1
|
Ribeiro IML, Albuquerque ALS, Vieira LG, Oliveira LAM, Bezerra FS, Teixeira RR, de Menezes RCA, Nogueira KDOPC. Protective role of isobenzofuran-1(3H)-one derivative against neuronal injury and enzyme activity dysregulation induced by bipyridyl herbicide in adult male rats. Neurotoxicology 2025; 108:368-376. [PMID: 40345337 DOI: 10.1016/j.neuro.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/17/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Diquat (DQT) is a bipyridyl herbicide widely used in agriculture. The exposure to DQT may overwhelm the antioxidant system, leading to oxidative damage in organs such as the brain, liver, and kidneys. The brain is particularly susceptible to oxidative damage because of its high oxygen consumption, relatively low antioxidant levels, and limited regenerative capacity. Faced with stressful agents, using compounds capable of repairing and replacing oxidized biomolecules is a promising option to maintain redox homeostasis. Isobenzofuran-1 (3H)-one derivative (named compound 1) emerges as a potential candidate to control the redox imbalance induced by DQT in the brain. The present study aimed to explore the neurotoxicity mechanisms of DQT and the antioxidant potential of compound 1 within the hippocampus and cortex, brain regions impacted by AD, using in vitro and in vivo models. Treatment with compound 1 reduced intracellular levels of ROS and lipid peroxidation in primary cultures of hippocampal neurons caused by DQT. Moreover, compound 1 significantly elevated GSH levels in the brains of rats subjected to DQT treatment. Treatment with compound 1 also decreased lipid peroxidation and carbonylated protein levels in the brain induced by DQT. The exposure to DQT resulted in neuronal injury in the hippocampus. Compound 1, protected against neuronal damage in CA1 and dentate gyrus hippocampal regions. The findings of our study suggest that DQT leads to oxidative damage and neuronal death in both the hippocampus and the cortex. Furthermore, compound 1 exhibits neuroprotective qualities by reducing oxidative damage caused by exposure to DQT.
Collapse
Affiliation(s)
| | | | - Lucas Gabriel Vieira
- Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, MG, Brazil
| | - Laser Antonio Machado Oliveira
- Laboratory of Neurobiology and Biomaterials, Federal University of Ouro Preto, MG, Brazil; Department of Biological Science, Federal University of Ouro Preto, MG, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Physiology, Federal University of Ouro Preto, MG, Brazil; Department of Biological Science, Federal University of Ouro Preto, MG, Brazil
| | | | - Rodrigo Cunha Alvim de Menezes
- Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, MG, Brazil; Department of Biological Science, Federal University of Ouro Preto, MG, Brazil
| | - Katiane de Oliveira Pinto Coelho Nogueira
- Laboratory of Neurobiology and Biomaterials, Federal University of Ouro Preto, MG, Brazil; Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, MG, Brazil; Laboratory of Experimental Physiology, Federal University of Ouro Preto, MG, Brazil; Department of Biological Science, Federal University of Ouro Preto, MG, Brazil.
| |
Collapse
|
2
|
Wang B, Yin Z, Liu J, Tang C, Zhang Y, Wang L, Li H, Luo Y. Diquat Induces Cell Death and dopamine Neuron Loss via Reactive Oxygen Species Generation in Caenorhabditis elegans. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:152-162. [PMID: 39745087 PMCID: PMC11740995 DOI: 10.1021/acs.est.4c07783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025]
Abstract
Diquat (DQ), a contact herbicide extensively utilized in both agricultural and nonagricultural domains, exhibits a high correlation with neuronal disorders. Nevertheless, the toxicity and underlying mechanisms associated with exposure to environmental concentrations of DQ remain ambiguous. Here, we report dose-dependent cellular neurotoxicity of DQ in Caenorhabditis elegans. First, DQ significantly compromised the development and brood size of worms, shortened the lifespan, and caused epidermal abnormalities. An unbiased transcriptomic analysis disclosed several pathways related to cell death and peroxisome homeostasis underlying this organismal-level toxicity. Moreover, exposure of DQ to C. elegans led to a notable increase of embryonic cell death. Concurrently, DQ exposure specifically caused the loss of dopamine neurons but not two other types of neurons in adulthood, which is in accordance with DQ-induced muscle-related defects such as pharyngeal pumping, body bends, and head thrashes. Mechanistically, DQ exposure induces the generation of reactive oxygen species (ROS) and enhances glutathione-related ROS scavenging pathway. Protein levels and activities of mitochondrial electron transport chain complexes were specifically impaired in the DQ-treated worms. Collectively, this study suggests an ROS-mediated cell death pathway involving the neuronal and behavioral toxicity of DQ, which offers a novel mitochondria-related perspective to elucidate the general toxicity caused by a widely distributed herbicide, DQ, at near-environment concentrations.
Collapse
Affiliation(s)
- Bing Wang
- School
of Tropical Agriculture and Forestry, Hainan
University, Haikou 570228, China
| | - Zibo Yin
- School
of Tropical Agriculture and Forestry, Hainan
University, Haikou 570228, China
| | - Jusong Liu
- School
of Tropical Agriculture and Forestry, Hainan
University, Haikou 570228, China
| | - Cheng Tang
- School
of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Yunfei Zhang
- School
of Tropical Agriculture and Forestry, Hainan
University, Haikou 570228, China
| | - Lanying Wang
- School
of Tropical Agriculture and Forestry, Hainan
University, Haikou 570228, China
| | - Hanzeng Li
- School
of Environmental Science and Engineering, Hainan University, Haikou 570228, China
| | - Yanping Luo
- School
of Tropical Agriculture and Forestry, Hainan
University, Haikou 570228, China
| |
Collapse
|
3
|
Wang C, Hu H, Liu J, Rong X, Zhang J, Du Y. Study on the mechanism of brain injury caused by acute diquat poisoning based on metabolomics. Toxicol Appl Pharmacol 2025; 494:117161. [PMID: 39571689 DOI: 10.1016/j.taap.2024.117161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/15/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
Brain injury following acute diquat poisoning has become increasingly common in moderate to severe cases, with unclear pathogenesis and high mortality. To investigate this, we conducted metabolomics on brain tissue from poisoned rats, combined with clinical biochemical and pathological analyses. In the high-dose group, 24 metabolites showed significant differences compared to the control group: 18 were upregulated, including cytosine, sedoheptulose-7-phosphate, indole, 3-dehydroshikimate, etc.; 6 were downregulated, including 6-phosphogluconic acid, 3-hydroxybenzoic acid, dAMP, etc. In the low-dose group, 10 metabolites showed significant differences: 4 were upregulated, including pentamidine, γ-tocotrienol, benzoylecgonine, etc.; and 6 were downregulated, including dAMP, glutathione, 3-hydroxybenzoic acid, etc. Enrichment analysis identified two key pathways-phenylalanine, tyrosine, and tryptophan biosynthesis, and the pentose phosphate pathway-as involved in brain injury. ROC analysis of six differential metabolites showed that sedoheptulose-7-phosphate, (2R)-2-hydroxy-3-(phosphonatooxy)propanoate, and 3-hydroxybenzoic acid had AUC values above 0.8. These findings suggest that these three metabolites demonstrate strong diagnostic potential for brain injury induced by diquat poisoning. Correlation analysis linked these biomarkers to clinical indicators such as neutrophil count and the eutrophil to lymphocyte ratio, supporting their relevance. This study provides insights into the mechanisms and biomarkers of diquat-induced brain injury, offering a foundation for future treatment and rapid detection.
Collapse
Affiliation(s)
- Chaocheng Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Hu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junzhao Liu
- Department of Emergency and Critical Care Medicine, West China School of Public Health, West China Fourth Hospital, Si Chuan, China
| | - Xia Rong
- Department of Emergency and Critical Care Medicine, West China School of Public Health, West China Fourth Hospital, Si Chuan, China
| | - Jing Zhang
- Department of Clinical Laboratory, West China School of Public Health, West China Fourth Hospital, Si Chuan, China
| | - Yu Du
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Emergency and Critical Care Medicine, West China School of Public Health, West China Fourth Hospital, Si Chuan, China; West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu 610041, Si Chuan, China.
| |
Collapse
|
4
|
Cao X, Sui B, Wu B, Geng Z, Song B. MR study on white matter injury in patients with acute diquat poisoning. Neurotoxicology 2025; 106:37-45. [PMID: 39643089 DOI: 10.1016/j.neuro.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE To explore the microstructural damage of white matter in acute diquat (DQ) poisoning patients using diffusion kurtosis imaging (DKI) and Tract-based Spatial Statistics (TBSS). METHODS This study included 19 DQ poisoning patients and 19 age-matched controls. MRI was performed using a 3.0 T Philips Achieva scanner with sequences including 3D T1WI, T2WI, DWI, 3D T2WI-FLAIR, and DKI (3 b-values, 15 directions). DICOM to NIFTI image form conversion was done using MRIcron's Dcm2niigui, followed by motion and eddy current correction with FSL to create a brain mask. Scalar indicators (MK, AK, RK, FAK) were calculated with DKE software. TBSS was used for spatial normalization, skeletonization, and projection of DKI indices for group analysis with TFCE for multiple comparison correction (P < 0.025). RESULTS After the screening and enrollment process, 19 DQ-poisoned patients and 19 healthy volunteers were analyzed. No significant age or sex differences were found between groups. For Mean Kurtosis (MK), the right corticospinal tract showed a significant difference with a mean difference of 0.21 (95 % CI: 0.15-0.27) and P = 0.000503. Axial Kurtosis (AK) in the left superior longitudinal fasciculus had a mean difference of 0.18 (95 % CI: 0.12-0.24) and P = 0.0024. Fractional Anisotropy of Kurtosis (FAK) in the right corticospinal tract showed a mean difference of 0.19 (95 % CI: 0.13-0.25) and P = 0.0000318. Axial Kurtosis (AK) positively correlated with blood drug levels (r = 0.52, P < 0.05). Seven patients developed subcortical leukodystrophy, mainly in the frontal parietal lobe, with possible insular lobe involvement. CONCLUSIONS DQ poisoning primarily damages the right corticospinal tract, right cingulate gyrus, and left superior longitudinal fasciculus, potentially causing movement and visual impairments. The injury involves demyelination and axonal degeneration, with asymmetrical damage between hemispheres. The left superior longitudinal fasciculus injury is dose-dependent, and unlike prior studies, dopaminergic nuclei were unaffected. The frontal parietal lobe is predominantly affected, with some insular lobe involvement in DQ poisoning patients.
Collapse
Affiliation(s)
- Xueshan Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China
| | - Bo Sui
- Department of Radiology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Bailin Wu
- Department of Radiology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Zuojun Geng
- Department of Radiology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China.
| | - Bo Song
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China.
| |
Collapse
|
5
|
Cheng J, Yang L, Zhang Z, Xu D, Hua R, Chen H, Li X, Duan J, Li Q. Diquat causes mouse testis injury through inducing heme oxygenase-1-mediated ferroptosis in spermatogonia. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116562. [PMID: 38850704 DOI: 10.1016/j.ecoenv.2024.116562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Diquat dibromide (DQ) is a globally used herbicide in agriculture, and its overuse poses an important public health issue, including male reproductive toxicity in mammals. However, the effects and molecular mechanisms of DQ on testes are limited. In vivo experiments, mice were intraperitoneally injected with 8 or 10 mg/kg/ day of DQ for 28 days. It has been found that heme oxygenase-1 (HO-1) mediates DQ-induced ferroptosis in mouse spermatogonia, thereby damaging testicular development and spermatogenesis. Histopathologically, we found that DQ exposure caused seminiferous tubule disorders, reduced germ cells, and increased sperm malformation, in mice. Reactive oxygen species (ROS) staining of frozen section and transmission electron microscopy (TEM) displayed DQ promoted ROS generation and mitochondrial morphology alterations in mouse testes, suggesting that DQ treatment induced testicular oxidative stress. Subsequent RNA-sequencing further showed that DQ treatment might trigger ferroptosis pathway, attributed to disturbed glutathione metabolism and iron homeostasis in spermatogonia cells in vitro. Consistently, results of western blotting, measurements of MDA and ferrous iron, and ROS staining confirmed that DQ increased oxidative stress and lipid peroxidation, and accelerated ferrous iron accumulation both in vitro and in vivo. Moreover, inhibition of ferroptosis by deferoxamine (DFO) markedly ameliorated DQ-induced cell death and dysfunction. By RNA-sequencing, we found that the expression of HO-1 was significantly upregulated in DQ-treated spermatogonia, while ZnPP (a specific inhibitor of HO-1) blocked spermatogonia ferroptosis by balancing intracellular iron homeostasis. In mice, administration of the ferroptosis inhibitor ferrostatin-1 effectively restored the increase of HO-1 levels in the spermatogonia, prevented spermatogonia death, and alleviated the spermatogenesis disorders induced by DQ. Overall, these findings suggest that HO-1 mediates DQ-induced spermatogonia ferroptosis in mouse testes, and targeting HO-1 may be an effective protective strategy against male reproductive disorders induced by pesticides in agriculture.
Collapse
Affiliation(s)
- Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Li Yang
- Health Management Center, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Dejun Xu
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Rongmao Hua
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518000, China
| | - Huali Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621000, China
| | - Xiaoya Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Jiaxin Duan
- College of Animal Science, Shanxi Agricultural University, Taiyuan 030801, China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
6
|
Braun G, Escher BI. Prioritization of mixtures of neurotoxic chemicals for biomonitoring using high-throughput toxicokinetics and mixture toxicity modeling. ENVIRONMENT INTERNATIONAL 2023; 171:107680. [PMID: 36502700 DOI: 10.1016/j.envint.2022.107680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Modern society continues to pollute the environment with larger quantities of chemicals that have also become more structurally and functionally diverse. Risk assessment of chemicals can hardly keep up with the sheer numbers that lead to complex mixtures of increasing chemical diversity including new chemicals, substitution products on top of still abundant legacy compounds. Fortunately, over the last years computational tools have helped us to identify and prioritize chemicals of concern. These include toxicokinetic models to predict exposure to chemicals as well as new approach methodologies such as in-vitro bioassays to address toxicodynamic effects. Combined, they allow for a prediction of mixtures and their respective effects and help overcome the lack of data we face for many chemicals. In this study we propose a high-throughput approach using experimental and predicted exposure, toxicokinetic and toxicodynamic data to simulate mixtures, to which a virtual population is exposed to and predict their mixture effects. The general workflow is adaptable for any type of toxicity, but we demonstrated its applicability with a case study on neurotoxicity. If no experimental data for neurotoxicity were available, we used baseline toxicity predictions as a surrogate. Baseline toxicity is the minimal toxicity any chemical has and might underestimate the true contribution to the mixture effect but many neurotoxicants are not by orders of magnitude more potent than baseline toxicity. Therefore, including baseline-toxic effects in mixture simulations yields a more realistic picture than excluding them in mixture simulations. This workflow did not only correctly identify and prioritize known chemicals of concern like benzothiazoles, organochlorine pesticides and plasticizers but we were also able to identify new potential neurotoxicants that we recommend to include in future biomonitoring studies and if found in humans, to also include in neurotoxicity screening.
Collapse
Affiliation(s)
- Georg Braun
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany.
| | - Beate I Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Zhou JN, Lu YQ. Lethal diquat poisoning manifests as acute central nervous system injury and circulatory failure: A retrospective cohort study of 50 cases. EClinicalMedicine 2022; 52:101609. [PMID: 35990582 PMCID: PMC9386369 DOI: 10.1016/j.eclinm.2022.101609] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/09/2022] [Accepted: 07/21/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The mortality rate of patients with diquat (DQ) poisoning is extremely high due to insufficient understanding of DQ-induced injury. This study aimed to summarize the characteristics of DQ poisoning as well as analyse the correlation between plasma DQ concentration and patient outcomes, thus providing a new strategy for diagnosis and treatment. METHODS This single-centre retrospective cohort study was conducted at the Emergency Department of the First Affiliated Hospital, Zhejiang University School of Medicine, China, between Oct 9, 2019 and March 10, 2022. 50 patients, whose plasma or urine samples tested positive for diquat and negative for paraquat by high performance liquid chromatography-tandem mass spectrometry, were included in the study. FINDINGS The mortality rate of acute DQ poisoning was 25 (50%) of 50. Compared with the survival group, the death group presented significantly higher initial plasma DQ concentration (Cp1), aspartate aminotransferase, alanine aminotransferase, serum creatinine, and creatine kinase-MB (P < 0.05). We found that six (24.0%) patients died of central nervous system injury, six (24.0%) patients died of refractory circulatory failure, and 13 (52.0%) patients died of central nervous system injury combined with circulatory failure. Receiver operator characteristic curve analysis showed that the area under the curve of Cp1 was 0.967 (95% CI: 0.911, 1.000), and the cut-off value was 3516.885 ng/mL (sensitivity, 90.9%; specificity, 96.0%). INTERPRETATION Lethal DQ poisoning is primarily associated with serious brain and vascular injury, as well as a high rate of mortality. Further research into the mechanisms of refractory circulatory failure and central nerve system damage could help reduce mortality. FUNDING There are no funding sources to declare.
Collapse
Affiliation(s)
- Jia-Ning Zhou
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, People's Republic of China
- Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou 310003, Zhejiang, People's Republic of China
| | - Yuan-Qiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, People's Republic of China
- Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou 310003, Zhejiang, People's Republic of China
- Correspondence author at: 79 Qingchun Road, Hangzhou 310003, People's Republic of China.
| |
Collapse
|
8
|
Xiao Y, Lin X, Zhou M, Ren T, Gao R, Liu Z, Shen W, Wang R, Xie X, Song Y, Hu W. Metabolomics analysis of the potential toxicological mechanisms of diquat dibromide herbicide in adult zebrafish (Danio rerio) liver. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1039-1055. [PMID: 35831485 DOI: 10.1007/s10695-022-01101-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/30/2022] [Indexed: 06/15/2023]
Abstract
Although diquat is a widely used water-soluble herbicide in the world, its sublethal adverse effects to fish have not been well characterised. In this study, histopathological examination and biochemical assays were applied to assess hepatotoxicity and combined with gas chromatography-mass spectrometry (GC-MS)-based metabolomics analysis to reveal overall metabolic mechanisms in the liver of zebrafish (Danio rerio) after diquat exposure at concentrations of 0.34 and 1.69 mg·L-1 for 21 days. Results indicated that 1.69 mg·L-1 diquat exposure caused cellular vacuolisation and degeneration with nuclear abnormality and led to the disturbance of antioxidative system and dysfunction in the liver. No evident pathological injury was detected, and changes in liver biochemistry were not obvious in the fish exposed to 0.34 mg·L-1 diquat. Multivariate statistical analysis revealed differences between profiles obtained by GC-MS spectrometry from control and two treatment groups. A total of 17 and 22 metabolites belonging to different classes were identified following exposure to 0.34 and 1.69 mg·L-1 diquat, respectively. The metabolic changes in the liver of zebrafish are mainly manifested as inhibition of energy metabolism, disorders of amino acid metabolism and reduction of antioxidant capacity caused by 1.69 mg·L-1 diquat exposure. The energy metabolism of zebrafish exposed to 0.34 mg·L-1 diquat was more inclined to rely on anaerobic glycolysis than that of normal zebrafish, and interference effects on lipid metabolism were observed. The metabolomics approach provided an innovative perspective to explore possible hepatic damages on fish induced by diquat as a basis for further research.
Collapse
Affiliation(s)
- Ye Xiao
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Xiang Lin
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Meilan Zhou
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Tianyu Ren
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Ruili Gao
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Zhongqun Liu
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Wenjing Shen
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Rong Wang
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Xi Xie
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Yanting Song
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China
| | - Wenting Hu
- School of Pharmaceutical Sciences, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China.
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, 58 Renmin Rd, Haikou, 570228, People's Republic of China.
| |
Collapse
|
9
|
Chen J, Su Y, Lin R, Lin F, Shang P, Hussain R, Shi D. Effects of Acute Diquat Poisoning on Liver Mitochondrial Apoptosis and Autophagy in Ducks. Front Vet Sci 2021; 8:727766. [PMID: 34458360 PMCID: PMC8385319 DOI: 10.3389/fvets.2021.727766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022] Open
Abstract
Diquat (DQ) is an effective herbicide and is widely used in agriculture. Due to persistent and frequent applications, it can enter into aquatic ecosystem and induce toxic effects to exposed aquatic animals. The residues of DQ via food chain accumulate in different tissues of exposed animals including humans and cause adverse toxic effects. Therefore, it is crucial and important to understand the mechanisms of toxic effects of DQ in exposed animals. We used ducks as test specimens to know the effects of acute DQ poisoning on mechanisms of apoptosis and autophagy in liver tissues. Results on comparison of various indexes of visceral organs including histopathological changes, apoptosis, autophagy-related genes, and protein expression indicated the adverse effects of DQ on the liver. The results of our experimental trial showed that DQ induces non-significant toxic effects on pro-apoptotic factors like BAX, BAK1, TNF-α, caspase series, and p53. The results revealed that anti-apoptotic gene Parkin was significantly upregulated, while an upward trend was also observed for Bcl2, suggesting that involvement of the anti-apoptotic factors in ducklings plays an important role in DQ poisoning. Results showed that DQ significantly increased the protein expression level of the autophagy factor Beclin 1 in the liver. Results on key autophagy factors like LC3A, LC3B, and p62 showed an upward trend at gene level, while the protein expression level of both LC3B and p62 reduced that might be associated with process of translation affected by the pro-apoptotic components such as apoptotic protease that inhibits the occurrence of autophagy while initiating cell apoptosis. The above results indicate that DQ can induce cell autophagy and apoptosis and the exposed organism may resist the toxic effects of DQ by increasing anti-apoptotic factors.
Collapse
Affiliation(s)
- Jiaxin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yalin Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Renzhao Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Fei Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry College, Linzhi, China
| | - Riaz Hussain
- Department of Pathology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Dayou Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
10
|
Paraoxonase Role in Human Neurodegenerative Diseases. Antioxidants (Basel) 2020; 10:antiox10010011. [PMID: 33374313 PMCID: PMC7824310 DOI: 10.3390/antiox10010011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
The human body has biological redox systems capable of preventing or mitigating the damage caused by increased oxidative stress throughout life. One of them are the paraoxonase (PON) enzymes. The PONs genetic cluster is made up of three members (PON1, PON2, PON3) that share a structural homology, located adjacent to chromosome seven. The most studied enzyme is PON1, which is associated with high density lipoprotein (HDL), having paraoxonase, arylesterase and lactonase activities. Due to these characteristics, the enzyme PON1 has been associated with the development of neurodegenerative diseases. Here we update the knowledge about the association of PON enzymes and their polymorphisms and the development of multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Parkinson's disease (PD).
Collapse
|
11
|
Lovejoy PC, Fiumera AC. Effects of Dual Exposure to the Herbicides Atrazine and Paraquat on Adult Climbing Ability and Longevity in Drosophila melanogaster. INSECTS 2019; 10:E398. [PMID: 31717666 PMCID: PMC6920984 DOI: 10.3390/insects10110398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/17/2019] [Accepted: 11/05/2019] [Indexed: 06/10/2023]
Abstract
Anthropomorphic effects are changing the planet, and therefore, organisms are being exposed to many new biotic and abiotic stressors. Exposure to multiple stressors can affect organisms in ways that are different than the sum of their individual effects, and these interactions are often difficult to predict. Atrazine and paraquat are two of the most widely used herbicides in the United States, and are individually known to increase oxidative damage, affect dopaminergic functioning, reduce longevity, and alter motor ability in non-target organisms. We measured the effects of individual and combined exposure to low doses of atrazine and paraquat on climbing ability and longevity of Drosophila melanogaster. Atrazine and paraquat interact to affect D. melanogaster climbing ability and longevity in different ways. Atrazine appeared to have a weak mitigative effect against the decrease in climbing ability caused by paraquat. In contrast, combined exposure to atrazine and paraquat had detrimental synergistic effects on female longevity. Overall, this study shows that atrazine and paraquat can interact and that it is important to measure several traits when assessing the consequences of exposure to multiple stressors. Future studies should continue to assess the impacts of stressor interactions on organisms, as many combinations have never been examined.
Collapse
Affiliation(s)
| | - Anthony C. Fiumera
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA;
| |
Collapse
|
12
|
Magalhães N, Carvalho F, Dinis-Oliveira RJ. Human and experimental toxicology of diquat poisoning: Toxicokinetics, mechanisms of toxicity, clinical features, and treatment. Hum Exp Toxicol 2018; 37:1131-1160. [PMID: 29569487 DOI: 10.1177/0960327118765330] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Diquat (1,1'-ethylene-2,2'-bipyridinium ion; DQ) is a nonselective quick-acting herbicide, which is used as contact and preharvest desiccant to control terrestrial and aquatic vegetation. Several cases of human poisoning were reported worldwide mainly due to intentional ingestion of the liquid formulations. Its toxic potential results from its ability to produce reactive oxygen and nitrogen species through redox cycling processes that can lead to oxidative stress and potentially cell death. Kidney is the main target organ due to DQ toxicokinetics and redox cycling. There is no antidote against DQ intoxications, and the efficacy of treatments currently applied is still unsatisfactory. The aim of this work was to review the most relevant human and experimental findings related to DQ, characterizing its chemistry, activity as herbicide, mechanisms of toxicity, consequences of poisoning, and potential therapeutic approaches taking into account previous experience in developing antidotes for paraquat, a more toxic bipyridinium herbicide.
Collapse
Affiliation(s)
- N Magalhães
- 1 UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - F Carvalho
- 1 UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - R J Dinis-Oliveira
- 1 UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.,2 IINFACTS-Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS-CESPU), Gandra, Portugal.,3 Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
13
|
Wang XH, Souders CL, Zhao YH, Martyniuk CJ. Mitochondrial bioenergetics and locomotor activity are altered in zebrafish (Danio rerio) after exposure to the bipyridylium herbicide diquat. Toxicol Lett 2018; 283:13-20. [DOI: 10.1016/j.toxlet.2017.10.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/06/2017] [Accepted: 10/29/2017] [Indexed: 12/19/2022]
|
14
|
Patel M, McElroy PB. Mitochondrial Dysfunction in Parkinson’s Disease. OXIDATIVE STRESS AND REDOX SIGNALLING IN PARKINSON’S DISEASE 2017. [DOI: 10.1039/9781782622888-00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders where oxidative stress and mitochondrial dysfunction have been implicated as etiological factors. Mitochondria are the major producers of reactive oxygen species (ROS) that can have damaging effects to cellular macromolecules leading to neurodegeneration. The most compelling evidence for the role of mitochondria in the pathogenesis of PD has been derived from toxicant-induced models of parkinsonism. Over the years, epidemiological studies have suggested a link between exposure to environmental toxins such as pesticides and the risk of developing PD. Data from human and experimental studies involving the use of chemical agents like paraquat, diquat, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rotenone and maneb have provided valuable insight into the underlying mitochondrial mechanisms contributing to PD and associated neurodegeneration. In this review, we have discussed the role of mitochondrial ROS and dysfunction in the pathogenesis of PD with a special focus on environmental agent-induced parkinsonism. We have described the various mitochondrial mechanisms by which such chemicals exert neurotoxicity, highlighting some landmark epidemiological and experimental studies that support the role of mitochondrial ROS and oxidative stress in contributing to these effects. Finally, we have discussed the significance of these studies in understanding the mechanistic underpinnings of PD-related dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| | - Pallavi Bhuyan McElroy
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| |
Collapse
|
15
|
Karuppagounder SS, Xiong Y, Lee Y, Lawless MC, Kim D, Nordquist E, Martin I, Ge P, Brahmachari S, Jhaldiyal A, Kumar M, Andrabi SA, Dawson TM, Dawson VL. LRRK2 G2019S transgenic mice display increased susceptibility to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated neurotoxicity. J Chem Neuroanat 2016; 76:90-97. [PMID: 26808467 DOI: 10.1016/j.jchemneu.2016.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/20/2016] [Accepted: 01/20/2016] [Indexed: 12/21/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common causes of late onset autosomal dominant form of Parkinson disease (PD). Gain of kinase activity due to the substitution of Gly 2019 to Ser (G2019S) is the most common mutation in the kinase domain of LRRK2. Genetic predisposition and environmental toxins contribute to the susceptibility of neurodegeneration in PD. To identify whether the genetic mutations in LRRK2 increase the susceptibility to environmental toxins in PD models, we exposed transgenic mice expressing human G2019S mutant or wild type (WT) LRRK2 to the environmental toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP treatment resulted in a greater loss of tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta (SNpc) in LRRK2 G2019S transgenic mice compared to the LRRK2 WT overexpressing mice. Similarly loss of dopamine levels were greater in the striatum of LRRK2 G2019S mice when compared to the LRRK2 WT mice when both were treated with MPTP. This study suggests a likely interaction between genetic and environmental risk factors in the PD pathogenesis and that the G2019S mutation in LRRK2 increases the susceptibility of dopamine neurons to PD-causing toxins.
Collapse
Affiliation(s)
- Senthilkumar S Karuppagounder
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Yulan Xiong
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Yunjong Lee
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Maeve C Lawless
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biological Science, Louisiana State University, Baton Rouge, LA 70803, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Donghyun Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily Nordquist
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Krieger School of Arts and Sciences, Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Ian Martin
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Preston Ge
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Krieger School of Arts and Sciences, Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Saurav Brahmachari
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Aanishaa Jhaldiyal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Manoj Kumar
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Shaida A Andrabi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA USA.
| |
Collapse
|
16
|
Dietary administration of diquat for 13 weeks does not result in a loss of dopaminergic neurons in the substantia nigra of C57BL/6J mice. Regul Toxicol Pharmacol 2015; 75:81-8. [PMID: 26683030 DOI: 10.1016/j.yrtph.2015.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 11/24/2022]
Abstract
Male and female C57BL/6J mice were administered diquat dibromide (DQ∙Br2) in their diets at concentrations of 0 (control), 12.5 and 62.5 ppm for 13 weeks to assess the potential effects of DQ on the nigrostriatal dopaminergic system. Achieved dose levels at 62.5 ppm were 6.4 and 7.6 mg DQ (ion)/kg bw/day for males and females, respectively. A separate group of mice was administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) ip as a positive control. The comparative effects of DQ and MPTP on the substantia nigra pars compacta (SNpc) and/or striatum were assessed using neurochemical, neuropathological and stereological endpoints. Morphological and stereological assessments were performed by investigators who were "blinded" to dose group. DQ had no effect on striatal dopamine concentration or dopamine turnover. There was no evidence of neuronal degeneration, astrocytic or microglial activation, or a reduction in the number of tyrosine hydroxylase positive (TH(+)) neurons in the SNpc or neuronal processes in the striatum of DQ-treated mice. These results are consistent with the rapid clearance of DQ from the brain following a single dose of radiolabeled DQ. In contrast, MPTP-treated mice exhibited decreased striatal dopamine concentration, reduced numbers of TH(+) neurons in the SNpc, and neuropathological changes, including neuronal necrosis, as well as astrocytic and microglial activation in the striatum and SNpc.
Collapse
|
17
|
Lopert P, Patel M. Mitochondrial mechanisms of redox cycling agents implicated in Parkinson's disease. J Neural Transm (Vienna) 2015; 123:113-23. [PMID: 25749885 DOI: 10.1007/s00702-015-1386-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/20/2015] [Indexed: 12/21/2022]
Abstract
Environmental agents have been implicated in Parkinson's disease (PD) based on epidemiological studies and the ability of toxicants to replicate features of PD. However, the precise mechanisms by which toxicants induce dopaminergic toxicity observed in the idiopathic form of PD remain to be fully understood. The roles of ROS and mitochondria are strongly suggested in the mechanisms by which these toxicants exert dopaminergic toxicity. There are marked differences and similarities shared by the toxicants in increasing steady-state levels of mitochondrial ROS. Furthermore, toxicants increase steady-state mitochondrial ROS levels by stimulating the production, inhibiting the antioxidant pathways of both. This review will focus on the role of mitochondria and ROS in PD associated with environmental exposures to redox-based toxicants.
Collapse
Affiliation(s)
- Pamela Lopert
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Manisha Patel
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
18
|
|
19
|
Solid-Phase Microextraction Combined with GC–MS for Determination of Diquat and Paraquat Residues in Water. Chromatographia 2014. [DOI: 10.1007/s10337-014-2809-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
20
|
Karuppagounder SS, Bhattacharya D, Ahuja M, Suppiramaniam V, DeRuiter J, Clark R, Dhanasekaran M. Elucidating the neurotoxic effects of MDMA and its analogs. Life Sci 2014; 101:37-42. [DOI: 10.1016/j.lfs.2014.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 01/21/2014] [Accepted: 02/08/2014] [Indexed: 10/25/2022]
|
21
|
Bouétard A, Besnard AL, Vassaux D, Lagadic L, Coutellec MA. Impact of the redox-cycling herbicide diquat on transcript expression and antioxidant enzymatic activities of the freshwater snail Lymnaea stagnalis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2013; 126:256-265. [PMID: 23237706 DOI: 10.1016/j.aquatox.2012.11.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/06/2012] [Accepted: 11/17/2012] [Indexed: 06/01/2023]
Abstract
The presence of pesticides in the environment results in potential unwanted effects on non-target species. Freshwater organisms inhabiting water bodies adjacent to agricultural areas, such as ditches, ponds and marshes, are good models to test such effects as various pesticides may reach these habitats through several ways, including aerial drift, run-off, and drainage. Diquat is a non-selective herbicide used for crop protection or for weed control in such water bodies. In this study, we investigated the effects of diquat on a widely spread aquatic invertebrate, the holarctic freshwater snail Lymnaea stagnalis. Due to the known redox-cycling properties of diquat, we studied transcript expression and enzymatic activities relative to oxidative and general stress in the haemolymph and gonado-digestive complex (GDC). As diquat is not persistent, snails were exposed for short times (5, 24, and 48 h) to ecologically relevant concentrations (22.2, 44.4, and 222.2 μg l(-1)) of diquat dibromide. RT-qPCR was used to quantify the transcription of genes encoding catalase (cat), a cytosolic superoxide dismutase (Cu/Zn-sod), a selenium-dependent glutathione peroxidase (gpx), a glutathione reductase (gred), the retinoid X receptor (rxr), two heat shock proteins (hsp40 and hsp70), cortactin (cor) and the two ribosomal genes r18S and r28s. Enzymatic activities of SOD, Gpx, Gred and glutathione S-transferase (GST) were investigated in the GDC using spectrophoto/fluorometric methods. Opposite trends were obtained in the haemolymph depending on the herbicide concentration. At the lowest concentration, effects were mainly observed after 24 h of exposure, with over-transcription of cor, hsp40, rxr, and sod, whereas higher concentrations down-regulated the expression of most of the studied transcripts, especially after 48 h of exposure. In the GDC, earlier responses were observed and the fold-change magnitude was generally much higher: transcription of all target genes increased significantly (or non-significantly for cat) after 5 h of exposure, and went back to control levels afterwards, suggesting the onset of an early response to oxidative stress associated to the unbalance of reactive oxygen species (ROS) in hepatocytes. Although increases obtained for Gred and SOD activities were globally consistent with their respective transcript expressions, up-regulation of transcription was not always correlated with increase of enzymatic activity, indicating that diquat might affect steps downstream of transcription. However, constitutive levels of enzymatic activities were at least maintained. In conclusion, diquat was shown to affect expression of the whole set of studied transcripts, reflecting their suitability as markers of early response to oxidative stress in L. stagnalis.
Collapse
Affiliation(s)
- Anthony Bouétard
- INRA, UMR INRA-Agrocampus Ouest ESE, Equipe Ecotoxicologie et Qualité des Milieux Aquatiques, Rennes, France.
| | | | | | | | | |
Collapse
|