1
|
McAtee D, Abdelmoneim A. A zebrafish-based acoustic motor response (AMR) assay to evaluate chemical-induced developmental neurotoxicity. Neurotoxicology 2024; 103:60-70. [PMID: 38851595 DOI: 10.1016/j.neuro.2024.06.003] [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: 08/21/2023] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Behavioral assays using early-developing zebrafish (Danio rerio) offer a valuable supplement to the in vitro battery adopted as new approach methodologies (NAMs) for assessing risk of chemical-induced developmental neurotoxicity. However, the behavioral assays primarily adopted rely on visual stimulation to elicit behavioral responses, known as visual motor response (VMR) assays. Ocular deficits resulting from chemical exposures can, therefore, confound the behavioral responses, independent of effects on the nervous system. This highlights the need for complementary assays employing alternative forms of sensory stimulation. In this study, we investigated the efficacy of acoustic stimuli as triggers of behavioral responses in larval zebrafish, determined the most appropriate data acquisition mode, and evaluated the suitability of an acoustic motor response (AMR) assay as means to assess alterations in brain activity and risk of chemical-induced developmental neurotoxicity. We quantified the motor responses of 120 h post-fertilization (hpf) larvae to acoustic stimuli with varying patterns and frequencies, and determined the optimal time intervals for data acquisition. Following this, we examined changes in acoustic and visual motor responses resulting from exposures to pharmacological agents known to impact brain activity (pentylenetetrazole (PTZ) and tricaine-s (MS-222)). Additionally, we examined the AMR and VMR of larvae following exposure to two environmental contaminants associated with developmental neurotoxicity: arsenic (As) and cadmium (Cd). Our findings indicate that exposure to a 100 Hz sound frequency in 100 ms pulses elicits the strongest behavioral response among the acoustic stimuli tested and data acquisition in 2 s time intervals is suitable for response assessment. Exposure to PTZ exaggerated and depressed both AMR and VMR in a concentration-dependent manner, while exposure to MS-222 only depressed them. Similarly, exposure to As and Cd induced respective hyper- and hypo-activation of both motor responses. This study highlights the efficiency of the proposed zebrafish-based AMR assay in demonstrating risk of chemical-induced developmental neurotoxicity and its suitability as a complement to the widely adopted VMR assay.
Collapse
Affiliation(s)
- Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
2
|
Alymbaeva D, Szabo C, Jocsak G, Bartha T, Zsarnovszky A, Kovago C, Ondrasovicova S, Kiss DS. Analysis of arsenic-modulated expression of hypothalamic estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptor gamma mRNA and simultaneous mitochondrial morphology and respiration rates in the mouse. PLoS One 2024; 19:e0303528. [PMID: 38753618 PMCID: PMC11098319 DOI: 10.1371/journal.pone.0303528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/26/2024] [Indexed: 05/18/2024] Open
Abstract
Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,β; TRα,β) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,β, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRβ and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.
Collapse
Affiliation(s)
- Daiana Alymbaeva
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Csaba Szabo
- Department of Animal Physiology and Health, Hungarian University of Agricultural and Life Sciences, Godollo, Hungary
| | - Gergely Jocsak
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Tibor Bartha
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Attila Zsarnovszky
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
- Department of Animal Physiology and Health, Hungarian University of Agricultural and Life Sciences, Godollo, Hungary
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Animal Physiology and Health, Institute of Physiology and Nutrition, Hungarian University of Agricultural and Life Sciences, Kaposvar, Hungary
| | - Csaba Kovago
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| | - Silvia Ondrasovicova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - David Sandor Kiss
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
3
|
Ding B, Ma X, Liu Y, Ni B, Lu S, Chen Y, Liu X, Zhang W. Arsenic-Induced, Mitochondria-Mediated Apoptosis Is Associated with Decreased Peroxisome Proliferator-Activated Receptor γ Coactivator α in Rat Brains. TOXICS 2023; 11:576. [PMID: 37505542 PMCID: PMC10384476 DOI: 10.3390/toxics11070576] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023]
Abstract
Chronic exposure to arsenic in drinking water damages cognitive function, and nerve cell apoptosis is one of the primary characteristics. The damage to mitochondrial structure and/or function is one of the main characteristics of apoptosis. Peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) is involved in the regulation of mitochondrial biogenesis, energy metabolism, and apoptosis. In this study, we aimed to study the role of PGC-1α in sodium arsenite (NaAsO2)-induced mitochondrial apoptosis in rat hippocampal cells. We discovered that increased arsenic-induced apoptosis in rat hippocampus increased with NaAsO2 (0, 2, 10, and 50 mg/L, orally via drinking water for 12 weeks) exposure by TUNEL assay, and the structure of mitochondria was incomplete and swollen and had increased lysosomes, lipofuscins, and nuclear membrane shrinkage observed via transmission electron microscopy. Furthermore, NaAsO2 reduced the levels of Bcl-2 and PGC-1α and increased the levels of Bax and cytochrome C expression. Moreover, correlation analysis showed that brain arsenic content was negatively correlated with PGC-1α levels and brain ATP content; PGC-1α levels were negatively correlated with apoptosis rate; and brain ATP content was positively correlated with PGC-1α levels, but no significant correlation between ATP content and apoptosis has been observed in this study. Taken together, the results of this study indicate that NaAsO2-induced mitochondrial pathway apoptosis is related to the reduction of PGC-1α, accompanied by ATP depletion.
Collapse
Affiliation(s)
- Bo Ding
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Xinbo Ma
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yang Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Bangyao Ni
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Siqi Lu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Yuting Chen
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin 150081, China
| |
Collapse
|
4
|
Zhang C, Li Y, Yu H, Ye L, Li T, Zhang X, Wang C, Li P, Ji H, Gao Q, Dong S. Nanoplastics promote arsenic-induced ROS accumulation, mitochondrial damage and disturbances in neurotransmitter metabolism of zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 863:161005. [PMID: 36539083 DOI: 10.1016/j.scitotenv.2022.161005] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
As a carrier, nanoplastics (NPs) can adsorb other toxic substances and thus modify their biological toxicity. Numerous studies have investigated the neurotoxic of high concentrations of arsenic (As, 2.83 mg/L-5 mg/L). However, it is still unknown whether the relatively low environmentally relevant concentrations of As (200 μg/L) can damage the structure and function of fish brains with the presence of NPs. In this study, zebrafish were exposed to polystyrene NPs, As and their mixture for 30 days respectively. Firstly, we found that the presence of NPs promoted the accumulation of As in zebrafish brains. Thereby the co-exposure of NPs and As further promoted the production of reactive oxygen species (ROS) in zebrafish brains compared with the single exposure of NPs or As, resulting in severe oxidative stress. Moreover, accumulated ROS directly damaged the mitochondrial membrane and mtDNA in zebrafish brains. Moreover, the mitochondrial damage was further aggravated due to inhibited mitochondrial fusion and activated mitochondrial division and mitophagy. Ultimately, the co-exposure led to mitochondrial damage in the zebrafish brain. Damaged mitochondria may not meet the high energy metabolic requirement for neuronal function. As a result, the normal function of nerve cells was adversely affected and eventually cell apoptosis may occur. Besides, the co-exposure caused more significant structural alterations in zebrafish brain tissue. Finally, the co-exposure of NPs and As caused abnormal biosynthesis and degradation of dopamine and acetylcholine. These resulted in decreased dopamine levels and increased acetylcholine levels in zebrafish brains. In conclusion, the presence of NPs promoted the accumulation of As, thereby inducing severe oxidative stress, which caused structural alterations and mitochondrial damage in the zebrafish brain, thus disordering neuromodulation, which may ultimately cause neurological dysfunction in zebrafish. This study will provide a risk assessment for evaluating the biotoxicity of NPs and As to fish and even other animals.
Collapse
Affiliation(s)
- Cheng Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yanyao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Haibo Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Limin Ye
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Tian Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaotian Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chi Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Pengju Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Qinfeng Gao
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao 266100, China
| | - Shuanglin Dong
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao 266100, China
| |
Collapse
|
5
|
Hamdan DI, Tawfeek N, El-Shiekh RA, Khalil HMA, Mahmoud MY, Bakr AF, Zaafar D, Farrag N, Wink M, El-Shazly AM. Salix subserrata Bark Extract-Loaded Chitosan Nanoparticles Attenuate Neurotoxicity Induced by Sodium Arsenate in Rats in Relation with HPLC-PDA-ESI-MS/MS Profile. AAPS PharmSciTech 2022; 24:15. [PMID: 36522541 DOI: 10.1208/s12249-022-02478-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Pollution is a worldwide environmental risk. Arsenic (As) is an environmental pollutant with a major health concern due to its toxic effects on multiple body organs, including the brain. Humans are exposed to As through eating contaminated food and water or via skin contact. Salix species (willow) are plants with medicinal efficacy. Salix subserrata Willd bark extract-loaded chitosan nanoparticles (SBE.CNPs) was formulated, characterized, and evaluated against As-induced neurotoxicity. The stem bark was selected for nanoparticle formulation based on HPLC-PDA-ESI-MS/MS profiling and in vitro antioxidant assessment using free radical scavenging activity. SBE.CNPs demonstrated an average un-hydrated diameter of 193.4 ± 24.5 nm and zeta potential of + 39.6 ± 0.4 mV with an encapsulation efficiency of 83.7 ± 4.3%. Compared to As-intoxicated rats, SBE.CNP-treated rats exhibited anxiolytic activity and memory-boosting as evidenced in open field test, light-dark activity box, and Y-maze. Also, it increased the antioxidant biomarkers, including superoxide dismutase and glutathione peroxidase associated with reducing the malondialdehyde levels and apoptotic activity. Besides this, SBE.CNPs maintained the brain architecture and downregulated both nuclear factor-kappa B and heme oxygenase-1 expression. These results suggest that SBE.CNP administration showed promising potent neuroprotective and antioxidative efficiencies against arsenic-induced oxidative threats.
Collapse
Affiliation(s)
- Daila I Hamdan
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University, Shibin Elkom, 32511, Egypt.
| | - Nora Tawfeek
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El Aini st., Cairo, 11562, Egypt
| | - Heba M A Khalil
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Mohamed Y Mahmoud
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Alaa F Bakr
- Pathology Department, Faculty of Vet. Medicine, Cairo University, Giza, 12211, Egypt
| | - Dalia Zaafar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Information and Technology, El Mokattam, Egypt
| | - Nawaal Farrag
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | - Assem Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
- Faculty of Pharmacy, El Saleheya El Gadida University, 44813 El Saleheya El Gadida, El Saleheya, Egypt
| |
Collapse
|
6
|
Flora SJS, Jain K, Panghal A, Patwa J. Chemistry, Pharmacology, and Toxicology of Monoisoamyl Dimercaptosuccinic Acid: A Chelating Agent for Chronic Metal Poisoning. Chem Res Toxicol 2022; 35:1701-1719. [PMID: 35972774 DOI: 10.1021/acs.chemrestox.2c00129] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arsenic, a metalloid, is known to cause deleterious effects in various body organs, particularly the liver, urinary bladder, and brain, and these effects are primarily mediated through oxidative stress. Chelation therapy has been considered one of the promising medical treatments for arsenic poisoning. Meso 2,3- dimercaptosuccinic acid (DMSA) has been recognized as one of the most effective chelating drugs to treat arsenic poisoning. However, the drug is compromised with a number of shortcomings, including the inability to treat chronic arsenic poisoning due to its extracellular distribution. Monoisoamyl 2,3-dimercaptosuccinic acid, one of the analogues of meso 2,3-dimeraptosuccinic acid (DMSA), is a lipophilic chelator and has shown promise to be considered as a potential future chelating agent/antidote not only for arsenic but also for a few other heavy metals like lead, mercury, cadmium, and gallium arsenide. The results from numerous studies carried out in the recent past, mainly from our group, strongly support the clinical application of MiADMSA. This review paper summarizes most of the scientific details including the chemistry, pharmacology, and safety profile of MiADMSA. The efficacy of MiADMSA mainly against arsenic toxicity but also a few other heavy metals was also discussed. We also reviewed a few other strategies in order to achieve the optimum effects of MiADMSA, like combination therapy using two chelating agents or coadministration of a natural and synthetic antioxidant (including phytomedicine) along with MiADMSA for treatment of metal/metalloid poisoning. We also briefly discussed the use of nanotechnology (nano form of MiADMSA i.e. nano-MiADMSA) and compared it with bulk MiADMSA. All these strategies have been shown to be beneficial in getting more pronounced therapeutic efficacy of MiADMSA, as an adjuvant or as a complementary agent, by significantly increasing the chelating efficacy of MiADMSA.
Collapse
Affiliation(s)
- Swaran J S Flora
- National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Near CRPF Base, Lucknow, Uttar Pradesh 226002, India.,National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, Sahibzada Ajit Singh Nagar, Mohali, Punjab 160062, India
| | - Keerti Jain
- National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Near CRPF Base, Lucknow, Uttar Pradesh 226002, India
| | - Archna Panghal
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, Sahibzada Ajit Singh Nagar, Mohali, Punjab 160062, India
| | - Jayant Patwa
- National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Near CRPF Base, Lucknow, Uttar Pradesh 226002, India
| |
Collapse
|
7
|
Mozaffarian F, Dehghani MA, Vanani AR, Mahdavinia M. Protective Effects of Alpha Lipoic Acid Against Arsenic Induced Oxidative Stress in Isolated Rat Liver Mitochondria. Biol Trace Elem Res 2022; 200:1190-1200. [PMID: 33866498 DOI: 10.1007/s12011-021-02712-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 01/09/2023]
Abstract
Arsenic as a heavy metal and toxic pollutant has been established that has the hepatotoxic effect in animal and human models. Previous studies showed that mitochondria as the first target of arsenic toxicity has a pathogenic role in liver diseases. This study investigated alpha lipoic acid (ALA) as an antioxidant could ameliorate against liver toxicity induced by arsenic in rat mitochondria. First, mitochondria were isolated by the liver tissue centrifugation protocol. Then, isolated mitochondria were exposed with different concentrations of ALA and arsenic in different times for receiving the optimum dose and time. Finally, mitochondria were pretreated with the optimum concentrations and times of ALA and then treated with optimum concentration and time of arsenic (160 μg/ml; 30 min). The results demonstrated a significant decrease in total mitochondrial dehydrogenase activity (mitochondrial complex II) by 3, 4 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay after arsenic exposure. Mitochondria treated with arsenic also showed a significant increase in ROS generation, MMP, and MDA levels. The activity of mitochondrial catalase and mitochondrial GSH significantly decreased after exposure of mitochondria with arsenic. Pretreatment of mitochondria with ALA improved mitochondrial complex II activity; decreased mitochondrial membrane damage, MDA, and ROS amounts; and ameliorated mitochondrial GSH levels and mitochondrial catalase activity. These findings revealed that arsenic induced oxidative stress and mitochondria dysfunction, while ALA improved mitochondrial function through increasing of antioxidant defense or preserving of complex II, but suggested that ALA could prevent from mitochondria dysfunction.
Collapse
Affiliation(s)
- Farzan Mozaffarian
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Amin Dehghani
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Atefeh Raesi Vanani
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
8
|
Thakur M, Rachamalla M, Niyogi S, Datusalia AK, Flora SJS. Molecular Mechanism of Arsenic-Induced Neurotoxicity including Neuronal Dysfunctions. Int J Mol Sci 2021; 22:10077. [PMID: 34576240 PMCID: PMC8471829 DOI: 10.3390/ijms221810077] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Arsenic is a key environmental toxicant having significant impacts on human health. Millions of people in developing countries such as Bangladesh, Mexico, Taiwan, and India are affected by arsenic contamination through groundwater. Environmental contamination of arsenic leads to leads to various types of cancers, coronary and neurological ailments in human. There are several sources of arsenic exposure such as drinking water, diet, wood preservatives, smoking, air and cosmetics, while, drinking water is the most explored route. Inorganic arsenic exhibits higher levels of toxicity compared its organic forms. Exposure to inorganic arsenic is known to cause major neurological effects such as cytotoxicity, chromosomal aberration, damage to cellular DNA and genotoxicity. On the other hand, long-term exposure to arsenic may cause neurobehavioral effects in the juvenile stage, which may have detrimental effects in the later stages of life. Thus, it is important to understand the toxicology and underlying molecular mechanism of arsenic which will help to mitigate its detrimental effects. The present review focuses on the epidemiology, and the toxic mechanisms responsible for arsenic induced neurobehavioral diseases, including strategies for its management from water, community and household premises. The review also provides a critical analysis of epigenetic and transgenerational modifications, mitochondrial oxidative stress, molecular mechanisms of arsenic-induced oxidative stress, and neuronal dysfunction.
Collapse
Affiliation(s)
- Manisha Thakur
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
- Toxicology Centre, Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Swaran Jeet Singh Flora
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| |
Collapse
|
9
|
Abdollahzade N, Majidinia M, Babri S. Melatonin: a pleiotropic hormone as a novel potent therapeutic candidate in arsenic toxicity. Mol Biol Rep 2021; 48:6603-6618. [PMID: 34453671 DOI: 10.1007/s11033-021-06669-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Arsenic is a natural element which exists in the environment in inorganic and organic forms. In humans, the main reason for the toxicity of arsenic is its uptake via water sources. As polluted water and the problems associated with it can be found in many countries. Therefore, considering all these positive effects of melatonin, this review is aimed at melatonin supplementation therapy on arsenic toxicity which seems to be a suitable therapeutic agent to eliminate the adverse effects of arsenic. METHODS AND RESULTS It is seen in previous studies that chronic exposure to arsenic could cause serious dys functions of organs and induce different degrees of toxicities that is one of the first hazardous materials in the classification of substances by the United States Environmental Protection Agency so leads to costly cleanup operations burdening the economy. Arsenic harmfulness degree depends on the bioavailability, chemical form, valence state, detoxification, and metabolism of human body. The oxidative stress has a major role in arsenic-induced toxicity; on the other hand, it was discovered that melatonin is a powerful scavenger for free radical and it's an extensive-spectrum antioxidant. CONCLUSION Due to its highly lipophilic and small size properties, melatonin accesses all intracellular organs by easily passing via the cell membrane and prevents protein, DNA damage, and lipid peroxidation. In particular, melatonin, by protecting and reducing oxidative stress in mitochondria, can normalize homeostasis and mitochondrial function and ultimately prevent apoptosis and cell death.
Collapse
Affiliation(s)
- Naseh Abdollahzade
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Shirin Babri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Oxidative Stress as a Common Key Event in Developmental Neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6685204. [PMID: 34336113 PMCID: PMC8315852 DOI: 10.1155/2021/6685204] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/29/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022]
Abstract
The developing brain is extremely sensitive to many chemicals. Perinatal exposure to neurotoxicants has been implicated in several neurodevelopmental disorders, including autism spectrum disorder, attention-deficit hyperactive disorder, and schizophrenia. Studies of the molecular and cellular events related to developmental neurotoxicity have identified a number of “adverse outcome pathways,” many of which share oxidative stress as a key event. Oxidative stress occurs when the balance between the production of free oxygen radicals and the activity of the cellular antioxidant system is dysregulated. In this review, we describe some of the developmental neurotoxins that target the antioxidant system and the mechanisms by which they elicit stress, including oxidative phosphorylation in mitochondria and plasma membrane redox system in rodent models. We also discuss future directions for identifying adverse outcome pathways related to oxidative stress and developmental neurotoxicity, with the goal of improving our ability to quickly and accurately screen chemicals for their potential developmental neurotoxicity.
Collapse
|
11
|
Cheng H, Yang B, Ke T, Li S, Yang X, Aschner M, Chen P. Mechanisms of Metal-Induced Mitochondrial Dysfunction in Neurological Disorders. TOXICS 2021; 9:142. [PMID: 34204190 PMCID: PMC8235163 DOI: 10.3390/toxics9060142] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 01/31/2023]
Abstract
Metals are actively involved in multiple catalytic physiological activities. However, metal overload may result in neurotoxicity as it increases formation of reactive oxygen species (ROS) and elevates oxidative stress in the nervous system. Mitochondria are a key target of metal-induced toxicity, given their role in energy production. As the brain consumes a large amount of energy, mitochondrial dysfunction and the subsequent decrease in levels of ATP may significantly disrupt brain function, resulting in neuronal cell death and ensuing neurological disorders. Here, we address contemporary studies on metal-induced mitochondrial dysfunction and its impact on the nervous system.
Collapse
Affiliation(s)
- Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
| | - Bobo Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China;
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| |
Collapse
|
12
|
Patwa J, Flora SJS. MiADMSA abrogates chronic copper-induced hepatic and immunological changes in Sprague Dawley rats. Food Chem Toxicol 2020; 145:111692. [PMID: 32871191 DOI: 10.1016/j.fct.2020.111692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
Wilson disease (WD) is an autosomal-recessive disorder associated with the impaired copper metabolism, resulting in hepatic and neurologic manifestations. D-Pencillamine (DPA) is a first-line of treatment however, monoisoamyl 2, 3-dimercaptosuccinic acid (MiADMSA), is gaining recognition recently as a future chelating agent of choice. We evaluated the effects of MiADMSA against copper-induced (20 mg/kg, orally, once, daily for 16 weeks) hepatic and immunological changes in the male Sprague Dawley (SD) rats. Copper overload increased the levels of pro-oxidant and concurrently decreased the levels of antioxidant enzymes in the liver. Increased oxidative stress triggered the up-regulation of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) in the liver and down-regulated the anti-inflammatory cytokine IL-4. Altered liver function parameters as well as serum immunoglobulins' (IgG, IgA, IgE, and IgM) levels, were also noted. MiADMSA treatment restored most of copper altered biochemical and immunological changes. Further, the histopathological changes proved that MiADMSA treatment ameliorated copper induced hepatic injury. Infra red spectra of liver tissue indicated shift in the characteristic -OH peak during copper exposure while the shifting came to normal in MiADMSA administered rat liver. We conclude that MiADMSA could be a promising antidote for the chronic copper toxicity and possibly in the clinical management of WD.
Collapse
Affiliation(s)
- Jayant Patwa
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, Uttar Pradesh, 226002, India
| | - S J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, Uttar Pradesh, 226002, India; National Institute of Pharmaceutical Education and Research, Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
13
|
Naqvi S, Kumar P, Flora SJS. Comparative efficacy of Nano and Bulk Monoisoamyl DMSA against arsenic-induced neurotoxicity in rats. Biomed Pharmacother 2020; 132:110871. [PMID: 33069968 DOI: 10.1016/j.biopha.2020.110871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022] Open
Abstract
Chelation therapy is considered as a safe and effective strategy to combat metal poisoning. Arsenic is known to cause neurological dysfunctions such as impaired memory, encephalopathy, and peripheral neuropathy as it easily crosses the blood-brain barrier. Oxidative stress is one of the mechanisms suggested for arsenic-induced neurotoxicity. We prepared Solid Lipid nanoparticles loaded with Monoisoamyl 2, 3-dimercaptosuccinic acid (Nano-MiADMSA), and compared their efficacy with bulk MiADMSA for treating arsenic-induced neurological and other biochemical effects. Solid lipid nanoparticles entrapping MiADMSA were synthesized and particle characterization was carried out by transmission electron microscopy (TEM) and dynamic light scattering (DLS). An in vivo study was planned to investigate the therapeutic efficacy of MiADMSA-encapsulated solid lipid nanoparticles (Nano-MiADMSA; 50 mg/kg orally for 5 days) and compared it with bulk MiADMSA against sodium meta-arsenite exposed rats (25 ppm in drinking water, for 12 weeks) in male rats. The results suggested the size of Nano-MiADMSA was between 100-120 nm ranges. We noted enhanced chelating properties of Nano-MiADMSA compared with bulk MiADMSA as evident by the reversal of oxidative stress variables like blood δ-aminolevulinic acid dehydratase (δ-ALAD), Reactive Oxygen Species (ROS), Catalase activity, Superoxide Dismutase (SOD), Thiobarbituric Acid Reactive Substances (TBARS), Reduced Glutathione (GSH) and Oxidized Glutathione (GSSG), Glutathione Peroxidase (GPx), Glutathione-S-transferase (GST) and efficient removal of arsenic from the blood and tissues. Recoveries in neurobehavioral parameters further confirmed nano-MiADMSA to be more effective than bulk MiADMSA. We conclude that treatment with Nano-MiADMSA is a better therapeutic strategy than bulk MiADMSA in reducing the effects of arsenic-induced oxidative stress and associated neurobehavioral changes.
Collapse
Affiliation(s)
- Saba Naqvi
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India
| | - Prince Kumar
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India.
| |
Collapse
|
14
|
Shi L, Hu X, Wang N, Liang H, Wu C, Cao H. Histopathological examination and transcriptome analyses to assess the acute toxic effects of arsenite exposure on rare minnows (Gobiocypris rarus). ECOTOXICOLOGY (LONDON, ENGLAND) 2020; 29:613-624. [PMID: 32385600 DOI: 10.1007/s10646-020-02222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/25/2020] [Indexed: 06/11/2023]
Abstract
Arsenic is ubiquitously present in the aquatic environment. We investigated the acute toxic effects of arsenite [As(III)] exposure on rare minnows (Gobiocypris rarus) in vivo. The 96-h LC50 value for exposure to As(III) was 13.73 mg/L. As(III) bioaccumulation in different tissues was measured using inductively-coupled plasma mass spectrometry, and the extent of As(III) accumulation was, from greatest to least, liver > intestine > gills > muscle > kidney > testis > brain. Histological examination revealed that in As(III)-treated fish, numerous cellular and tissue alterations were present in the gill, liver, and intestine tissues. Moreover, transmission electron microscopy showed ultrastructural alterations in hepatocytes. We also performed transcriptome analyses to investigate As(III)-induced toxicity response in the liver of As(III)-treated fish; various oxidative-related genes were differentially expressed, and their expression levels were further validated using qPCR. This study is one of the many steps we aim to take on the way to promote the rare minnow to an international standard laboratory animal.
Collapse
Affiliation(s)
- Lixia Shi
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Key Lab of Freshwater Biodiversity Conservation Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, CAFS, Wuhan, 430223, China
- School of Life Sciences, Huizhou University, Huizhou, 516007, China
| | - Xudong Hu
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nenghan Wang
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenxi Wu
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Hong Cao
- The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
- Key Lab of Freshwater Biodiversity Conservation Ministry of Agriculture and Rural Affairs, Yangtze River Fisheries Research Institute, CAFS, Wuhan, 430223, China.
| |
Collapse
|
15
|
Dey T, Ghosh A, Mishra S, Pal PK, Chattopadhyay A, Pattari SK, Bandyopadhyay D. Attenuation of arsenic induced high fat diet exacerbated oxidative stress mediated hepatic and cardiac injuries in male Wistar rats by piperine involved antioxidative mechanisms. Food Chem Toxicol 2020; 142:111477. [PMID: 32525072 DOI: 10.1016/j.fct.2020.111477] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/02/2020] [Accepted: 05/27/2020] [Indexed: 11/28/2022]
Abstract
The current study explored the efficacy of piperine in attenuating arsenic induced high fat diet aggravated oxidative stress mediated injury in hepatic and cardiac tissues of male Wistar rats. Oral administration of piperine significantly (p < 0.05) reduced the levels of organ specific and oxidative stress biomarkers in arsenic and high fat diet treated rat hepatic and cardiac tissues in a dose dependant manner with the dose of 60 mg/kg b.w. exhibiting maximum protection. Arsenic induced high fat diet aggravated oxidative stress mediated damages in liver and heart tissues led to decreased activities of antioxidant enzymes, ROS generation, diminished activities of Krebs' cycle and respiratory chain enzymes, collapsed mitochondrial membrane potential, mitochondrial DNA damage along with altered lipid metabolism and inflammatory cytokine levels. Histochemical and histopathological studies supported the above findings. Piperine efficiently counteracted the arsenic induced high fat diet aggravated oxidative stress mediated damages by modulating antioxidant defense mechanism along with free radical quenching ability. These findings indicate that piperine protected the arsenic induced high fat diet aggravated hepatic and cardiac injuries which underline the importance of piperine in providing a possible therapeutic regime for the amelioration of arsenic-induced high fat diet aggravated oxidative stress mediated organ damages.
Collapse
Affiliation(s)
- Tiyasa Dey
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata, 700009, India
| | - Auroma Ghosh
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata, 700009, India
| | - Sanatan Mishra
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata, 700009, India; Department of Physiology, Vidyasagar College, 39, Sankar Ghosh Lane, Kolkata, 700006, India
| | - Palash Kumar Pal
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata, 700009, India
| | - Aindrila Chattopadhyay
- Department of Physiology, Vidyasagar College, 39, Sankar Ghosh Lane, Kolkata, 700006, India
| | - Sanjib K Pattari
- R. N. Tagore International Institute of Cardiac Sciences, Mukundapur, Kolkata, 700099, India
| | - Debasish Bandyopadhyay
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata, 700009, India.
| |
Collapse
|
16
|
Abstract
Exposure to arsenic in contaminated drinking water is a worldwide public health problem that affects more than 200 million people. Protein quality control constitutes an evolutionarily conserved mechanism for promoting proper folding of proteins, refolding of misfolded proteins, and removal of aggregated proteins, thereby maintaining homeostasis of the proteome (i.e., proteostasis). Accumulating lines of evidence from epidemiological and laboratory studies revealed that chronic exposure to inorganic arsenic species can elicit proteinopathies that contribute to neurodegenerative disorders, cancer, and type II diabetes. Here, we review the effects of arsenic exposure on perturbing various elements of the proteostasis network, including mitochondrial homeostasis, molecular chaperones, inflammatory response, ubiquitin-proteasome system, autophagy, as well as asymmetric segregation and axonal transport of misfolded proteins. We also discuss arsenic-induced disruptions of post-translational modifications of proteins, for example, ubiquitination, and their implications in proteostasis. Together, studies in the past few decades support that disruption of protein quality control may constitute an important mechanism underlying the arsenic-induced toxicity.
Collapse
|
17
|
Sathua K, Srivastava S, Flora SJS. MiADMSA ameliorate arsenic induced urinary bladder carcinogenesis in vivo and in vitro. Biomed Pharmacother 2020; 128:110257. [PMID: 32474354 DOI: 10.1016/j.biopha.2020.110257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/05/2020] [Accepted: 05/10/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Arsenicosis is a major threat to public health and is a major cause of the development of urinary bladder cancer. Oxidative/ nitrosative stress is one of the key factors for these effects but the involvement of other associated factors is less known. There is a lack of data for the efficacy of chelator against urinary bladder carcinogenesis. The present study demonstrates the early signs of arsenic exposed urinary bladder carcinogenesis and its attenuation by Monoisoamyl dimercaptosuccinic acid (MiADMSA). METHODS Male rats were exposed to 50 ppm of sodium arsenite and dimethylarsinic acid (DMA) via drinking water for 18 weeks and treated with MiADMSA (50 mg/kg, orally once daily for 5 days) for 3 weeks with a gap one week between the two courses of treatments. We compared in vivo data with in vitro by co-exposing 100 nM of sodium arsenite and DMA to rat (NBT-II) as well as human transitional epithelial carcinoma (T-24) cells with 100 nM of MiADMSA. RESULTS The data showed that sodium arsenite and DMA exposure significantly increased the tissue arsenic contents, ROS, TBARS levels, catalase, SOD activities and significantly decreased GSH level which might be responsible for an increased 8-OHdG level. These changes might have increased pro-oncogenic biomarkers like MMP-9 and survivin in serum, bladder tissues, NBT-II, and T-24 cells. High cell migration and clonogenic potential in NBT-II and T-24 cells exposed to arsenic suggest pronounced carcinogenic potential. Significant recovery in these biomarkers was noted on treatment with MiADMSA. CONCLUSION Early signs of urinary bladder carcinogenesis were observed in arsenic and DMA exposed rats which were linked to metal accumulation, oxidative/ nitrosative stress, 8-OHdG, MMP-9 and survivin which were reduced by MiADMSA possibly via its efficient chelation abilities in vivo and in vitro.
Collapse
Affiliation(s)
- Kshirod Sathua
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, U.P., 226002, India
| | - Sakshi Srivastava
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, U.P., 226002, India
| | - S J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, U.P., 226002, India.
| |
Collapse
|
18
|
Pei F, Wang Y, Fang Y, Li P, Yang W, Ma N, Ma G, Hu Q. Concentrations of heavy metals in muscle and edible offal of pork in Nanjing city of China and related health risks. J Food Sci 2020; 85:493-499. [PMID: 31985835 DOI: 10.1111/1750-3841.15014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/28/2019] [Accepted: 11/14/2019] [Indexed: 02/03/2023]
Abstract
The concentrations of heavy metals, such as chromium (Cr), arsenic (As), cadmium (Cd), mercury (Hg), and lead (Pb), in different muscles and edible offal of consumed pork from the city of Nanjing and related health risks were evaluated. The results showed that the detected Hg concentrations from 6 of 80 samples exceeded the maximum allowable concentration (GB 2762-2017). Moreover, most of the edible offal, especially the kidneys, contained more heavy metals than the other parts, although the concentrations among the samples were significantly different (P < 0.05). In addition, the health risk evaluation revealed that the estimated daily intake of all the samples was far below the recommended limit, and all the target hazard quotients and total target hazard quotients were less than 1, which is considered safe for human health. Nevertheless, the Hg concentrations over the acceptable limit should receive sufficient attention, considering the long-term consumption of pork. PRACTICAL APPLICATION: Currently, no reports have been available on the heavy metal assessment of pork, though pork is widely consumed in many non-Muslim communities. In this study, the concentrations of heavy metals in different muscles and edible offal of consumed pork were determined and their related health risks were evaluated. This study will provide a significant reference to understand the quality and safety of pork in China or other similar developing countries.
Collapse
Affiliation(s)
- Fei Pei
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Yueying Wang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Yong Fang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Peng Li
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Wenjian Yang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Ning Ma
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Gaoxing Ma
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| | - Qiuhui Hu
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing Univ. of Finance and Economics, Nanjing, 210023, China
| |
Collapse
|
19
|
Patwa J, Thakur A, Sharma A, Flora SJS. Monoisoamyl DMSA reduced copper-induced neurotoxicity by lowering 8-OHdG level, amyloid beta and Tau protein expressions in Sprague-Dawley rats. Metallomics 2020; 12:1428-1448. [DOI: 10.1039/d0mt00083c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Copper dyshomeostasis has long been linked with several neurodegenerative disorders.
Collapse
Affiliation(s)
- Jayant Patwa
- Department of Pharmacology and Toxicology
- National Institute of Pharmaceutical Education and Research (NIPER-R)
- Transit Campus
- Near CRPF Camp
- Lucknow
| | - Ashima Thakur
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER-R)
- Transit Campus
- Lucknow
- India
| | - Abha Sharma
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER-R)
- Transit Campus
- Lucknow
- India
| | - S. J. S. Flora
- Department of Pharmacology and Toxicology
- National Institute of Pharmaceutical Education and Research (NIPER-R)
- Transit Campus
- Near CRPF Camp
- Lucknow
| |
Collapse
|
20
|
Wang Y, Zhao H, Yang X, Mu M, Zong H, Luo L, Xing M. Excessive Cu 2+ deteriorates arsenite-induced apoptosis in chicken brain and resulting in immunosuppression, not in homeostasis. CHEMOSPHERE 2020; 239:124758. [PMID: 31514009 DOI: 10.1016/j.chemosphere.2019.124758] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 08/31/2019] [Accepted: 09/03/2019] [Indexed: 06/10/2023]
Abstract
Trace elements such as copper (Cu) and arsenic (As) are two of the major contaminants and well-known inducers of cognitive deficits and neurobehavioral changes. This study evaluated the immunotoxicity of their individual or combined exposure on different brain regions in chickens. Consequently, nuclear damage and organelle lesions, especially mitochondria were observed under Cu or/and As stress, in which positive regulation of key proteins, dynamin-related protein 1 (Drp1), Cytochrome C (Cyt c), BCL2-associated X (Bax), Caspases 3 and P53 was detected by qRCR and Western blot analyses, indicating disturbed mitochondrial dynamic equilibrium and apoptosis execution. In addition, qRCR analysis confirmed the involvement of cytokines secreted by different populations of helper T cells, indicative of cellular immunity. Gene expression studies showed marked up regulation of Th1/Th17 cytokines along with heat shock protein (HSP) 70, a synergism was noted in co-administration group. Interesting, lower apoptosis index was noted in brainstem compared to cerebrum and cerebellum. An intense immunosuppression and heat shock response against Cu or/and As was also seen in cerebrum and cerebellum but not in brainstem. In conclusion, our study suggests a synergistic neurotoxicity in chickens under Cu and As exposure. These findings provide a basic understanding of mitochondrial abnormality-initiated neuropathology in response to environmental pollutant mixtures, suggesting an adaptive response to the frangibility of the central nerve system.
Collapse
Affiliation(s)
- Yu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| | - Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| | - Xin Yang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Mengyao Mu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Hui Zong
- Guangdong Vocational College of Science and Trade, Guangzhou, PR China
| | - Liyang Luo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| |
Collapse
|
21
|
Javadipour M, Rezaei M, Keshtzar E, Khodayar MJ. Metformin in contrast to berberine reversed arsenic‐induced oxidative stress in mitochondria from rat pancreas probably via Sirt3‐dependent pathway. J Biochem Mol Toxicol 2019; 33:e22368. [DOI: 10.1002/jbt.22368] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Mansoureh Javadipour
- Toxicology Research CenterAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Toxicology, Faculty of PharmacyAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Mohsen Rezaei
- Toxicology Research CenterAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Toxicology, Faculty of PharmacyAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Toxicology, Faculty of Medical SciencesTarbiat Modares University Tehran Iran
| | - Elham Keshtzar
- Toxicology Research CenterAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Toxicology, Faculty of PharmacyAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Mohammad Javad Khodayar
- Toxicology Research CenterAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Toxicology, Faculty of PharmacyAhvaz Jundishapur University of Medical Sciences Ahvaz Iran
| |
Collapse
|
22
|
Niño SA, Morales-Martínez A, Chi-Ahumada E, Carrizales L, Salgado-Delgado R, Pérez-Severiano F, Díaz-Cintra S, Jiménez-Capdeville ME, Zarazúa S. Arsenic Exposure Contributes to the Bioenergetic Damage in an Alzheimer's Disease Model. ACS Chem Neurosci 2019; 10:323-336. [PMID: 30141907 DOI: 10.1021/acschemneuro.8b00278] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Worldwide, every year there is an increase in the number of people exposed to inorganic arsenic (iAs) via drinking water. Human populations present impaired cognitive function as a result of prenatal and childhood iAs exposure, while studies in animal models demonstrate neurobehavioral deficits accompanied by neurotransmitter, protein, and enzyme alterations. Similar impairments have been observed in close association with Alzheimer's disease (AD). In order to determine whether iAs promotes the pathophysiological progress of AD, we used the 3xTgAD mouse model. Mice were exposed to iAs in drinking water from gestation until 6 months (As-3xTgAD group) and compared with control animals without arsenic (3xTgAD group). We investigated the behavior phenotype on a test battery (circadian rhythm, locomotor behavior, Morris water maze, and contextual fear conditioning). Adenosine triphosphate (ATP), reactive oxygen species, lipid peroxidation, and respiration rates of mitochondria were evaluated, antioxidant components were detected by immunoblots, and immunohistochemical studies were performed to reveal AD markers. As-3xTgAD displayed alterations in their circadian rhythm and exhibited longer freezing time and escape latencies compared to the control group. The bioenergetic profile revealed decreased ATP levels accompanied by the decline of complex I, and an oxidant state in the hippocampus. On the other hand, the cortex showed no changes of oxidant stress and complex I; however, the antioxidant response was increased. Higher immunopositivity to amyloid isoforms and to phosphorylated tau was observed in frontal cortex and hippocampus of exposed animals. In conclusion, mitochondrial dysfunction may be one of the triggering factors through which chronic iAs exposure exacerbates brain AD-like pathology.
Collapse
Affiliation(s)
- Sandra Aurora Niño
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, CP 78210 San Luis Potosí, SLP, México
| | - Adriana Morales-Martínez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur 3877, CP 14269, México D.F., México
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosi, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Leticia Carrizales
- Centro de Investigación Aplicada en Ambiente y Salud, CIACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Roberto Salgado-Delgado
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava Martínez S/N, CP 78290 San Luis Potosí, SLP, Mexico
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur 3877, CP 14269, México D.F., México
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, CP 76230 Juriquilla, Querétaro, México
| | - María E. Jiménez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosi, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Sergio Zarazúa
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, CP 78210 San Luis Potosí, SLP, México
| |
Collapse
|
23
|
Chandravanshi LP, Gupta R, Shukla RK. Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions. Biol Trace Elem Res 2018; 186:185-198. [PMID: 29502250 DOI: 10.1007/s12011-018-1286-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/23/2018] [Indexed: 12/31/2022]
Abstract
Over the last decade, there has been an increased concern about the health risks from exposure to arsenic at low doses, because of their neurotoxic effects on the developing brain. The exact mechanism underlying arsenic-induced neurotoxicity during sensitive periods of brain development remains unclear, although enhanced oxidative stresses, leading to mitochondrial dysfunctions might be involved. Here, we highlight the generation of reactive oxygen species (ROS) and oxidative stress which leads to mitochondrial dysfunctions and apoptosis in arsenic-induced developmental neurotoxicity. Here, the administration of sodium arsenite at doses of 2 or 4 mg/kg body weight in female rats from gestational to lactational (GD6-PD21) resulted to increased ROS, led to oxidative stress, and increased the apoptosis in the frontal cortex, hippocampus, and corpus striatum of developing rats on PD22, compared to controls. Enhanced levels of ROS were associated with decreased mitochondrial membrane potential and the activity of mitochondrial complexes, and hampered antioxidant levels. Further, neuronal apoptosis, as measured by changes in the expression of pro-apoptotic (Bax, Caspase-3), anti-apoptotic (Bcl2), and stress marker proteins (p-p38, pJNK) in arsenic-exposed rats, was discussed. The severities of changes were found to more persist in the corpus striatum than in other brain regions of arsenic-exposed rats even after the withdrawal of exposure on PD45 as compared to controls. Therefore, our results indicate that perinatal arsenic exposure leads to abrupt changes in ROS, oxidative stress, and mitochondrial functions and that apoptotic factor in different brain regions of rats might contribute to this arsenic-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Lalit P Chandravanshi
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research, Post Box No. 80, MG Marg, Lucknow, 226 001, India.
| | - Richa Gupta
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research, Post Box No. 80, MG Marg, Lucknow, 226 001, India
| | - Rajendra K Shukla
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| |
Collapse
|
24
|
Kushwaha P, Yadav A, Samim M, Flora SJS. Combinatorial drug delivery strategy employing nano-curcumin and nano-MiADMSA for the treatment of arsenic intoxication in mouse. Chem Biol Interact 2018; 286:78-87. [PMID: 29548727 DOI: 10.1016/j.cbi.2018.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/23/2018] [Accepted: 03/13/2018] [Indexed: 12/27/2022]
Abstract
Chelation therapy is the mainstream treatment for heavy metal poisoning. Apart from this, therapy using antioxidant/herbal extracts are the other strategies now commonly being tried for the treatment. We have previously reported individual beneficial efficacy of nanoparticle mediated administration of an antioxidant like 'curcumin' and an arsenic chelator 'monoisoamyl 2,3-dimercaptosuccinic acid (MiADMSA)' for the treatment of arsenic toxicity compared to bulk drugs. The present paper investigates our hypothesis that a combination drug delivery therapy employing two nanosystems, a chelator and a strong antioxidant, may produce more pronounced therapeutic effects compared to individual effects in the treatment of arsenic toxicity. An in-vivo study was conducted wherein arsenic as sodium arsenite (100 ppm) was administered in drinking water for 5 months to Swiss albino mice. This was followed by a treatment protocol comprising of curcumin encapsulated chitosan nanoparticles (nano-curcumin, 15 mg/kg, orally for 1 month) either alone or in combination with MiADMSA encapsulated polymeric nanoparticles (nano-MiADMSA, 50 mg/kg for last 5 days) to evaluate the therapeutic potential of the combination treatment. Our results demonstrated that co-treatment with nano-curcumin and nano-MiADMSA provided beneficial effects in a synergistic way on the adverse changes in oxidative stress parameters and metal status induced by arsenic.
Collapse
Affiliation(s)
- Pramod Kushwaha
- Division of Regulatory Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474002, M.P., India
| | - Abhishek Yadav
- Division of Regulatory Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474002, M.P., India
| | - M Samim
- Jamia Hamdard, New Delhi, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research, Raebareli 209010, U.P., India.
| |
Collapse
|
25
|
Thymoquinone alleviates arsenic induced hippocampal toxicity and mitochondrial dysfunction by modulating mPTP in Wistar rats. Biomed Pharmacother 2018; 102:1152-1160. [PMID: 29710533 DOI: 10.1016/j.biopha.2018.03.159] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Arsenic is a pervasive environmental pollutant that is found in ground waters globally and is related to numerous morbidities in the high-risk population areas in countries including Bangladesh, India, and the USA. Arsenic exposure has been ubiquitously reported for exacerbating free radical generation, mitochondrial dysfunction, and apoptosis by interfering with the mPTP functioning. Over the past decades, nutraceuticals with antioxidant properties have shown promising efficacy in arsenic poisoning. In the present study, we have examined, the protective efficacy of thymoquinone (TQ), an active component of seed oil of Nigella sativa with antioxidant and anti-inflammatory activity on arsenic-induced toxicity in hippocampi of Wistar rats. In our results, arsenic conditioning (10 mg/kg b.wt.; p.o.) for 8 days has caused a significant increase in intracellular ROS generation, mitochondrial dysfunction and apoptotic events. On the contrary pretreatment with TQ (2.5 and 5 mg/kg b.wt.; p.o.) inhibited arsenic-induced mitochondrial dysfunction such as lowering of mitochondrial membrane potential (Δψm). Our results indicated that the neuroprotective efficacy of TQ in arsenic-induced stress is mediated through or in part by inhibition of mPTP opening. Demonstration of neuroprotective action of TQ provides insight into the pathogenesis of arsenic-related neuropathological morbidities.
Collapse
|
26
|
Sun H, Yang Y, Shao H, Sun W, Gu M, Wang H, Jiang L, Qu L, Sun D, Gao Y. Sodium Arsenite-Induced Learning and Memory Impairment Is Associated with Endoplasmic Reticulum Stress-Mediated Apoptosis in Rat Hippocampus. Front Mol Neurosci 2017; 10:286. [PMID: 28936164 PMCID: PMC5594089 DOI: 10.3389/fnmol.2017.00286] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
Chronic arsenic exposure has been associated to cognitive deficits. However, mechanisms remain unknown. The present study investigated the neurotoxic effects of sodium arsenite in drinking water over different dosages and time periods. Based on results from the Morris water maze (MWM) and morphological analysis, an exposure to sodium arsenite could induce neuronal damage in the hippocampus, reduce learning ability, and accelerate memory impairment. Sodium arsenite significantly increased homocysteine levels in serum and brain. Moreover, sodium arsenite triggered unfolded protein response (UPR), leading to the phosphorylation of RNA-regulated protein kinase-like ER kinase (PERK) and eukaryotic translation initiation factor 2 subunit α (eIF2α), and the induction of activating transcription factor 4 (ATF4). Arsenite exposure also stimulated the expression of the endoplasmic reticulum (ER) stress markers, glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and the cleavage of caspase-12. Furthermore, exposure to arsenite enhanced apoptosis as demonstrated by expression of caspase-3 and TUNEL assay in the hippocampus. The results suggest that exposure to arsenite can significantly decrease learning ability and accelerate memory impairment. Potential mechanisms are related to enhancement of homocysteine and ER stress-induced apoptosis in the hippocampus.
Collapse
Affiliation(s)
- Hongna Sun
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Yanmei Yang
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Hanwen Shao
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Weiwei Sun
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Muyu Gu
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Hui Wang
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Lixin Jiang
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Lisha Qu
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Dianjun Sun
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China
| | - Yanhui Gao
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin, China.,Institution of Environmentally Related Diseases, Harbin Medical UniversityHarbin, China
| |
Collapse
|
27
|
Srivastava P, Dhuriya YK, Gupta R, Shukla RK, Yadav RS, Dwivedi HN, Pant AB, Khanna VK. Protective Effect of Curcumin by Modulating BDNF/DARPP32/CREB in Arsenic-Induced Alterations in Dopaminergic Signaling in Rat Corpus Striatum. Mol Neurobiol 2016; 55:445-461. [DOI: 10.1007/s12035-016-0288-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/02/2016] [Indexed: 01/04/2023]
|
28
|
Keshtzar E, Khodayar MJ, Javadipour M, Ghaffari MA, Bolduc DL, Rezaei M. Ellagic acid protects against arsenic toxicity in isolated rat mitochondria possibly through the maintaining of complex II. Hum Exp Toxicol 2016; 35:1060-72. [DOI: 10.1177/0960327115618247] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic arsenic exposure has been linked to many health problems including diabetes and cancer. In the present study, we assessed the protective effect of ellagic acid (EA) against toxicity induced by arsenic in isolated rat liver mitochondria. Reactive oxygen species (ROS) and mitochondrial membrane potential decline were assayed using dichlorofluorescein diacetate and rhodamine 123, respectively, and dehydrogenase activity obtained by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide conversion assay. Arsenic increased ROS levels and mitochondrial dysfunction, which led to a reduction in mitochondrial total dehydrogenase activity. Mitochondria pretreated with EA exposed to arsenic at various concentrations led to a reversal of ROS production and mitochondrial damage. Our results showed that mitochondria were significantly affected when exposed to arsenic, which resulted in excessive ROS production and mitochondrial membrane disruption. Pretreatment with EA, reduced ROS amounts, mitochondrial damage, and restored total dehydrogenase activity specifically associated with mitochondrial complex II. EA protective characteristics may be accomplished particularly throughout the mitochondrial maintenance either directly by its antioxidant property or indirectly through its maintaining of complex II. These findings also suggest a potential role for EA in treating or preventing mitochondria associated disorders.
Collapse
Affiliation(s)
- E Keshtzar
- Diabetes Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - MJ Khodayar
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - M Javadipour
- Diabetes Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - MA Ghaffari
- Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - DL Bolduc
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - M Rezaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
29
|
Prakash C, Kumar V. Arsenic-induced mitochondrial oxidative damage is mediated by decreased PGC-1α expression and its downstream targets in rat brain. Chem Biol Interact 2016; 256:228-35. [PMID: 27425645 DOI: 10.1016/j.cbi.2016.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/21/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022]
Abstract
The present study was carried out to investigate the molecular mechanism of arsenic-induced mitochondrial oxidative damage and its relation to biogenesis in rat brain. Chronic sodium arsenite (25 ppm, orally) administration for 12 weeks decreased mitochondrial complexes activities and mRNA expression of selective complexes subunits. The expression of mitochondrial biogenesis regulator PGC-1α, and its downstream targets NRF-1, NRF-2 and Tfam were decreased significantly both at mRNA and protein levels suggesting impaired biogenesis following chronic arsenic-exposure. In addition to this, protein expression analysis also revealed activation of Bax and caspase-3, leading to translocation of cytochrome c from mitochondria to cytosol suggesting induction of apoptotic pathway under oxidative stress. This was further confirmed by electron microscopy study which depicted morphological changes in mitochondria in terms of altered nuclear and mitochondrial shape and chromatin condensation in arsenic-treated rats. The immunohistochemical studies showed both nuclear and cytosolic localization of NRF-1 and NRF-2 in arsenic-exposed rat brain further suggesting regulatory role of these transcription factors under arsenic neurotoxicity. The results of present study indicate that arsenic-induced mitochondrial oxidative damage is associated with decreased mitochondrial biogenesis in rat brain that may present as important target to reveal the mechanism for arsenic-induced neurotoxicity.
Collapse
Affiliation(s)
- Chandra Prakash
- Department of Biochemistry, Maharshi Dayanand University, Rohtak 124001, Haryana, India
| | - Vijay Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak 124001, Haryana, India.
| |
Collapse
|
30
|
Inorganic Arsenic Induces NRF2-Regulated Antioxidant Defenses in Both Cerebral Cortex and Hippocampus in Vivo. Neurochem Res 2016; 41:2119-28. [PMID: 27165637 DOI: 10.1007/s11064-016-1927-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/15/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
Inorganic arsenic is reported to induce the reactive oxygen species-mediated oxidative stress, which is supposed to be one of the main mechanisms of arsenic-related neurological diseases. Nuclear factor erythroid 2-related factor 2 (NRF2), a master regulator of antioxidant defense systems, up-regulates the expression of target genes to fight against oxidative damages caused by harmful substances, including metals. In the present study, mice were used as a model to investigate the oxidative stress levels and the expressions of NRF2-regulated antioxidant substances in both cerebral cortex and hippocampus with 5, 10 and 20 mg/kg NaAsO2 exposure intra-gastrically. Our results showed that acute NaAsO2 treatment resulted in decreased total anti-oxidative capacity (T-AOC) and increased maleic dialdehyde production in the nervous system. We also detected rapidly elevation of NRF2 protein levels by enhancement of Nrf2 transcription, especially at 20 mg/kg NaAsO2 exposure group. In the meantime, mRNA and protein levels of Nrf2 encoding antioxidant enzymes heme oxygenase-1 (HO-1), NAD(P)H: quinine oxidoreductase 1 (NQO1) and glutathione S-transferase (GST) were consistently elevated time- and dose-dependently both in the cerebral cortex and hippocampus. Taken together, the presence study demonstrated the activation of NRF2 pathway, an early antioxidant defensive response, in both cerebral cortex and hippocampus upon inorganic arsenic (iAs) exposure in vivo. A better knowledge on the roles of NRF2 pathway in maintaining cellular redox homeostasis would be helpful for the strategies on improvement of neurotoxicity related to this metalloid.
Collapse
|
31
|
Yadav A, Flora SJS. Nano drug delivery systems: a new paradigm for treating metal toxicity. Expert Opin Drug Deliv 2016; 13:831-41. [PMID: 27030893 DOI: 10.1517/17425247.2016.1160890] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION The standard medical treatment for metal toxicity is chelation therapy. Chelating agents work by forming less toxic complexes with the toxic metal ions which are readily excreted from the body. These compounds, based on their hydrophilic/lipophilic property, can either remove toxic metal ions from extracellular sites or can penetrate the intracellular compartments to facilitate the removal of toxic metal ions. However, there are various disadvantages associated with this kind of therapy, notably, selectivity. Other problems and challenges are that the therapy regime is expensive, time consuming and has poor patient compliance. Two chelating agents, dimercaptosuccinic acid (DMSA) and dimercaptopropionicsulfonate (DMPS) have gained increased acceptance among clinicians, undoubtedly improving the management of metal intoxications. AREAS COVERED The present review provides an insight into the conventional chelating agents, new chelators under development, and the new opportunities presented by the use of nanotherapy for the treatment of metal poisoning cases. EXPERT OPINION Today's research should not only focus towards development of alternate chelators but also targeted therapy such as the nanotherapy.
Collapse
Affiliation(s)
- Abhishek Yadav
- a Directorate of Planning & Coordination , Defence Research and Development Organisation, Ministry of Defence, Govt. of India , Delhi , India
| | - Swaran Jeet Singh Flora
- b Division of Regulatory Toxicology , Defence Research and Development Establishment , Gwalior , Madhya Pradesh , India
| |
Collapse
|
32
|
Prakash C, Soni M, Kumar V. Mitochondrial oxidative stress and dysfunction in arsenic neurotoxicity: A review. J Appl Toxicol 2015; 36:179-88. [DOI: 10.1002/jat.3256] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/01/2015] [Accepted: 09/28/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Chandra Prakash
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| | - Manisha Soni
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| | - Vijay Kumar
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| |
Collapse
|
33
|
Prakash C, Soni M, Kumar V. Biochemical and Molecular Alterations Following Arsenic-Induced Oxidative Stress and Mitochondrial Dysfunction in Rat Brain. Biol Trace Elem Res 2015; 167:121-9. [PMID: 25764338 DOI: 10.1007/s12011-015-0284-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/24/2015] [Indexed: 01/08/2023]
Abstract
Oxidative stress is associated with the generation of reactive oxygen species (ROS), which is supposed to be one of the mechanisms of arsenic-induced neurodegeneration. Mitochondria, being the major source of ROS generation may present an important target of arsenic-mediated neurotoxicity. Hence, we planned the study to elucidate the possible biochemical and molecular alterations induced by arsenic exposure in rat brain mitochondria. Chronic sodium arsenite treatment (25 ppm for 12 weeks) resulted in decreased activity of mitochondrial complexes I, II, and IV followed by increased ROS generation. There was decrease in mitochondrial superoxide dismutase (MnSOD) activity in arsenic-treated rat brain further showing increased superoxide radical generation in mitochondria. The decrease in MnSOD activity might be responsible for the increased protein and lipid oxidation as observed in our study. Protein and messenger RNA (mRNA) levels of MnSOD and mitochondrial uncoupling protein 2 (UCP-2) were downregulated suggesting decreased removal of ROS in rat brain. Fourier transform infrared (FTIR) spectroscopy analysis revealed significant decrease in amide A, amide I, amide II, and Olefinic = CH stretching band area suggesting molecular alteration in proteins and lipids after arsenic treatment. The results of present study indicate that arsenic-induced disturbed mitochondrial metabolism, decreased removal of ROS, decrease in protein synthesis, and altered membrane lipid polarity and fluidity may be responsible for the mitochondrial oxidative damage in rat brain that may further be implicated as contributing factor in arsenic-induced neurodegeneration.
Collapse
Affiliation(s)
- Chandra Prakash
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | | | | |
Collapse
|
34
|
Pachauri V, Flora S. Combined Efficacy of Gallic Acid and MiADMSA with Limited Beneficial Effects Over MiADMSA Against Arsenic-induced Oxidative Stress in Mouse. BIOCHEMISTRY INSIGHTS 2015; 8:1-10. [PMID: 26339189 PMCID: PMC4552275 DOI: 10.4137/bci.s30505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/26/2015] [Accepted: 08/01/2015] [Indexed: 11/05/2022]
Abstract
Gallic acid is an organic acid known for its antioxidant and anticancer properties. The present study is focused on evaluating the role of gallic acid in providing better therapeutic outcomes against arsenic-induced toxicity. Animals pre-exposed to arsenic were treated with monoisoamyl meso-2,3-dimercaptosuccinic acid (MiADMSA), a new chelating drug, alone and in combination with gallic acid, consecutively for 10 days. The study suggests that (1) gallic acid in presence of MiADMSA is only moderately beneficial against arsenic, (2) monotherapy with gallic acid is more effective than in combination with MiADMSA after arsenic exposure in reducing oxidative injury, and (3) MiADMSA monotherapy as reported previously provides significant therapeutic efficacy against arsenic. Thus, based on the present results, we conclude that gallic acid is effective against arsenic-induced oxidative stress but provides limited additional beneficial effects when administered in combination with MiADMSA. We still recommend that lower doses of gallic acid be evaluated both individually and in combination with MiADMSA, as it might not exhibit the shortcomings we observed with higher doses in this study.
Collapse
Affiliation(s)
- Vidhu Pachauri
- Division of Regulatory Toxicology, Defence Research and Development Establishment, Gwalior, India
| | - Sjs Flora
- Division of Regulatory Toxicology, Defence Research and Development Establishment, Gwalior, India
| |
Collapse
|
35
|
Teng YC, Tai YI, Huang HJ, Lin AMY. Melatonin Ameliorates Arsenite-Induced Neurotoxicity: Involvement of Autophagy and Mitochondria. Mol Neurobiol 2015; 52:1015-22. [DOI: 10.1007/s12035-015-9250-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Qi Y, Li SY, Piao FY, Wang ZM, Chen RL, Liu S, Shen JS. 2,5-Hexanedione induces apoptosis via a mitochondria-mediated pathway in PC12 cells. Mol Cell Toxicol 2015. [DOI: 10.1007/s13273-015-0010-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
37
|
Aung KH, Tsukahara S, Maekawa F, Nohara K, Nakamura K, Tanoue A. Role of Environmental Chemical Insult in Neuronal Cell Death and Cytoskeleton Damage. Biol Pharm Bull 2015; 38:1109-12. [DOI: 10.1248/bpb.b14-00890] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kyaw Htet Aung
- Department of Pharmacology, National Research Institute for Child Health and Development
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Fumihiko Maekawa
- Molecular Toxicology Section, Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Keiko Nohara
- Molecular Toxicology Section, Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development
| | - Akito Tanoue
- Department of Pharmacology, National Research Institute for Child Health and Development
| |
Collapse
|
38
|
CHEN R, LIU S, PIAO F, WANG Z, QI Y, LI S, ZHANG D, SHEN J. 2,5-hexanedione induced apoptosis in mesenchymal stem cells from rat bone marrow via mitochondria-dependent caspase-3 pathway. INDUSTRIAL HEALTH 2015; 53:222-35. [PMID: 25739802 PMCID: PMC4466875 DOI: 10.2486/indhealth.2014-0182] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
2,5-hexanedione (HD) induces apoptosis of nerve cells. However,the mechanism of HD-induced apoptosis remains unknown. Mesenchymal stem cells (MSCs) are multipotential stem cells with the ability to differentiate into various cell types. This study is designed to investigate the apoptosis induced by HD in rat bone marrow MSCs (BMSCs) and the related underlying mechanisms. The fifth generation of MSCs was treated with 0, 10, 20 and 40 mM HD respectively. The viability of BMSCs was observed by MTT. Apoptosis were estimated by Hoechst 33342 staining and TUNEL assay. The disruption of mitochondrial transmembrane potential (MMP) was examined by JC-1 staining. Moreover, the expression of Bax and Bcl-2, cytochrome c release, and caspase-3 activity were determined by real-time RT-PCR, Western blot and Spectrophotometry. Our results showed that HD induced apoptosis in MSCs in a dose dependent manner. Moreover, HD downregulated the Bcl-2 expression,upregulated the Bax expression and the Bax/Bcl-2 ratio, promoted the disruption of MMP, induced the release of cytochrome c from mitochondria to cytosol, and increased the activity of caspase-3 in MSCs. These results indicate that HD induces apoptosis in MSCs and the activated mitochondria-dependent caspase-3 pathway may be involved in the HD-induced apoptosis.
Collapse
Affiliation(s)
- Ruolin CHEN
- Department of Occupational and Environmental Health, Dalian
Medical University, China
| | - Shuang LIU
- Department of Occupational and Environmental Health, Dalian
Medical University, China
| | - Fengyuan PIAO
- Department of Occupational and Environmental Health, Dalian
Medical University, China
- To whom correspondence should be addressed. E-mail: (F.
Piao); (J.
Shen)
| | - Zhemin WANG
- Department of Occupational and Environmental Health, Dalian
Medical University, China
| | - Yuan QI
- Department of Occupational and Environmental Health, Dalian
Medical University, China
| | - Shuangyue LI
- Department of Occupational and Environmental Health, Dalian
Medical University, China
| | - Dongmei ZHANG
- Department of Physiology, Dalian Medical University,
China
| | - Jingshun SHEN
- Department of Neurology, First Affiliated Hospital of Dalian
Medical University, China
- To whom correspondence should be addressed. E-mail: (F.
Piao); (J.
Shen)
| |
Collapse
|
39
|
Abstract
Metals are frequently used in industry and represent a major source of toxin exposure for workers. For this reason governmental agencies regulate the amount of metal exposure permissible for worker safety. While essential metals serve physiologic roles, metals pose significant health risks upon acute and chronic exposure to high levels. The central nervous system is particularly vulnerable to metals. The brain readily accumulates metals, which under physiologic conditions are incorporated into essential metalloproteins required for neuronal health and energy homeostasis. Severe consequences can arise from circumstances of excess essential metals or exposure to toxic nonessential metal. Herein, we discuss sources of occupational metal exposure, metal homeostasis in the human body, susceptibility of the nervous system to metals, detoxification, detection of metals in biologic samples, and chelation therapeutic strategies. The neurologic pathology and physiology following aluminum, arsenic, lead, manganese, mercury, and trimethyltin exposures are highlighted as classic examples of metal-induced neurotoxicity.
Collapse
Affiliation(s)
- Samuel Caito
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA; The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN, USA; Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
40
|
Yadav A, Mathur R, Samim M, Lomash V, Kushwaha P, Pathak U, Babbar AK, Flora SJS, Mishra AK, Kaushik MP. Nanoencapsulation of DMSA monoester for better therapeutic efficacy of the chelating agent against arsenic toxicity. Nanomedicine (Lond) 2014; 9:465-81. [PMID: 24910877 DOI: 10.2217/nnm.13.17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS Exposure to toxic metals remains a widespread occupational and environmental problem in world. Chelation therapy is a mainstream treatment used to treat heavy metal poisoning. This paper describes the synthesis, characterization and therapeutic evaluation of monoisoamyl 2,3-dimercaptosuccinic acid (MiADMSA)-encapsulated polymeric nanoparticles as a detoxifying agent for arsenic poisoning. MATERIALS & METHODS Polymeric nanoparticles entrapping the DMSA monoester, which can evade the reticulo-endothelial system and have a long circulation time in the blood, were prepared. Particle characterization was carried out by transmission electron microscopy and dynamic light scattering. An in vivo study was conducted to investigate the therapeutic efficacy of MiADMSA-encapsulated polymeric nanoparticles (nano- MiADMSA; 50 mg/kg orally for 5 days) and comparison drawn with bulk MiADMSA. Swiss albino mice exposed to sodium arsenite for 4 weeks were treated for 5 days to evaluate alterations in blood, brain, kidney and liver oxidative stress variables. The study also evaluated the histopathological changes in tissues and the chelating potential of the nanoformulation. RESULTS Our results show that nano-MiADMSA have a narrow size distribution in the 50-nm range. We observed an enhanced chelating potential of nano-MiADMSA compared with bulk MiADMSA as evident in the reversal of biochemical changes indicative of oxidative stress and efficient removal of arsenic from the blood and tissues. Histopathological changes and urinary 8-OHdG levels also prove better therapeutic efficacy of the novel formulation for arsenic toxicity. CONCLUSION The results from our study show better therapeutic efficacy of nano-MiADMSA in removing arsenic burden from the brain and liver.
Collapse
|
41
|
Early life arsenic exposure and brain dopaminergic alterations in rats. Int J Dev Neurosci 2014; 38:91-104. [DOI: 10.1016/j.ijdevneu.2014.08.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/09/2014] [Accepted: 08/23/2014] [Indexed: 11/21/2022] Open
|
42
|
Srivastava P, Yadav RS, Chandravanshi LP, Shukla RK, Dhuriya YK, Chauhan LKS, Dwivedi HN, Pant AB, Khanna VK. Unraveling the mechanism of neuroprotection of curcumin in arsenic induced cholinergic dysfunctions in rats. Toxicol Appl Pharmacol 2014; 279:428-440. [PMID: 24952339 DOI: 10.1016/j.taap.2014.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/20/2022]
Abstract
Earlier, we found that arsenic induced cholinergic deficits in rat brain could be protected by curcumin. In continuation to this, the present study is focused to unravel the molecular mechanisms associated with the protective efficacy of curcumin in arsenic induced cholinergic deficits. Exposure to arsenic (20mg/kg body weight, p.o) for 28 days in rats resulted to decrease the expression of CHRM2 receptor gene associated with mitochondrial dysfunctions as evident by decrease in the mitochondrial membrane potential, activity of mitochondrial complexes and enhanced apoptosis both in the frontal cortex and hippocampus in comparison to controls. The ultrastructural images of arsenic exposed rats, assessed by transmission electron microscope, exhibited loss of myelin sheath and distorted cristae in the mitochondria both in the frontal cortex and hippocampus as compared to controls. Simultaneous treatment with arsenic (20mg/kg body weight, p.o) and curcumin (100mg/kg body weight, p.o) for 28 days in rats was found to protect arsenic induced changes in the mitochondrial membrane potential and activity of mitochondrial complexes both in frontal cortex and hippocampus. Alterations in the expression of pro- and anti-apoptotic proteins and ultrastructural damage in the frontal cortex and hippocampus following arsenic exposure were also protected in rats simultaneously treated with arsenic and curcumin. The data of the present study reveal that curcumin could protect arsenic induced cholinergic deficits by modulating the expression of pro- and anti-apoptotic proteins in the brain. More interestingly, arsenic induced functional and ultrastructural changes in the brain mitochondria were also protected by curcumin.
Collapse
Affiliation(s)
- Pranay Srivastava
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajesh S Yadav
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India; Department of Crimnology and Forensic Science, Harisingh Gour University, Sagar 470 003, India
| | - Lalit P Chandravanshi
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajendra K Shukla
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Yogesh K Dhuriya
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Lalit K S Chauhan
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Hari N Dwivedi
- Babu Banarasi Das University, BBD City, Faizabad Road, Lucknow 227 015, India
| | - Aditiya B Pant
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Vinay K Khanna
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India.
| |
Collapse
|
43
|
Lu TH, Tseng TJ, Su CC, Tang FC, Yen CC, Liu YY, Yang CY, Wu CC, Chen KL, Hung DZ, Chen YW. Arsenic induces reactive oxygen species-caused neuronal cell apoptosis through JNK/ERK-mediated mitochondria-dependent and GRP 78/CHOP-regulated pathways. Toxicol Lett 2013; 224:130-40. [PMID: 24157283 DOI: 10.1016/j.toxlet.2013.10.013] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 10/26/2022]
Abstract
Arsenic (As), a well-known high toxic metal, is an important environmental and industrial contaminant, and it induces oxidative stress, which causes many adverse health effects and diseases in humans, particularly in inorganic As (iAs) more harmful than organic As. Recently, epidemiological studies have suggested a possible relationship between iAs exposure and neurodegenerative disease development. However, the toxicological effects and underlying mechanisms of iAs-induced neuronal cell injuries are mostly unknown. The present study demonstrated that iAs significantly decreased cell viability and induced apoptosis in Neuro-2a cells. iAs also increased oxidative stress damage (production of malondialdehyde (MDA) and ROS, and reduction of Nrf2 and thioredoxin protein expression) and induced several features of mitochondria-dependent apoptotic signals, including: mitochondrial dysfunction, the activations of PARP and caspase cascades, and the increase in caspase-3 activity. Pretreatment with the antioxidant N-acetylcysteine (NAC) effectively reversed these iAs-induced responses. iAs also increased the phosphorylation of JNK and ERK1/2, but did not that p38-MAPK, in treated Neuro-2a cells. NAC and the specific JNK inhibitor (SP600125) and ERK1/2 inhibitor (PD98059) abrogated iAs-induced cell cytotoxicity, caspase-3/-7 activity, and JNK and ERK1/2 activation. Additionally, exposure of Neuro-2a cells to iAs triggered endoplasmic reticulum (ER) stress identified through several key molecules (GRP 78, CHOP, XBP-1, and caspase-12), which was prevented by NAC. Transfection with GRP 78- and CHOP-specific si-RNA dramatically suppressed GRP 78 and CHOP expression, respectively, and attenuated the activations of caspase-12, -7, and -3 in iAs-exposed cells. Therefore, these results indicate that iAs induces ROS causing neuronal cell death via both JNK/ERK-mediated mitochondria-dependent and GRP 78/CHOP-triggered apoptosis pathways.
Collapse
Affiliation(s)
- Tien-Hui Lu
- Department of Physiology, and Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 404, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Inorganic Arsenic Exposure and Children’s Neurodevelopment: A Review of the Evidence. TOXICS 2013. [DOI: 10.3390/toxics1010002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Ding X, Su Q, Jiang M, Xie H, Cong J, Wang L. Arsenic affects on cerebellar development of mice. Toxicol Mech Methods 2013; 23:672-7. [PMID: 23998907 DOI: 10.3109/15376516.2013.840347] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To evaluate arsenic affects on the development of cerebellum, the mother mice received drinking water containing arsenic trioxide (As2O3) 4 ppm during gestation and lactation period. The cytogenesis was observed by immunohistochemical technique using 5'-bromo-2'-deoxyuridine (BrdU) antibody. To characterize the arsenic neurotoxicity, 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), an oxidative DNA lesion marker, and 8-nitroguanine, a nitrative DNA lesion marker were used to verify DNA damage. Arsenic-exposed litters showed neuron necrosis. The mitosis of granule cells decreased dramatically in arsenic-exposed mice as compared with arsenic unexposed mice. 8-OxodG was formed in neurons of all the layers, especially in the granular layer in cerebellum of arsenic-exposed mice. There is no significant difference, however, in the expression of 8-nitroguanine between arsenic-exposed and -unexposed mice. These results indicate that arsenic can disturb the mitosis of granule cells and interfere with the normal development of mice cerebellum. Arsenic-induced pathological changes in vivo may be caused by oxidative DNA damage other than nitrative DNA damage.
Collapse
Affiliation(s)
- Xiaohui Ding
- Histology and Embryology Department, Shenyang Medical College , Shenyang , China
| | | | | | | | | | | |
Collapse
|
46
|
Monensin potentiates lead chelation efficacy of MiADMSA in rat brain post chronic lead exposure. Food Chem Toxicol 2012; 50:4449-60. [PMID: 22982480 DOI: 10.1016/j.fct.2012.08.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 08/29/2012] [Accepted: 08/30/2012] [Indexed: 01/04/2023]
Abstract
The present study evaluates combination therapy with a chelating agent, MiADMSA and a Na(+) ionophore, monensin against sub-chronic lead toxicity in rats. Animals were exposed to 0.1% lead in drinking water for 16 weeks and then treated with either MiADMSA at 50mg/kg body weight, or monensin at 10mg/kg, or both in combination for a period of 5 days was administered. Biomarkers indicative of oxidative stress like ROS, GSH, GSSG and TBARS demonstrated lead-induced toxic manifestations in blood, kidney and brain. Antioxidants like SOD, catalase and glutathione peroxidase along with specific lead biomarker, blood ALAD were also severely depleted in lead intoxicated animals. Serum parameters and histopathological findings supported the said results. MiADMSA treatment during both mono- and combination therapy with monensin, restored the antioxidant status and recovered biochemical and haematological variables due to lead. However, monensin alone was not found to be effective in the given scenario. Interestingly, combination therapy in its ability to revert lead-induced overall systemic toxicity was only found at par with the MiADMSA monotherapy except for its chelation potential. Monensin given in combination with MiADMSA potentiated its lead chelation ability especially from brain, along with maintaining the normal copper concentrations in the organ unlike MiADMSA monotherapy.
Collapse
|