bradman A, Harley K, Holland N, et al. Pesticide Toxicity and the Developing Brain. Basic Clin Pharmacol Toxicol. févr 2008;102(2):228‑36. PMid:18226078 https://doi.org/10.1111/j.1742-7843.2007.00171.x
37. Modgil S, Lahiri DK, Sharma VL, Anand A. Role of early life exposure and environment on neurodegeneration: implications on brain disorders. Transl Neurodegener. déc 2014;3(1):9. https://doi.org/10.1186/2047-9158-3-9 PMid:24847438 PMCid:PMC4028099
38. Borgert CJ, Quill TF, McCarty LS, Mason AM. Can mode of action predict mixture toxicity for risk assessment? Toxicol Appl Pharmacol. 2004;201(2):85‑96. https://doi.org/10.1016/j.taap.2004.05.005 PMid:15541748
39. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72(1‑2):248‑54. https://doi.org/10.1006/abio.1976.9999 PMid:942051
40. Patlolla AK, Barnes C, Yedjou C, Velma VR, Tchounwou PB. Oxidative stress, DNA damage, and antioxidant enzyme activity induced by hexavalent chromium in Sprague‐Dawley rats. Environ Toxicol. févr 2009;24(1):66‑73. https://doi.org/10.1002/tox.20395 PMid:18508361 PMCid:PMC2769560
41. Sedlak J, Lindsay HR. Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman's reagent. Anal Biochem. 1968;25:192‑205. https://doi.org/10.1016/0003-2697(68)90092-4 PMid:4973948
42. Habig WH, Pabst MJ, Jakoby WB. Glutathione S-transferases: the first enzymatic step in mercapturic acid formation. J Biol Chem. 1974;249(22):7130‑9. https://doi.org/10.1016/S0021-9258(19)42083-8 PMid:4436300
43. Aebi H. [13] Catalase in vitro. In: Methods in enzymology [Internet]. Elsevier; 1984 [cité 30 oct 2025]. p. 121‑6. Disponible sur: https://www.sciencedirect.com/science/article/pii/S0076687984050163 https://doi.org/10.1016/S0076-6879(84)05016-3 PMid:6727660
44. Marklund S, Marklund G. Involvement of the superoxide anion radical in the autoxidation of pyrogallol and a convenient assay for superoxide dismutase. Eur J Biochem. 1974;47(3):469‑74. https://doi.org/10.1111/j.1432-1033.1974.tb03714.x PMid:4215654
45. Hassan I, Chibber S, Khan AA, Naseem I. Cisplatin-Induced Neurotoxicity In Vivo Can Be Alleviated by Riboflavin Under Photoillumination. Cancer Biother Radiopharm. mars 2013;28(2):160‑8. https://doi.org/10.1089/cbr.2012.1312 PMid:23215961
46. Baldi I, Cordier S, Coumoul X, Elbaz A, Gamet-Payrastre L, Lebailly P, et al. Pesticides: Effets sur la santé [Doctoral dissertation, Institut national de la santé et de la recherche médicale]. INSERM; 2013.
47. Slotkin TA, Tate CA, Ryde IT, Levin ED, Seidler FJ. Organophosphate Insecticides Target the Serotonergic System in Developing Rat Brain Regions: Disparate Effects of Diazinon and Parathion at Doses Spanning the Threshold for Cholinesterase Inhibition. Environ Health Perspect. oct 2006;114(10):1542‑6. https://doi.org/10.1289/ehp.9337 PMid:17035140 PMCid:PMC1626396
48. Feoli AM, Siqueira IR, Almeida L, Tramontina AC, Vanzella C, Sbaraini S, et al. Effects of protein malnutrition on oxidative status in rat brain. Nutrition. 2006;22(2):160‑5. https://doi.org/10.1016/j.nut.2005.06.007 PMid:16459228
49. Mayne ST. Antioxidant Nutrients and Chronic Disease: Use of Biomarkers of Exposure and Oxidative Stress Status in Epidemiologic Research. J Nutr. 1 mars 2003;133(3):933S-940S. https://doi.org/10.1093/jn/133.3.933S PMid:12612179
50. Stegeman JJ, Brouwer B, Di Giulio RT, Lars F, Fowler BA, Sanders BM, et al. Molecular responses to environmental contamination: enzyme and protein systems as indicators of chemical exposure and effect. In: Molecular responses to environmental contamination: enzyme and protein systems as indicators of chemical exposure and effect. CRC Press; 2018. p. 235‑336. (In Biomarkers). https://doi.org/10.1201/9781351070270-7 PMid:29503637 PMCid:PMC5820305
51. Meredith SC. Protein Denaturation and Aggregation: Cellular Responses to Denatured and Aggregated Proteins. Ann N Y Acad Sci. mars 2006;1066(1):181‑221. https://doi.org/10.1196/annals.1363.030 PMid:16533927
52. Yang L, Chen JH, Xu T, Zhou AS, Yang HK. Rice protein improves oxidative stress by regulating glutathione metabolism and attenuating oxidative damage to lipids and proteins in rats. Life Sci. 2012;91(11‑12):389‑94. https://doi.org/10.1016/j.lfs.2012.08.003 PMid:22906634
53. Tauffenberger A, Magistretti PJ. Reactive Oxygen Species: Beyond Their Reactive Behavior. Neurochem Res. janv 2021;46(1):77‑87. https://doi.org/10.1007/s11064-020-03208-7 PMid:33439432 PMCid:PMC7829243
54. Mailloux RJ. An Update on Mitochondrial Reactive Oxygen Species Production. Antioxidants. juin 2020;9(6):472. https://doi.org/10.3390/antiox9060472 PMid:32498250 PMCid:PMC7346187
55. Wang X, Martínez MA, Dai M, Chen D, Ares I, Romero A, et al. Permethrin-induced oxidative stress and toxicity and metabolism. A review. Environ Res. 2016;149:86‑104. https://doi.org/10.1016/j.envres.2016.05.003 PMid:27183507
56. Bhardwaj JK, Mittal M, Saraf P, Kumari P. Pesticides induced oxidative stress and female infertility: a review. Toxin Rev. 2 janv 2020;39(1):1‑13. https://doi.org/10.1080/15569543.2018.1474926
57. Aoun M, Tiranti V. Mitochondria: a crossroads for lipid metabolism defect in neurodegeneration with brain iron accumulation diseases. Int J Biochem Cell Biol. 2015;63:25‑31. https://doi.org/10.1016/j.biocel.2015.01.018 PMid:25668476
58. Georgiou-Siafis SK, Tsiftsoglou AS. The key role of GSH in keeping the redox balance in mammalian cells: mechanisms and significance of GSH in detoxification via formation of conjugates. Antioxidants. 2023;12(11):1953. PMid:38001806 PMCid:PMC10669396 https://doi.org/10.3390/antiox12111953
59. Rjeibi I, Ben Saad A, Hfaiedh N. Oxidative damage and hepatotoxicity associated with deltamethrin in rats: The protective effects of Amaranthus spinosus seed extract. Biomed Pharmacother. 1 déc 2016;84:853‑60. https://doi.org/10.1016/j.biopha.2016.10.010 PMid:27728895
60. Raj Rai S, Bhattacharyya C, Sarkar A, Chakraborty S, Sircar E, Dutta S, et al. Glutathione: Role in Oxidative/Nitrosative Stress, Antioxidant Defense, and Treatments. ChemistrySelect. 14 mai 2021;6(18):4566‑90. https://doi.org/10.1002/slct.202100773
61. Bekkairi S, Bouchiha H, Gasmi S, Rouabhi R, Boualleg O. Abamectin-induced neurotoxicity and preventive effect of Ephedra essential oils against this toxicity in rats. Stud Eng Exact Sci. 2024;5(3):e12359‑e12359. https://doi.org/10.54021/seesv5n3-004
62. Sharma P, Firdous S, Singh R. Neurotoxic effect of cypermethrin and protective role of resveratrol in Wistar rats. Int J Nutr Pharmacol Neurol Dis. 2014;4(2):104. https://doi.org/10.4103/2231-0738.129598
63. Vaish S, Gupta D, Mehrotra R, Mehrotra S, Basantani MK. Glutathione S-transferase: a versatile protein family. 3 Biotech. juill 2020;10(7):321. https://doi.org/10.1007/s13205-020-02312-3 PMid:32656054 PMCid:PMC7320970
64. Sauer E, Moro AM, Brucker N, Nascimento S, Gauer B, Fracasso R, et al. Liver δ-Aminolevulinate Dehydratase Activity is Inhibited by Neonicotinoids and Restored by Antioxidant Agents. Int J Environ Res Public Health. nov 2014;11(11):11676‑90. https://doi.org/10.3390/ijerph111111676 PMid:25402564 PMCid:PMC4245637
65. Board PG, Menon D. Glutathione transferases, regulators of cellular metabolism and physiology. Biochim Biophys Acta Bba-Gen Subj. 2013;1830(5):3267‑88. https://doi.org/10.1016/j.bbagen.2012.11.019 PMid:23201197
66. Ademi M. CATALASE ACTIVITY IN INTERACTION WITH IONIZED WATER AND OTHER ANTIOXIDANTS IN BLOOD PLASMA, LIVER, AND KIDNEY OF THE RAT DURING HYPERTHERMIC STRESS. MEDIS - Int J Med Sci Res. 14 déc 2022;1:21‑5. https://doi.org/10.35120/medisij010421a
67. Madkour L. Citation: Loutfy H Madkour. Function of Reactive Oxygen Species (ROS) Inside the Living Organisms and Sources of Oxidants Function of Reactive Oxygen Species (ROS) Inside the Living Organisms and Sources of Oxidants. J Glob Pharma Technol. 3 juill 2019;2:1‑23.
68. Ighodaro OM, Akinloye OA. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alex J Med. 2018;54(4):287‑93. https://doi.org/10.1016/j.ajme.2017.09.001
69. Ileriturk M, Kandemir O, Kandemir FM. Evaluation of protective effects of quercetin against cypermethrin‐induced lung toxicity in rats via oxidative stress, inflammation, apoptosis, autophagy, and endoplasmic reticulum stress pathway. Environ Toxicol. nov 2022;37(11):2639‑50. https://doi.org/10.1002/tox.23624 PMid:35876585
70. Abd El-Moneim Ibrahim K, Mohamed Abdelrahman S, K. A. Elhakim H, Ali Ragab E. Single or combined exposure to chlorpyrifos and cypermethrin provoke oxidative stress and downregulation in monoamine oxidase and acetylcholinesterase gene expression of the rat's brain. Environ Sci Pollut Res. avr 2020;27(11):12692‑703. https://doi.org/10.1007/s11356-020-07864-8 PMid:32006337
71. Adegbesan BO, Adenuga GA. Effect of lead exposure on liver lipid peroxidative and antioxidant defense systems of protein-undernourished rats. Biol Trace Elem Res. mai 2007;116(2):219‑25. https://doi.org/10.1007/s12011-007-9029-8 PMid:17646689
72. Yekti R, Bukhari A, Jafar N, Thaha AR. Measurement of malondialdehyde (MDA) as a good indicator of lipid peroxidation. Int J Allied Med Sci Clin Res IJAMSCR. 2018;6(4):1‑3.
73. Valgimigli L. Lipid peroxidation and antioxidant protection. Biomolecules. 2023;13(9):1291. PMid:37759691 PMCid:PMC10526874 https://doi.org/10.3390/biom13091291
74. Alsemeh A, AbdelRahman M, Mahmoud F, Abd El Fattah E. Neurotoxicity of Cypermethrin on Rat Cerebral Cortex and the Alleviated effect of Hydroxytyrosol. J Med Histol. 2020;4(2):214‑36. https://doi.org/10.21608/jmh.2021.54223.1087
75. Ali HF. Cellular Mechanism involved in cypermethrin induced neurotoxicity. Rec Pharm Biomed Sci. 2020;4(1):32‑9. https://doi.org/10.21608/rpbs.2019.17969.1043
76. Wang B, Wang Y, Zhang J, Hu C, Jiang J, Li Y, et al. ROS-induced lipid peroxidation modulates cell death outcome: mechanisms behind apoptosis, autophagy, and ferroptosis. Arch Toxicol. juin 2023;97(6):1439‑51. https://doi.org/10.1007/s00204-023-03476-6 PMid:37127681
77. Bayır H, Anthonymuthu TS, Tyurina YY, Patel SJ, Amoscato AA, Lamade AM, et al. Achieving life through death: redox biology of lipid peroxidation in ferroptosis. Cell Chem Biol. 2020;27(4):387‑408. https://doi.org/10.1016/j.chembiol.2020.03.014 PMid:32275865 PMCid:PMC7218794
78. Gęgotek A, Skrzydlewska E. Biological effect of protein modifications by lipid peroxidation products. Chem Phys Lipids. 2019;221:46‑52. PMid:30922835 https://doi.org/10.1016/j.chemphyslip.2019.03.011
79. Abramov AY, Potapova EV, Dremin VV, Dunaev AV. Interaction of oxidative stress and misfolded proteins in the mechanism of neurodegeneration. Life. 2020;10(7):101. https://doi.org/10.3390/life10070101 PMid:32629809 PMCid:PMC7400128
80. Gandhi S, Abramov AY. Mechanism of Oxidative Stress in Neurodegeneration. Oxid Med Cell Longev. 2012;2012:1‑11. https://doi.org/10.1155/2012/428010 PMid:22685618 PMCid:PMC3362933
81. Boudefar S, Sayoud A. Etude de la neurotoxicité liée au stress oxydant induit par les pyréthrinoides chez le rat wistar. [PhD Thesis]. université de jijel; 2017.
82. Abdel-Razik RK, Hamed NA. Deleterious effect of abamectin on rat brain mitochondria. Alex Sci Exch J. 2015;36(OCTOBER-DECEMBER):423‑8. https://doi.org/10.21608/asejaiqjsae.2015.2964
83. DiMauro S, Schon EA. Mitochondrial Disorders in the Nervous System. Annu Rev Neurosci. 1 juill 2008;31(1):91‑123. https://doi.org/10.1146/annurev.neuro.30.051606.094302 PMid:18333761
84. Sarailoo M, Afshari S, Asghariazar V, Safarzadeh E, Dadkhah M. Cognitive Impairment and Neurodegenerative Diseases Development Associated with Organophosphate Pesticides Exposure: a Review Study. Neurotox Res. oct 2022;40(5):1624‑43. https://doi.org/10.1007/s12640-022-00552-0 PMid:36066747
85. Rodrigues JA, Narasimhamurthy RK, Joshi MB, Dsouza HS, Mumbrekar KD. Pesticides Exposure-Induced Changes in Brain Metabolome: Implications in the Pathogenesis of Neurodegenerative Disorders. Neurotox Res. oct 2022;40(5):1539‑52. https://doi.org/10.1007/s12640-022-00534-2 PMid:35781222 PMCid:PMC9515138
86. Ghosh N, Das A, Chaffee S, Roy S, Sen CK. Reactive oxygen species, oxidative damage and cell death. In: Immunity and inflammation in health and disease [Internet]. Elsevier; 2018 [cité 30 oct 2025]. p. 45‑55. https://www.sciencedirect.com/science/article/pii/B9780128054178000044 https://doi.org/10.1016/B978-0-12-805417-8.00004-4
87. Obeng E. Apoptosis (programmed cell death) and its signals - A review. Braz J Biol. 2021;81:1133‑43. https://doi.org/10.1590/1519-6984.228437 PMid:33111928
88. Nagata S, Tanaka M. Programmed cell death and the immune system. Nat Rev Immunol. mai 2017;17(5):333‑40. https://doi.org/10.1038/nri.2016.153 PMid:28163302
89. Chipuk JE, Green DR. Do inducers of apoptosis trigger caspase-independent cell death? Nat Rev Mol Cell Biol. 2005;6(3):268‑75. https://doi.org/10.1038/nrm1573 PMid:15714200
90. Kumar V, Sami N, Kashav T, Islam A, Ahmad F, Hassan MI. Protein aggregation and neurodegenerative diseases: From theory to therapy. Eur J Med Chem. 29 nov 2016;124:1105‑20. https://doi.org/10.1016/j.ejmech.2016.07.054 PMid:27486076
91. Borza LR. A review on the cause-effect relationship between oxidative stress and toxic proteins in the pathogenesis of neurodegenerative diseases. Med-Surg J. 2014;118(1):19‑27.
92. Alqahtani T, Deore SL, Kide AA, Shende BA, Sharma R, Chakole RD, et al. Mitochondrial dysfunction and oxidative stress in Alzheimer's disease, and Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis-an updated review. Mitochondrion. 2023;71:83‑92. https://doi.org/10.1016/j.mito.2023.05.007 PMid:37269968
93. Erekat NS. Apoptosis and its therapeutic implications in neurodegenerative diseases. Clin Anat. janv 2022;35(1):65‑78. https://doi.org/10.1002/ca.23792 PMid:34558138
94. Herawati IE, Lesmana R, Levita J, Subarnas A. Molecular interaction of ricin-a with caspase-3, caspase-8, caspase-9 and autophagy-related gene5 (ATG5) to understand its role as anticancer agent. Chem. 2021;14(3):1790‑4. https://doi.org/10.31788/RJC.2021.1436298
95. Morris G, Berk M. The many roads to mitochondrial dysfunction in neuroimmune and neuropsychiatric disorders. BMC Med. déc 2015;13(1):68. https://doi.org/10.1186/s12916-015-0310-y PMid:25889215 PMCid:PMC4382850
96. Unnisa A, Greig NH, Kamal MA. Inhibition of Caspase 3 and Caspase 9 Mediated Apoptosis: A MultimodalTherapeutic Target in Traumatic Brain Injury. Curr Neuropharmacol. avr 2023;21(4):1001‑12. https://doi.org/10.2174/1570159X20666220327222921 PMid:35339178 PMCid:PMC10227914
97. Muhammed RE, El-Desouky MA, Abo-Seda SB, Nahas AA, Elhakim HKA, Alkhalaf MI. The protecting role of Moringa oleifera in cypermethrin-induced mitochondrial dysfunction and apoptotic events in rats brain. J King Saud Univ - Sci. sept 2020;32(6):2717‑22. https://doi.org/10.1016/j.jksus.2020.06.006
98. Hussien HM, Abdou HM, Yousef MI. Cypermethrin induced damage in genomic DNA and histopathological changes in brain and haematotoxicity in rats: the protective effect of sesame oil. Brain Res Bull. 2013;92:76‑83. https://doi.org/10.1016/j.brainresbull.2011.10.020 PMid:22085743
99. Ashafaq M, Hussain S, Alshahrani S, Siddiqui R, Alam MI, Elhassan Taha MM, et al. Neuroprotective effects of nano-curcumin against cypermethrin associated oxidative stress and up-regulation of apoptotic and inflammatory gene expression in rat brains. Antioxidants. 2023;12(3):644. https://doi.org/10.3390/antiox12030644 PMid:36978892 PMCid:PMC10045852
100. Strasser A, Vaux DL. Cell death in the origin and treatment of cancer. Mol Cell. 2020;78(6):1045‑54. https://doi.org/10.1016/j.molcel.2020.05.014 PMid:32516599
101. Labi V, Erlacher M. How cell death shapes cancer. Cell Death Dis. 5 mars 2015;6(3):e1675‑e1675. https://doi.org/10.1038/cddis.2015.20 PMid:25741600 PMCid:PMC4385913
102. Nervous System | 22 | v2 | Toxicologic Pathology | Mark T. Butt, Alys [Internet]. [cité 30 oct 2025]. Disponible sur: https://www.taylorfrancis.com/chapters/edit/10.1201/9780429504624-22/nervous-system-mark-butt-alys-bradley-robert-sills
103. Vinters HV, Kleinschmidt-DeMasters BK. General pathology of the central nervous system. In: Greenfield's Neuropathology-Two Volume Set [Internet]. CRC Press; 2018 [cité 30 oct 2025]. p. 25‑82. https://www.taylorfrancis.com/chapters/edit/10.1201/9781315382715-6/general-pathology-central-nervous-system-harry-vinters-bk-kleinschmidt-demasters https://doi.org/10.1201/9781315382715-6 PMid:29124544
104. Astiz M, De Alaniz MJ, Marra CA. Effect of pesticides on cell survival in liver and brain rat tissues. Ecotoxicol Environ Saf. 2009;72(7):2025‑32. https://doi.org/10.1016/j.ecoenv.2009.05.001 PMid:19493570
105. Chalimoniuk M, Jagsz S, Sadowska-Krepa E, Chrapusta SJ, Klapcinska B, Langfort J. Diversity of endurance training effects on antioxidant defenses and oxidative damage in different brain regions of adolescent male rats. J Physiol Pharmacol. 2015;66(4):539‑47.
106. Mohamadin AM, Sheikh B, Abd El-Aal AA, Elberry AA, Al-Abbasi FA. Protective effects of Nigella sativa oil on propoxur-induced toxicity and oxidative stress in rat brain regions. Pestic Biochem Physiol. 2010;98(1):128‑34. https://doi.org/10.1016/j.pestbp.2010.05.011
107. Fujikawa DG. The role of excitotoxic programmed necrosis in acute brain injury. Comput Struct Biotechnol J. 2015;13:212‑21. https://doi.org/10.1016/j.csbj.2015.03.004 PMid:25893083 PMCid:PMC4398818
108. Xiong K, Liao H, Long L, Ding Y, Huang J, Yan J. Necroptosis contributes to methamphetamine-induced cytotoxicity in rat cortical neurons. Toxicol In Vitro. 2016;35:163‑8. https://doi.org/10.1016/j.tiv.2016.06.002 PMid:27288563
109. Sharma D, Sangha GK. Triazophos induced neuro-splenic toxicity and evaluation of antioxidative potential of aqueous Broccoli extract in Wistar albino rats. J Appl Nat Sci. 2021;13(2):616. https://doi.org/10.31018/jans.v13i2.2644
110. Syed F, Chandravanshi LP, Khanna VK, Soni I. Beta-cyfluthrin induced neurobehavioral impairments in adult rats. Chem Biol Interact. 2016;243:19‑28. https://doi.org/10.1016/j.cbi.2015.11.015 PMid:2660415