Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Type of study
Publication year range
1.
J Biochem Mol Toxicol ; 36(8): e23092, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35521929

ABSTRACT

The protective effects of thymol and carvacrol, two phenolic monoterpenes with a wide spectrum of pharmacological effects, against the oxidative stress produced by the di (2-ethylhexyl) phthalate (DEHP) in human embryonic kidney cells 293 cells (HEK-293 cells) were investigated in this study. The cytotoxicity was monitored by cell viability, while oxidative stress generation was assessed by reactive oxygen species (ROS) quantification, antioxidant enzyme activities measurement, glutathione concentration, and malondialdehyde (MDA) quantification. The genotoxicity was evaluated by the measurement of DNA fragmentation through the Comet assay. Our results demonstrated that the pretreatment of HEK-293 cells with thymol or carvacrol, 2 h before DEHP exposure, significantly increased the cell viability, decreased the ROS overproduction, modulated catalase (CAT), and superoxide dismutase (SOD) activities, restored the reduced glutathione content, and reduced the MDA level. The DNA fragmentation was also decreased by thymol and carvacrol pretreatment. These findings suggest that thymol and carvacrol could protect HEK-293 cells from DEHP-induced oxidative stress.


Subject(s)
Cymenes , Diethylhexyl Phthalate , Thymol , Antioxidants/pharmacology , Cymenes/pharmacology , Diethylhexyl Phthalate/toxicity , Glutathione , HEK293 Cells , Humans , Kidney/metabolism , Oxidative Stress , Reactive Oxygen Species , Thymol/pharmacology
2.
Pestic Biochem Physiol ; 174: 104797, 2021 May.
Article in English | MEDLINE | ID: mdl-33838701

ABSTRACT

Tebuconazole (TEB) is a common triazole fungicide that has been widely used for the control of plant pathogenic fungi, suggesting that mammal exposure occurs regularly. Several studies demonstrated that TEB exposure has been linked to a variety of toxic effects, including neurotoxicity, immunotoxicity, reprotoxicity and carcinogenicity. However, there is a few available data regarding the molecular mechanism involved in TEB-induced toxicity. The current study was undertaken to investigate the toxic effects of TEB in HCT116 cells. Our results showed that TEB caused cytotoxicity by inhibiting cell viability as assessed by the MTT assay. Furthermore, we have demonstrated that TEB induced a significant increase in the reactive oxygen species (ROS) production leading to the induction of lipid peroxidation and DNA fragmentation and increased superoxide dismutase (SOD) and catalase (CAT) activities. Moreover, TEB exposure induced mitochondrial membrane potential loss and caspase-9/-3 activation. Treatment with general caspases inhibitor (Z-VAD-fmk) significantly prevented the TEB-induced cell death, indicating that TEB induced caspases-dependent cell death. These findings suggest the involvement of oxidative stress and apoptosis in TEB-induced toxicity in HCT116.


Subject(s)
DNA Damage , Triazoles , Animals , Apoptosis , HCT116 Cells , Humans , Oxidative Stress , Reactive Oxygen Species , Triazoles/toxicity
3.
Chem Biol Interact ; 330: 109114, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32735800

ABSTRACT

Tebuconazole (TEB) is a broad-spectrum conazole fungicide that has been used in agriculture in the control of foliar and soil-borne diseases of many crops. The present study has investigated the adverse effects of subchronic exposure to TEB on the kidney of male rats. Animals were divided into four equal groups and treated with TEB at increasing doses 0.9, 9 and 27 mg/kg body weight for 28 consecutive days. The results showed that TEB induced oxidative stress in the kidney demonstrated by an increase in malondialdehyde (MDA), protein carbonyl (PC), advanced oxidation protein product (AOPP) levels and DNA damage, as compared to the controls. Furthermore, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activities were increased in the renal tissue of treated rats. Moreover, significant decrease in reduced glutathione (GSH) content in TEB-treated rats was observed, while oxidized glutathione (GSSG) levels were increased, thus a marked fall in GSH/GSSG ratio was registered in the kidney. Glutathione reductase (GR) activity showed a significant increase after TEB exposure. Moreover, TEB down-regulated the expression of Bcl2 and up-regulated the expression of Bax and caspase 3, which triggered apoptosis via the Bax/Bcl2 and caspase pathway. Also, TEB administration resulted in altered biochemical indicators of renal function and varying lesions in the overall histo-architecture of renal tissues. Taken together, our findings brought into light the renal toxicity induced by TEB, which was found to be significant at low doses.


Subject(s)
Apoptosis/drug effects , DNA Damage/drug effects , Kidney/pathology , Oxidative Stress/drug effects , Triazoles/toxicity , Animals , Dose-Response Relationship, Drug , Fungicides, Industrial/toxicity , Gene Expression Regulation , Glutathione/metabolism , Glutathione Disulfide/metabolism , Glutathione Reductase/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Oxidation-Reduction , Rats , Rats, Wistar
4.
Cell Stress Chaperones ; 25(6): 919-928, 2020 11.
Article in English | MEDLINE | ID: mdl-32500380

ABSTRACT

Environmental toxicants such as phthalate have been involved in multiple health disorders including renal diseases. Oxidative damage is implicated in many alterations caused by phthalate especially the di(2-ethylhexyl) phthalate (DEHP), which is the most useful phthalate. However, information regarding its mechanism of renal damage is lacking. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates gene expression implicated in free radical scavenging and cytoprotection including the antioxidant glutathione (GSH) pathway. The aim of this study was to assess whether DEHP affects the Nrf2 pathway and the GSH concentration. Mice were divided into four groups: a control group and three groups treated with DEHP at different concentrations (5, 50, and 200 mg/kg body weight) for 30 days. Our results showed that DEHP altered the normal levels of serum biochemical parameters creatinine (CREA), urea, and lactate dehydrogenase (LDH). This phthalate caused oxidative damage through the induction of lipid peroxidation and protein oxidation as marked by increase of protein carbonyl (PC) and loss of protein-bound sulfhydryls (PSH). Simultaneously, DEHP treatment decreased the protein level of Nrf-2, HO-1, and GCLC (responsible of GSH synthesis) and decreased the GSH level. Inhibition of the Nrf2 pathway is related to the activation of the mitochondrial pathway of apoptosis. This apoptotic process is evidenced by an upregulation of p53 and Bax protein levels in addition to a downregulation of Bcl-2. Collectively, our data demonstrated that depletion of Nrf2 and GSH was associated with the elevation of oxidative stress and the activation of intrinsic apoptosis in mouse kidney treated with DEHP.


Subject(s)
Diethylhexyl Phthalate/toxicity , Glutathione/metabolism , Homeostasis , Kidney/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Biomarkers/blood , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/metabolism , Homeostasis/drug effects , Kidney/drug effects , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Mice, Inbred BALB C , Oxidation-Reduction , Protein Carbonylation/drug effects , Signal Transduction/drug effects , Sulfhydryl Compounds/metabolism
5.
Food Chem Toxicol ; 137: 111134, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32006631

ABSTRACT

Tebuconazole is an effective systemic fungicide that belongs to the triazoles family. It has been widely used in both agricultural and medical sectors for the control of fungal diseases. Although TEB poses serious threats to mammals health, studies regarding its cardiotoxicity are very limited. Thus, we aimed to evaluate the effects of TEB on some biochemical parameters, the induction of apoptosis and DNA damage in the heart tissue. Male Wistar rats were treated with TEB at varied oral doses for 28 consecutive days. This study demonstrates that TEB decreased cardiac acetylcholinesterase, increased serum marker enzymes such as creatinine phosphokinase (CPK) and lactate dehydrogenase (LDH), and altered the lipid profile by increasing serum levels of total cholesterol (T-CHOL), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and reduced high-density lipoprotein cholesterol (HDL-C) levels. Furthermore, TEB increased levels of p53 and Bax/Bcl2 ratio, released the cytochrome c into the cytosol and activated caspase-9 and caspase-3. Besides, our results showed that TEB induced genotoxic effects. TEB induced DNA fragmentation and increased the frequency of micronucleated bone marrow cells. Moreover, TEB treatment developed fibrosis in the myocardium. Our results suggest that TEB exposure may affect myocardial cells normal functioning and triggers apoptosis.


Subject(s)
Cardiotoxicity/etiology , Fungicides, Industrial/toxicity , Triazoles/toxicity , Animals , Apoptosis/drug effects , Cardiotoxicity/genetics , Cardiotoxicity/metabolism , Cardiotoxicity/physiopathology , Cholesterol, LDL/metabolism , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Humans , Male , Rats , Rats, Wistar , Triglycerides/metabolism
6.
Environ Sci Pollut Res Int ; 27(8): 7957-7966, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31893363

ABSTRACT

The increased use of pesticides is the origin of multiple damages to the environment and to humans; thus, the search for new strategies to reduce or even protect the toxic effects caused by these synthetic products became a necessity. In this context, our study attempted to evaluate the protective effects of fennel essential oil (FEO), the main essential oil extracted from Faeniculum vulgare Mill., a plant with aromatic, flavorful, and medicinal uses, against toxicity induced by an insecticide-triflumuron (TFM)-in human carcinoma cells (HCT116). Our methodological approach consists of the cytotoxicity assay starting with the cell viability test, the ROS generation, the malondialdehyde (MDA) production, the DNA fragmentation, and the measurement of some antioxidant enzymes activities such as catalase (CAT) and superoxide dismutase (SOD). Also, we measured the mitochondrial transmembrane potential. The outcome of the current study showed clearly that after 2 h of HCT 116 cell pretreatment with FEO, there were increase in cell viability, reduction in ROS generation, and modulation in CAT and SOD activities induced by TFM. In the same manner, significant decreases in MDA levels were found. Mainly, the results indicated a perceptible decrease in DNA damages and a significant reduction in the mitochondrial membrane potential loss. Our work demonstrates that FEO can be an important protector against toxic effects induced by TFM in HCT 116 cells.


Subject(s)
Antioxidants/chemistry , Benzamides/chemistry , Catalase/chemistry , Colonic Neoplasms/physiopathology , Foeniculum , Insecticides , Oils, Volatile , Superoxide Dismutase/chemistry , Benzamides/toxicity , Catalase/metabolism , Colonic Neoplasms/chemistry , DNA Damage , Humans , Oxidative Stress
7.
Environ Toxicol ; 35(1): 78-86, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31486570

ABSTRACT

Di (2-ethylhexyl) phthalate (DEHP) is a plasticizer that gives flexibility to various polyvinyl chloride products. It is a pollutant easily released into the environment and can cause many adverse effects to living organisms including hepatotoxicity. The thioredoxin system is a determining factor in the redox balance maintaining in the liver, which is a vulnerable tissue of reactive oxygen species overproduction because of its high energy needs. In order to determine if the thioredoxin system is a target in the development of DEHP hepatotoxicity, Balb/c mice were administered with DEHP intraperitoneally daily for 30 days. Results demonstrated that after DEHP exposure, biochemical profile changes were observed. This phthalate causes oxidative damage through the induction of lipid peroxydation as well as the increase of superoxide dismutase and catalase activities. As new evidence provided in this study, we demonstrated that the DEHP affected the thioredoxin system by altering the expression and the activity of thioredoxin (Trx) and thioredoxin Reductase (TrxR1). The two enzyme activities of the oxidative phase of the pentose phosphate pathway: Glucose-6-phosphate dehydrogenase and 6-Phosphogluconate dehydrogenase were also affected by this phthalate. This leads to a decrease in the level of nicotinamide adenine dinucleotide phosphate used by the TrxR1 to maintain the regeneration of the reduced Trx. We also demonstrated that such effects can be responsible of DEHP-induced DNA damage.


Subject(s)
Diethylhexyl Phthalate/toxicity , Environmental Pollutants/toxicity , Liver/drug effects , Oxidative Stress/drug effects , Pentose Phosphate Pathway/drug effects , Plasticizers/toxicity , Thioredoxins/metabolism , Animals , DNA Damage , Dose-Response Relationship, Drug , Glucosephosphate Dehydrogenase/metabolism , Injections, Intraperitoneal , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
8.
Cell Stress Chaperones ; 25(1): 151-162, 2020 01.
Article in English | MEDLINE | ID: mdl-31848848

ABSTRACT

Phthalates, particularly di(2-ethylhexyl) phthalate (DEHP), are compounds widely used as plasticizers and have become serious global contaminants. Because of the bioaccumulation of such substances, the food chain is at risk. The food contamination by some phthalates has been linked to different side effects in experimental animals. That is why we have chosen the intestinal system's cells which represent the primary targets of these compounds to test their toxic effects. Human colon carcinoma cells (HCT 116) were chosen to elucidate whether DEHP triggers oxidative stress and apoptosis. Our results indicated that DEHP is cytotoxic; it induces the overexpression of Hsp70 protein and causes oxidative damage through the generation of free radicals leading to lipid peroxidation induction and the increase of superoxide dismutase (SOD) and catalase (CAT) activities. In addition, cell treatment with DEHP resulted in a glutathione (GSH) content decrease and a decrease in the glutathione reductase (GR) activity. As new evidence provided in this study, we demonstrated that the DEHP affected the two enzymes' activities of the oxidative phase of the pentose phosphate pathway: Glucose-6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD). This leads to a decrease in the level of NADPH used by the GR to maintain the regeneration of the reduced GSH. We also demonstrated that such effects can be responsible for DEHP-induced apoptosis.


Subject(s)
Carcinoma/drug therapy , Diethylhexyl Phthalate/pharmacology , Glutathione/drug effects , Pentose Phosphate Pathway/drug effects , Regeneration/drug effects , Antioxidants/metabolism , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phthalic Acids/pharmacology , Plasticizers/pharmacology , Superoxide Dismutase/metabolism
9.
Toxicol Mech Methods ; 29(8): 580-586, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31170869

ABSTRACT

Neonicotinoids are a group of pesticides widely used in agriculture and at home. Among those pesticides, acetamiprid (ACM) is a broad-spectrum insecticide used for the protection of vegetables and fruits from pest. The extensive use of this pesticide had led to contamination of environment including soil, water, as well as food products. However, there are few informations regarding the molecular mechanism by which ACM exerts its cytotoxic and genotoxic effects. The aim of the present study was to investigate the toxic effects of ACM in PC12 cells. We demonstrated that ACM significantly decreased cell viability as assessed by the MTT assay. We also shown that ACM-induced reactive oxygen species (ROS) generation followed by lipid peroxidation as evidenced by an increase in the MDA levels. The increase in cell death was accompanied by a reduction in the mitochondrial membrane potential. Besides, pretreatment with Z-VAD-FMK, a general caspases inhibitor, significantly decreased the ACM-induced cell death. Our results also indicate that ACM induced a concentration-dependent increase in DNA damage as evident by the Comet assay. These data indicate that ACM produces cytotoxicity and DNA damage in mammalian cells. Highlights ACM is cytotoxic toward rat pheochromocytoma adrenal medulla cells (PC12). ACM induces ROS generation, lipid peroxidation, and DNA fragmentation. ACM induces caspase-dependent apoptosis in PC12 cells.


Subject(s)
DNA Damage , Environmental Pollutants/toxicity , Insecticides/toxicity , Neonicotinoids/toxicity , Animals , Cell Survival/drug effects , Comet Assay , Dose-Response Relationship, Drug , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
10.
Environ Sci Pollut Res Int ; 26(8): 7540-7549, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30659488

ABSTRACT

Because of the extensive use of phthalates for domestic, medical, and industrial applications, the evaluation of their toxic effects is of major concern to public health. The aim of the present study was to assess the propensity of di (2-ethylhexyl) phthalate (DEHP), one of the most used phthalates, to cause oxidative cardiac damage in mice. DEHP was administered intraperitoneally at doses of 5, 50, and 200 mg/kg body weight for 30 consecutive days in BALB/c mice. We assessed the effect of DEHP on cardiac injury using biochemical profile (such as aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase (LDH), creatinine phosphokinase (CPK), total cholesterol (T-CHOL), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C)), parameters related to myocardiac oxidative stress, such as malondialdehyde (MDA) level, protein carbonyl (PC) concentration, and DNA fragmentation. In addition, we evaluated antioxidant status; enzymatic (catalase (CAT) and superoxide dismutase (SOD) activities) and non-enzymatic (protein-bound sulfhydryl concentration (PSH)) antioxidants. Acetylcholinesterase (AChE) activity and histopathological changes were also assessed in heart mice treated with DEHP. Our results showed that DEHP induced an elevation of serum marker enzymes and perturbated the lipid profile. In addition, this phthalate increased lipid peroxidation, protein carbonyl levels, and DNA fragmentation in the heart in a dose-dependent manner. Antioxidant status was also perturbated by the increase of the CAT and SOD activities and the decrease of the protein-bound sulfhydryl concentration. AChE activity was also inhibited in the heart following the treatment with DEHP. These biochemical alterations were also confirmed by histopathological changes. Increased free radical production at various doses of DEHP would result in impairment of the redox status leading to an enhanced dose-dependent cardiotoxicity.


Subject(s)
Diethylhexyl Phthalate/toxicity , Hazardous Substances/toxicity , Heart Diseases/chemically induced , Heart/drug effects , Animals , Antioxidants , Catalase , Lipid Peroxidation , Male , Malondialdehyde , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Oxidative Stress , Phthalic Acids , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...