1
|
Yi X, Wei R, Huang S, Wei P, Li H, Li Z, Aschner M, Jiang Y, Ou S, Li S. The effect of resveratrol on lead-induced oxidative damage and apoptosis in HT-22 cells. Food Chem Toxicol 2025; 197:115274. [PMID: 39864579 DOI: 10.1016/j.fct.2025.115274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVE The purpose of this work was to investigate whether resveratrol affects lead-induced oxidative damage in HT-22 cells, characterizing mechanisms and strategies for preventing and treating lead-induced neurotoxicity. METHODS Various lead and resveratrol concentrations were applied to HT-22 cells over different time periods. First, we established the lead treatment (12.5, 50 and 200 μmol/L) and resveratrol (40 μmol/L) intervention model for the study. MTT was used to analyze HT-22 cell survival rate. The rates of cell death, mitochondrial membrane potential, lipid peroxidation, and reactive oxygen species (ROS) generation were all measured by flow cytometry. Cellular oxidant (MDA) and antioxidant (SOD, GSH-Px) levels were measured with test kits. Western blotting was used to assess the expression of proteins related to autophagy and apoptosis. RESULTS Lead reduced HT-22 cell viability in a concentration/time-dependent manner. In addition, lead (200 μmol/L) decreased the protein expression of BCL2, while increasing PARP and BAX expression and apoptotic rate. Moreover, the lead-exposed group had significantly higher levels of ROS, lipid-ROS, and MDA than the control group. This was accompanied by increased MDA levels and decreased SOD, GSH-Px, and MMP levels in the lead-exposed cells. Furthermore, lead lowered SIRT1 protein expression, while increasing the levels of autophagy-related proteins, including P62, ATG5, Beclin-1 and LC3 Ⅱ/Ⅰ. Resveratrol (40 μmol/L), an agonist of SIRT1, restored the effects of lead (200 μmol/L) to levelsindistinguishable from controls. CONCLUSION Resveratrol inhibited mitochondrial damage and restored the lead-induced block of autophagic flux and oxidative stress by activating SIRT1, thereby alleviating lead-induced damage in HT-22 cells.
Collapse
Affiliation(s)
- Xiang Yi
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; School of Public Health, Xiangnan University, Chenzhou, 423000, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Ruokun Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaoni Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Peiqi Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - HuiShuai Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Zhenning Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shiyan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
2
|
Wang Y, Liu R, Li W, Bao P, Zhang J, Yue W. Global magnitude and temporal trends of stroke attributable to lead exposure from 1990 to 2021. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117865. [PMID: 39919592 DOI: 10.1016/j.ecoenv.2025.117865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/12/2025] [Accepted: 02/04/2025] [Indexed: 02/09/2025]
Abstract
OBJECTIVE Lead exposure is associated with an increased risk of stroke, but studies on the global burden of stroke attributable to lead exposure remain limited. This study evaluates the temporal and spatial trends in the disease burden of stroke and its three subtypes attributable to lead exposure in 204 countries and territories from 1990 to 2021. METHODS Data from Global Burden of Disease Study (GBD) 2021 were used to estimate annual deaths, disability-adjusted life years (DALYs), age-standardized mortality rate (ASMR), and age-standardized DALY rate (ASDR) of stroke attributable to lead exposure. The estimated annual percentage change (EAPC) was calculated to evaluate trends from 1990 to 2021. RESULTS Between 1990 and 2021, the global burden of stroke attributable to lead exposure increased significantly, with deaths rising from 341,294 to 556,595 and DALYs from 8.52 million to 12.02 million. However, ASMRs declined from 9.33 to 6.65 per 100,000 population (EAPC = -1.23), and ASDRs fell from 213.99 to 139.82 per 100,000 population (EAPC = -1.51). Among subtypes, ischemic stroke (IS) contributed the most to ASMRs, whereas intracerebral hemorrhage (ICH) accounted for the highest ASDRs. Males had higher ASMRs and ASDRs than females, although females exhibited greater reduction rates. The burden was disproportionately higher in older adults, particularly those over 70 years, with IS and ICH contributing significantly. Socio-demographic and regional disparities were evident, with low- and low-middle SDI regions bearing the highest burden. CONCLUSION The stroke burden attributable to lead exposure remains high, with significant variations by age, sex, region, and subtypes. Public health measures and policies are urgently needed to prevent and mitigate lead exposure globally.
Collapse
Affiliation(s)
- Ying Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Ran Liu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China; Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Wenxia Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - PengNan Bao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - JinWei Zhang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Wei Yue
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China; Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.
| |
Collapse
|
3
|
Rameshrad M, Memariani Z, Naraki K, Hosseinzadeh H. Investigating the protective properties of Panax ginseng and its constituents against biotoxins and metal toxicity: a mechanistic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1215-1242. [PMID: 39287674 DOI: 10.1007/s00210-024-03410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024]
Abstract
Natural toxins are toxic substances produced by living microorganisms and cause harmful effects to other creatures, but not the organisms themselves. Based on the sources, they are classified into fungal, microbial, herbal, algae, and animal biotoxins. Metals, the oldest toxicants, are not created or destroyed by human industry as elements, just concentrated in the biosphere. An antidote can counteract the toxic effects of a drug or toxin or mitigate the adverse effects of a harmful substance. The potential antidote effects of Panax ginseng in organ toxicity have been proved by many scientific research projects. Herein, we are going to gather a comprehensive mechanistic review of the antidotal effects of ginseng and its main constituents against natural toxins and metal toxicity. In this regard, a literate search has been done in PubMed/Medline, Science Direct, and Scopus from 2000 until 2024. The gathered data showed the protective impacts of this golden plant and its secondary metabolites against aflatoxin, deoxynivalenol, three-nitro propionic acid, ochratoxin A, lipopolysaccharide, nicotine, aconite, domoic acid, α-synuclein, amyloid β, and glutamate as well as aluminum, cadmium, chrome, copper, iron, and lead. These antidotal effects occur by multi-functional mechanisms. It may be attributed to antioxidant, anti-inflammatory, and anti-apoptotic effects. Future research directions on the antidotal effects of ginseng against natural toxins and metal toxicity involve broadening the scope of studies to include a wider range of toxins and metals, exploring synergistic interactions with other natural compounds, and conducting more human clinical trials to validate the efficacy and safety of ginseng-based treatments.
Collapse
Affiliation(s)
- Maryam Rameshrad
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Memariani
- Traditional Medicine and History of Medical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Karim Naraki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Science, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Jomova K, Alomar SY, Nepovimova E, Kuca K, Valko M. Heavy metals: toxicity and human health effects. Arch Toxicol 2025; 99:153-209. [PMID: 39567405 PMCID: PMC11742009 DOI: 10.1007/s00204-024-03903-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Heavy metals are naturally occurring components of the Earth's crust and persistent environmental pollutants. Human exposure to heavy metals occurs via various pathways, including inhalation of air/dust particles, ingesting contaminated water or soil, or through the food chain. Their bioaccumulation may lead to diverse toxic effects affecting different body tissues and organ systems. The toxicity of heavy metals depends on the properties of the given metal, dose, route, duration of exposure (acute or chronic), and extent of bioaccumulation. The detrimental impacts of heavy metals on human health are largely linked to their capacity to interfere with antioxidant defense mechanisms, primarily through their interaction with intracellular glutathione (GSH) or sulfhydryl groups (R-SH) of antioxidant enzymes such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and other enzyme systems. Although arsenic (As) is believed to bind directly to critical thiols, alternative hydrogen peroxide production processes have also been postulated. Heavy metals are known to interfere with signaling pathways and affect a variety of cellular processes, including cell growth, proliferation, survival, metabolism, and apoptosis. For example, cadmium can affect the BLC-2 family of proteins involved in mitochondrial death via the overexpression of antiapoptotic Bcl-2 and the suppression of proapoptotic (BAX, BAK) mechanisms, thus increasing the resistance of various cells to undergo malignant transformation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of antioxidant enzymes, the level of oxidative stress, and cellular resistance to oxidants and has been shown to act as a double-edged sword in response to arsenic-induced oxidative stress. Another mechanism of significant health threats and heavy metal (e.g., Pb) toxicity involves the substitution of essential metals (e.g., calcium (Ca), copper (Cu), and iron (Fe)) with structurally similar heavy metals (e.g., cadmium (Cd) and lead (Pb)) in the metal-binding sites of proteins. Displaced essential redox metals (copper, iron, manganese) from their natural metal-binding sites can catalyze the decomposition of hydrogen peroxide via the Fenton reaction and generate damaging ROS such as hydroxyl radicals, causing damage to lipids, proteins, and DNA. Conversely, some heavy metals, such as cadmium, can suppress the synthesis of nitric oxide radical (NO·), manifested by altered vasorelaxation and, consequently, blood pressure regulation. Pb-induced oxidative stress has been shown to be indirectly responsible for the depletion of nitric oxide due to its interaction with superoxide radical (O2·-), resulting in the formation of a potent biological oxidant, peroxynitrite (ONOO-). This review comprehensively discusses the mechanisms of heavy metal toxicity and their health effects. Aluminum (Al), cadmium (Cd), arsenic (As), mercury (Hg), lead (Pb), and chromium (Cr) and their roles in the development of gastrointestinal, pulmonary, kidney, reproductive, neurodegenerative (Alzheimer's and Parkinson's diseases), cardiovascular, and cancer (e.g. renal, lung, skin, stomach) diseases are discussed. A short account is devoted to the detoxification of heavy metals by chelation via the use of ethylenediaminetetraacetic acid (EDTA), dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), 2,3-dimercapto-1-propane sulfonic acid (DMPS), and penicillamine chelators.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, 949 74, Nitra, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
5
|
Luo Z, Zhu N, Xiong K, Qiu F, Cao C. Analysis of the relationship between sleep-related disorders and cadmium in the US population. Front Public Health 2024; 12:1476383. [PMID: 39525462 PMCID: PMC11544537 DOI: 10.3389/fpubh.2024.1476383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cadmium is a heavy metal that accumulates in the body due to environmental and occupational exposure. The neurotoxicity of cadmium received increasingly attention in recent years. Sleeping is regulated and coordinated by nervous system, however, little is known about the relationship between cadmium and sleep. This study aimed to examine the relationship between blood cadmium concentrations and sleep-related disorders in US adults. Methods This cross-sectional study used data on blood cadmium and sleep from the 2005-2008 and 2015-2020 National Health and Nutrition Examination Survey (NHANES). Weighted multiple regression, generalized weighted modeling, and weighted restricted cubic splines (RCS) were utilized to investigate the association between blood cadmium and sleep outcomes (sleep duration, trouble sleeping, symptoms of obstructive sleep apnea (OSA) and daytime sleepiness). Furthermore, subgroup analyses were conducted to investigate any differences in the associations between age, gender, ethnicity, education level, marital status, smoking status, alcohol consumption, diabetes mellitus (DM), cardiovascular disease (CVD) and hypertension groups. Results In 19,152 participants, the median blood cadmium concentration was 0.48 (IQR: 0.28, 0.82)μg/L. Compared with the lowest reference quartile, participants in the higher quartile had a significantly higher risk of insufficient sleeping (<7 h/night) in crude model (OR 1.53, 95% CI 1.33-1.74), Model 1 (OR 1.57, 95% CI 1.38-1.80) and Model 2 (OR 1.45, 95% CI 1.27-1.65). In the unadjusted model, individuals in the highest quartile of cadmium level had a significantly increased risk of OSA symptoms of 53% (OR = 1.53, 95% CI: 1.42, 1.65) compared with participants in the bottom quartile, and this risk increased by 35% (OR = 1.35, 95% CI: 1.23, 1.48) after adjusting for all covariates. Individuals in the highest quartile of cadmium level were 76% more likely to have a trouble sleeping than individuals in the lowest quartile in the unadjusted model (OR = 1.76, 95% CI: 1.31, 1.93), whereas in the fully adjusted model, this likelihood was 86% higher (OR = 1.86, 95% CI: 1.51, 1.96). A similar positive correlation was also observed for cadmium level and daytime sleepiness. However, no relationship was noted between cadmium and excessive sleep duration (≥9 h). A linear dose-response relationship was found between cadmium concentration and the risk of insufficient sleeping (P non-linearity = 0.321), OSA symptoms (P non-linearity = 0.176), trouble sleeping (P non-linearity = 0.682) and daytime sleepiness (P non-linearity = 0.565). Additionally, no significant interactions between cadmium concentrations and subgroup variables were identified (P for interaction>0.05). Conclusion Insufficient sleep, symptoms of OSA, trouble sleeping and daytime sleepiness were found to have a positive association with the blood cadmium concentration in US adults. However, further prospective studies are necessary to establish whether there is a causal relationship between these factors.
Collapse
Affiliation(s)
| | | | | | | | - Chao Cao
- Key Laboratory of Respiratory Disease of Ningbo, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Zhejiang, China
| |
Collapse
|
6
|
Alva S, Parithathvi A, Harshitha P, Dsouza HS. Influence of lead on cAMP-response element binding protein (CREB) and its implications in neurodegenerative disorders. Toxicol Lett 2024; 400:35-41. [PMID: 39117292 DOI: 10.1016/j.toxlet.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/03/2024] [Accepted: 08/06/2024] [Indexed: 08/10/2024]
Abstract
Lead (Pb2+) is one of the most common toxic metals present in the environment, and lead exposure causes serious health issues in humans. Lead is widely used because of its physio-chemical characteristics, which include softness, corrosion resistance, ductility, and low conductivity. Lead affects almost all human organs, specifically the central nervous system. Lead neurotoxicity is connected to various neural pathways, including brain-derived neurotrophic factor (BDNF) protein level alterations, cyclic adenosine 3',5'-monophosphate (cAMP) response element binding protein (CREB) pathway changes, and N-methyl-D-aspartate receptors (NMDARs) changes. Lead primarily affects protein kinase C (PKC) through the replacement of calcium (Ca2+) ions in the CREB pathway. In this review, we have discussed the effect of lead on the CREB pathway and its implications on the nervous system, highlighting its effects on learning, synaptic plasticity, memory, and cognitive deficits. This review provides an understanding of the lead-induced alterations in the CREB pathway, which can lead to the future prospect of its use as a diagnostic marker as well as a therapeutic target for neurodegenerative disorders.
Collapse
Affiliation(s)
- Sharal Alva
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| | - Aluru Parithathvi
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - P Harshitha
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Herman Sunil Dsouza
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
7
|
Huang M, Zhang H, Wang H, Niu J, Luo B, Wu G, Li X, Yan J. Effects of Cadmium and Lead Co-exposure on Sleep Status in Rural Areas Northwestern China. Biol Trace Elem Res 2024:10.1007/s12011-024-04243-z. [PMID: 38801624 DOI: 10.1007/s12011-024-04243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
In this study, we explored how cadmium and lead co-exposure affects sleep status among residents of a polluted area and nature reserve in rural northwestern China. Cadmium and lead levels were measured using blood samples, and sleep status was evaluated using sleep questionnaires, with the main sleep indicators including sleep duration, sleep quality, bedtime, and staying up. Furthermore, cadmium-lead co-exposure levels were divided into three groups: high exposure, medium exposure, and low exposure. Subjects in the contaminated area had significantly higher exposure levels (p < 0.001) and more negative sleep indicators (p < 0.01). Significant differences were found for all four sleep indicators in the high exposure group compared to the low exposure group (p < 0.01). Moreover, the overall evaluation of sleep status with high cadmium-lead co-exposure had a negative impact. Our data suggest that cadmium-lead co-exposure has a negative effect on sleep status and may have a synergistic effect on sleep.
Collapse
Affiliation(s)
- Min Huang
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430061, People's Republic of China
| | - Honglong Zhang
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Haiping Wang
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jingping Niu
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Bin Luo
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430061, People's Republic of China
| | - Xun Li
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, The First Hospital of Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jun Yan
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of General Surgery, The First Hospital of Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
8
|
Zahoor SM, Ishaq S, Ahmed T. Neurotoxic effects of metals on blood brain barrier impairment and possible therapeutic approaches. VITAMINS AND HORMONES 2024; 126:1-24. [PMID: 39029969 DOI: 10.1016/bs.vh.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Exposure to neurotoxic and heavy metals (Pb2+, As3+, Mn2+, Cd2+, etc) has increased over time and has shown to negatively affect brain health. Heavy metals can cross the blood brain barrier (BBB) in various ways including receptor or carrier-mediated transport, passive diffusion, or transport via gaps in the endothelial cells of the brain. In high concentrations, these metals have been shown to cause structural and functional impairment to the BBB, by inducing oxidative stress, ion dyshomeostasis, tight junction (TJ) loss, astrocyte/pericyte damage and interference of gap junctions. The structural and functional impairment of the BBB results in increased BBB permeability, which ultimately leads to accumulation of these heavy metals in the brain and their subsequent toxicity. As a result of these effects, heavy metals are correlated with various neurological disorders. The pathological effects of these heavy metals can be effectively mitigated via chelation. In addition, it is possible to treat the associated disorders by counteracting the molecular mechanisms associated with the brain and BBB impairment.
Collapse
Affiliation(s)
- Saba Mehak Zahoor
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sara Ishaq
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
9
|
You Y, Chen Y, Zhang Y, Zhang Q, Yu Y, Cao Q. Mitigation role of physical exercise participation in the relationship between blood cadmium and sleep disturbance: a cross-sectional study. BMC Public Health 2023; 23:1465. [PMID: 37525176 PMCID: PMC10391747 DOI: 10.1186/s12889-023-16358-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/20/2023] [Indexed: 08/02/2023] Open
Abstract
The neurotoxicity of heavy metals received increasingly attention in recent years. Sleeping is regulated and coordinated by nervous system, however, the health hazard of heavy metal like cadmium (Cd) exposure on sleep health remained unknown. Rescue strategies like physical exercise (PE) has emerged to mitigate such influence. An epidemiological design with cross-sectional data from National Health and Nutrition Examination Survey 2007-2010 was applied. The relationship between three blood heavy metals [cadmium (Cd), lead (Pb), mercury (Hg)] and sleep disturbance was analyzed. A total of 8,751 participants were finally included in and the weighted participants were 330,239,463. Weighted quantile sum (WQS) regression indicated that mixed blood metals were positively related to risk of sleep disturbance and the mixture effect of exposure to heavy metals was mainly attributable to Cd (89.1%). Weighted logistic regression showed a significant positive association between the highest quartile of blood Cd and sleep disturbance [(OR (95% CI)): 1.191 (1.014,1.400), p = 0.036] in the fully adjusted model, while no association was found under Pb and Hg exposure. In the association between Q3 and Q4 level of blood Cd and sleep disturbance, moderate-to-vigorous physical exercise group had lower risks than none and low exercise group. In the restricted cubic spline model, it was also verified that higher PE participation was associated with the lowest incidence of sleep disturbance with the increment in Cd concentration. Our study suggested that both policy makers and the public should minimize heavy metal exposure. Moreover, conducting moderate to vigorous physical exercise is a protecting factor to mitigate Cd's influence on sleep health.
Collapse
Affiliation(s)
- Yanwei You
- Division of Sports Science & Physical Education, Tsinghua University, Beijing, 100084, China
| | - Yuquan Chen
- Institute of Medical Information/Medical Library, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China
| | - Yangchang Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, 100169, China
| | - Qi Zhang
- Undergraduate Department, Taishan University, Taian, 250111, China
| | - Yaohui Yu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Qiang Cao
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, 650093, China.
- School of Pharmacy, Macau University of Science and Technology, Macau, Macau, 999078, China.
| |
Collapse
|
10
|
Zhang Z, Li J, Jiang S, Xu M, Ma T, Sun Z, Zhang J. Lactobacillus fermentum HNU312 alleviated oxidative damage and behavioural abnormalities during brain development in early life induced by chronic lead exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 251:114543. [PMID: 36640575 DOI: 10.1016/j.ecoenv.2023.114543] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
Lead exposure is a global public health safety issue that severely disrupts brain development and causes damage to the nervous system in early life. Probiotics and gut microbes have been highlighted for their critical roles in mitigating lead toxicity. However, the underlying mechanisms by which they work yet to be fully explored. Here, we designed a two-stage experiment using the probiotic Lactobacillus fermentum HNU312 (Lf312) to uncover how probiotics alleviate lead toxicity to the brain during early life. First, we explored the tolerance and adsorption of Lf312 to lead in vitro. Second, the adsorption capacity of the strain was determined and confirmed in vivo. The shotgun metagenome sequencing showed lead exposure-induced imbalance and dysfunction of the gut microbiome. In contrast, Lf312 intake significantly modulated the structure of the microbiome, increased the abundance of beneficial bacteria and short-chain fatty acids (SCFAs)-producing bacteria, and upregulated function-related metabolic pathways such as antioxidants. Notably, Lf312 enhanced the integrity of the blood-brain barrier by increasing the levels of SCFAs in the gut, alleviated inflammation in the brain, and ultimately improved anxiety-like and depression-like behaviours induced by lead exposure in mice. Subsequently, the effective mechanism was confirmed, highlighting that Lf312 worked through integrated strategies, including ionic adsorption and microbiota-gut-brain axis regulation. Collectively, this work elucidated the mechanism by which the gut microbiota mitigates the toxic effects of lead in the brain and provides preventive measures and intervention measures for brain damage due to mass lead poisoning in children.
Collapse
Affiliation(s)
- Zeng Zhang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Jiahe Li
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Shuaiming Jiang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Meng Xu
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Teng Ma
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education P. R. C., Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs China, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education P. R. C., Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs China, Inner Mongolia Agricultural University, Hohhot 010018, China.
| | - Jiachao Zhang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China; One Health Institute, Hainan University, Haikou, Hainan 570228, China.
| |
Collapse
|
11
|
Goel H, Goyal K, Pandey AK, Benjamin M, Khan F, Pandey P, Mittan S, Iqbal D, Alsaweed M, Alturaiki W, Madkhali Y, Kamal MA, Tanwar P, Upadhyay TK. Elucidations of Molecular Mechanism and Mechanistic Effects of Environmental Toxicants in Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:84-97. [PMID: 35352654 DOI: 10.2174/1871527321666220329103610] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 02/08/2023]
Abstract
Due to rising environmental and global public health concerns associated with environmental contamination, human populations are continually being exposed to environmental toxicants, including physical chemical mutagens widespread in our environment causing adverse consequences and inducing a variety of neurological disorders in humans. Physical mutagens comprise ionizing and non-ionizing radiation, such as UV rays, IR rays, X-rays, which produces a broad spectrum of neuronal destruction, including neuroinflammation, genetic instability, enhanced oxidative stress driving mitochondrial damage in the human neuronal antecedent cells, cognitive impairment due to alterations in neuronal function, especially in synaptic plasticity, neurogenesis repression, modifications in mature neuronal networks drives to enhanced neurodegenerative risk. Chemical Mutagens including alkylating agents (EMS, NM, MMS, and NTG), Hydroxylamine, nitrous acid, sodium azide, halouracils are the major toxic mutagen in our environment and have been associated with neurological disorders. These chemical mutagens create dimers of pyrimidine that cause DNA damage that leads to ROS generation producing mutations, chromosomal abnormalities, genotoxicity which leads to increased neurodegenerative risk. The toxicity of four heavy metal including Cd, As, Pb, Hg is mostly responsible for complicated neurological disorders in humans. Cadmium exposure can enhance the permeability of the BBB and penetrate the brain, driving brain intracellular accumulation, cellular dysfunction, and cerebral edema. Arsenic exerts its toxic effect by induction of ROS production in neuronal cells. In this review, we summarize the molecular mechanism and mechanistic effects of mutagens in the environment and their role in multiple neurological disorders.
Collapse
Affiliation(s)
- Harsh Goel
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Keshav Goyal
- Division of Molecular and Cellular Biology, Faculty of Biology, Ludwig Maximilians Universitat, Munchen, Germany
| | - Avanish Kumar Pandey
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Mercilena Benjamin
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering & Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, India
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering & Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, India
| | - Sandeep Mittan
- Department of Cardiology, Ichan School of Medicine, Mount Sinai Hospital, One Gustave L. Levy Place, New York, USA
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | - Mohammed Alsaweed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | - Yahya Madkhali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, China
- King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Bangladesh
- Enzymoics, 7 Peterlee Place, Hebersham NSW 2770, Novel Global Community Educational Foundation, Australia
| | - Pranay Tanwar
- Department of Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, Gujarat 391760, India
| |
Collapse
|
12
|
Liu M, Liu R, Wang R, Ba Y, Yu F, Deng Q, Huang H. Lead-induced neurodevelopmental lesion and epigenetic landscape: Implication in neurological disorders. J Appl Toxicol 2022. [PMID: 36433892 DOI: 10.1002/jat.4419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/20/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Lead (Pb) was implicated in multiple genotoxic, neuroepigenotoxic, and chromosomal-toxic mechanisms and interacted with varying synaptic plasticity pathways, likely underpinning previous reports of links between Pb and cognitive impairment. Epigenetic changes have emerged as a promising biomarker for neurological disorders, including cognitive disorders, Alzheimer's disease (AD), and Parkinson's disease (PD). In the present review, special attention is paid to neural epigenetic features and mechanisms that can alter gene expression patterns upon environmental Pb exposure in rodents, primates, and zebrafish. Epigenetic modifications have also been discussed in population studies and cell experiment. Further, we explore growing evidence of potential linkage between Pb-induced disruption of regulatory pathway and neurodevelopmental and neurological disorders both in vivo and in vitro. These findings uncover how epigenome in neurons facilitates the development and function of the brain in response to Pb insult.
Collapse
Affiliation(s)
- Mengchen Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Rundong Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Ruike Wang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Yue Ba
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Fangfang Yu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Qihong Deng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Hui Huang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| |
Collapse
|
13
|
Guan R, Wang T, Dong X, Du K, Li J, Zhao F, Xu J, Li B, Zheng G, Shen X, Cao B, Wang J, Aschner M, Liu M, Chen R. Effects of co-exposure to lead and manganese on learning and memory deficits. J Environ Sci (China) 2022; 121:65-76. [PMID: 35654517 PMCID: PMC9163452 DOI: 10.1016/j.jes.2021.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 06/15/2023]
Abstract
Lead (Pb) and manganese (Mn) are common neurotoxins. However, individuals are subject to co-exposures in real life, and it is therefore important to study these metals in combination. Weaning Sprague-Dawley rats were given ad libitum access to drinking water solutions containing Pb (100 mg/L), Mn (2.5 mg/mL) or a mixture, and each treatment had its own minocycline (50 mg/(kg•day)) supplement group. The results showed a significant difference in spatial memory and induction levels of hippocampal long-term potentiation (LTP) in all exposure groups when compared with controls. The combined-exposure group exhibited the most pronounced effect when compared with each of the single-metal exposure groups. Microglia displayed activation at day 3 after exposure alone or in combination, while astrocytes showed activation at day 5, accompanied by decreased expression levels of GLAST, GLT-1, and GS. Furthermore, the levels of glutamate in the synaptic cleft increased significantly. When microglial activation was inhibited by minocycline, the activation of astrocytes and the expression of GLAST, GLT-1, and GS were both reversed. In addition, upon minocycline treatment, hippocampal LTP impairment and cognitive injury were significantly alleviated in each of the exposure groups. These results suggest that combined exposure to Pb and Mn can cause greater effects on cognition and synaptic plasticity when compared to single-metal exposure groups. The reason may involve abnormal activation of microglia leading to excessive regulation of astrocytes, resulting in glutamate reuptake dysfunction in astrocytes and leading to perturbed cognition and synaptic plasticity.
Collapse
Affiliation(s)
- Ruili Guan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Tao Wang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoru Dong
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Kejun Du
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Juan Li
- Department of Physiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Fang Zhao
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Jie Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Bin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Gang Zheng
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Xuefeng Shen
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Baohua Cao
- School of Nursing, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Wang
- School of Nursing, Fourth Military Medical University, Xi'an 710032, China
| | - Michael Aschner
- School of Nursing, Fourth Military Medical University, Xi'an 710032, China; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mingchao Liu
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China; School of Nursing, Fourth Military Medical University, Xi'an 710032, China.
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
14
|
Chelyadina NS, Kapranov SV, Popov MA, Smirnova LL, Bobko NI. Trace elements in the detoxifying and accumulating body parts of Mytilus galloprovincialis Lamark, 1819 (Crimea, Black Sea): human health risks and effect of the sampling site location. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:61352-61369. [PMID: 35441295 DOI: 10.1007/s11356-022-20186-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/06/2022] [Indexed: 06/14/2023]
Abstract
The mussel M. galloprovincialis is one of the most popular species in world's mariculture and environment pollution bioindicators. Although this mollusk was in a focus of numerous publications, the detoxifying and accumulating role of some of its body parts was insufficiently studied. The goals of the present work were as follows: (a) to study the distribution of potentially toxic elements (Cr, Co, Ni, Cu, Zn, As, Cd, Hg, Pb) in soft tissues, byssus, and shell liquor of this bivalve as a function of sampling location in the Black Sea near the southwestern coast of Crimea and (b) to assess human health risks from consuming soft tissues of mussels cultivated on a mollusk farm. Multivariate analysis showed significant differences in the overall distribution of the elements among the body parts and sampling sites under consideration. The trace element contents in soft tissues of M. galloprovincialis decreased in the following order: Zn > Cu > As > Ni > Pb > Cd > Cr > Co > Hg. The noncarcinogenic hazard index from the cultivated mussel consumption was found to be well below one and the carcinogenic risk index was found within the tolerable limits, which indicate the safety of consuming these mussels for humans. Byssus of M. galloprovincialis was characterized as a perfect indicator of marine environment pollution with Ni, Cu, Pb, Сo, and Cr. For the first time, the concentrations of trace elements were determined in the shell liquor and the function of byssus and shell liquor as the systems of trace element excretion from soft tissues was demonstrated.
Collapse
Affiliation(s)
- Natalya S Chelyadina
- A.O. Kovalevsky Institute of Biology of the Southern Seas, Russian Academy of Sciences, Nakhimov ave. 2, Sevastopol, Russian Federation, 299011.
| | - Sergey V Kapranov
- A.O. Kovalevsky Institute of Biology of the Southern Seas, Russian Academy of Sciences, Nakhimov ave. 2, Sevastopol, Russian Federation, 299011
| | - Mark A Popov
- A.O. Kovalevsky Institute of Biology of the Southern Seas, Russian Academy of Sciences, Nakhimov ave. 2, Sevastopol, Russian Federation, 299011
| | - Lyudmila L Smirnova
- Institute of Natural and Technical Systems, Russian Academy of Sciences, Lenin str. 28, Sevastopol, Russian Federation, 299011
| | - Nikolay I Bobko
- A.O. Kovalevsky Institute of Biology of the Southern Seas, Russian Academy of Sciences, Nakhimov ave. 2, Sevastopol, Russian Federation, 299011
| |
Collapse
|
15
|
Zhang T, Yin X, Zhang Y, Chen H, Man J, Li Y, Chen J, Yang X, Lu M. Global Trends in Mortality and Burden of Stroke Attributable to Lead Exposure From 1990 to 2019. Front Cardiovasc Med 2022; 9:870747. [PMID: 35811690 PMCID: PMC9259800 DOI: 10.3389/fcvm.2022.870747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background Lead exposure is an important risk factor for stroke. However, the latest global spatiotemporal patterns of lead exposure-related stroke burden were unclear. In this study, we assessed this topic. Methods The data were obtained from the Global Burden of Disease Study (2019). The estimated annual percentage change (EAPC) was estimated to evaluate the temporal trends of the age-standardized mortality and disability-adjusted life years (DALYs) rates (ASMR and ASDR) of stroke attributable to lead exposure. Results In 2019, the numbers of global stroke deaths and DALYs attributable to lead exposure were 305.27 and 6738.78 thousand, respectively. The corresponding ASMR and ASDR were highest in males, the elderly population, low and middle-income countries, and the intracerebral hemorrhage subtype. From 1990 to 2019, the ASMR and ASDR of global stroke attributable to lead exposure decreased [ASMR: EAPC = −1.34, 95% confidence interval (CI): (−1.57, −1.10); ASDR: EAPC = −1.74, 95% CI: (−1.95, −1.52)], especially in females, the high-income countries, and the subarachnoid hemorrhage subtype. Conclusion This study emphasizes the importance of continued implementation of lead exposure prevention strategies and improved high-efficiency treatment and stroke acute health care, especially in low and middle-income countries.
Collapse
Affiliation(s)
- Tongchao Zhang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
| | - Xiaolin Yin
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuan Zhang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
| | - Hui Chen
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
| | - Jinyu Man
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yufei Li
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiaqi Chen
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaorong Yang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
- *Correspondence: Xiaorong Yang,
| | - Ming Lu
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center of Shandong University, Jinan, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Ming Lu,
| |
Collapse
|
16
|
Khedr NF, Talkan OFA. New insights into arsenic, lead, and iron neurotoxicity: Activation of MAPK signaling pathway and oxidative stress. J Biochem Mol Toxicol 2022; 36:e23040. [DOI: 10.1002/jbt.23040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/27/2022] [Accepted: 03/02/2022] [Indexed: 01/14/2023]
Affiliation(s)
- Naglaa F. Khedr
- Department of Biochemistry, Faculty of Pharmacy Tanta University Tanta Egypt
| | - Ola F. A. Talkan
- Chemistry Department, Animal Health Research Institute‐Shiben El‐Kom Lab. Agriculture Research Center Menofia Shiben El‐Kom Egypt
| |
Collapse
|
17
|
Liu J, Ghastine L, Um P, Rovit E, Wu T. Environmental exposures and sleep outcomes: A review of evidence, potential mechanisms, and implications. ENVIRONMENTAL RESEARCH 2021; 196:110406. [PMID: 33130170 PMCID: PMC8081760 DOI: 10.1016/j.envres.2020.110406] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 06/02/2023]
Abstract
Environmental exposures and poor sleep outcomes are known to have consequential effects on human health. This integrative review first seeks to present and synthesize existing literature investigating the relationship between exposure to various environmental factors and sleep health. We then present potential mechanisms of action as well as implications for policy and future research for each environmental exposure. Broadly, although studies are still emerging, empirical evidence has begun to show a positive association between adverse effects of heavy metal, noise pollution, light pollution, second-hand smoke, and air pollution exposures and various sleep problems. Specifically, these negative sleep outcomes range from subjective sleep manifestations, such as general sleep quality, sleep duration, daytime dysfunction, and daytime sleepiness, as well as objective sleep measures, including difficulties with sleep onset and maintenance, sleep stage or circadian rhythm interference, sleep arousal, REM activity, and sleep disordered breathing. However, the association between light exposure and sleep is less clear. Potential toxicological mechanisms are thought to include the direct effect of various environmental toxicants on the nervous, respiratory, and cardiovascular systems, oxidative stress, and inflammation. Nevertheless, future research is required to tease out the exact pathways of action to explain the associations between each environmental factor and sleep, to inform possible therapies to negate the detrimental effects, and to increase efforts in decreasing exposure to these harmful environmental factors to improve health.
Collapse
Affiliation(s)
- Jianghong Liu
- University of Pennsylvania School of Nursing, 418 Curie Blvd, Philadelphia, PA, 19104, USA.
| | - Lea Ghastine
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Phoebe Um
- Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Elizabeth Rovit
- University of Pennsylvania School of Nursing, 418 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Tina Wu
- University of Pennsylvania School of Nursing, 418 Curie Blvd, Philadelphia, PA, 19104, USA
| |
Collapse
|
18
|
Zeng X, Xu C, Xu X, Zhang Y, Huang Y, Huo X. Elevated lead levels in relation to low serum neuropeptide Y and adverse behavioral effects in preschool children with e-waste exposure. CHEMOSPHERE 2021; 269:129380. [PMID: 33383249 DOI: 10.1016/j.chemosphere.2020.129380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/05/2020] [Accepted: 12/17/2020] [Indexed: 02/05/2023]
Abstract
As a neurotoxicant, lead (Pb) primarily affects central nervous system, and particularly impacts developing brain. This study explores the associations of blood Pb level and children's behavioral health. A total of 213 preschool children aged 3-7 years old were recruited from Guiyu (the e-waste-exposed area) and Haojiang (the reference area). The behavioral health of children was assessed using the 'behavioral symptoms' subscale of the Strengths and Difficulties Questionnaire (SDQ). Results showed that there was a significant difference in percent of children categorized as "at risk" between Guiyu (48.2%) and Haojiang (13.9%) (p < 0.001). The blood Pb level of children in Guiyu was significantly higher than those in Haojiang (median: 5.19 μg/dL vs. 3.42 μg/dL, p < 0.001). The serum Neuropeptide Y (NPY) was significantly lower in Guiyu children than those in Haojiang. Spearman correlation analyses demonstrated that blood Pb levels was negatively correlated with NPY (rs = -0.25, p < 0.001), but positively correlated with behavioral symptom scores; while serum NPY levels were negatively associated with behavioral symptom scores. Behavioral symptom scores were higher in children with blood Pb level ≥5.00 μg/dL (high) than those with blood Pb level < 5.00 μg/dL (low). After adjusting for confounding factors, children with lower NPY levels were at higher risk of having behavioral difficulties. In conclusion, Pb exposure in e-waste-exposed areas may lead to decrease in serum NPY and increase in the risk of children's behavioral problems. In addition, NPY may mediate the association between Pb exposure and behavioral difficulties.
Collapse
Affiliation(s)
- Xiang Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Cheng Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, And Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, And Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yu Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, And Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, 9713, GZ, the Netherlands
| | - Yu Huang
- Laboratory of Environmental Medicine and Developmental Toxicology, And Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
19
|
Abstract
Lead (Pb2+) is a non-essential metal with numerous industrial applications that have led to ts ubiquity in the environment. Thus, not only occupational-exposed individuals' health is compromised, but also that of the general population and in particular children. Notably, although the central nervous system is particularly susceptible to Pb2+, other systems are affected as well. The present study focuses on molecular mechanisms that underlie the effects that arise from the presence of Pb2+ in situ in the brain, and the possible toxic effects that follows. As the brain barriers represent the first target of systemic Pb2+, mechanisms of Pb2+ entry into the brain are discussed, followed by a detailed discussion on neurotoxic mechanisms, with special emphasis on theories of ion mimicry, mitochondrial dysfunction, redox imbalance, and neuroinflammation. Most importantly, the confluence and crosstalk between these events is combined into a cogent mechanism of toxicity, by intertwining recent and old evidences from humans, in vitro cell culture and experimental animals. Finally, pharmacological interventions, including chelators, antioxidants substances, anti-inflammatory drugs, or their combination are reviewed as integrated approaches to ameliorate Pb2+ harmful effects in both developing or adult organisms.
Collapse
Affiliation(s)
- Miriam B. Virgolini
- IFEC CONICET. IFEC-CONICET. Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Haya de la Torre y Medina Allende, Ciudad Universitaria, 5016, Córdoba, Argentina
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA and IM Sechenov First Moscow State Medical University (Sechenov University), 119146, Moscow, Russia
| |
Collapse
|
20
|
Latham LE, Wang C, Patterson TA, Slikker W, Liu F. Neuroprotective Effects of Carnitine and Its Potential Application to Ameliorate Neurotoxicity. Chem Res Toxicol 2021; 34:1208-1222. [PMID: 33570912 DOI: 10.1021/acs.chemrestox.0c00479] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Carnitine is an essential metabolite that is absorbed from the diet and synthesized in the kidney, liver, and brain. It ferries fatty acids across the mitochondrial membrane to undergo β-oxidation. Carnitine has been studied as a therapy or protective agent for many neurological diseases and neurotoxicity (e.g., prolonged anesthetic exposure-induced developmental neurotoxicity in preclinical models). Preclinical and clinical data support the notion that carnitine or acetyl carnitine may improve a patient's quality of life through increased mitochondrial respiration, release of neurotransmitters, and global gene expression changes, showing the potential of carnitine beyond its approved use to treat primary and secondary carnitine deficiency. In this review, we summarize the beneficial effects of carnitine or acetyl carnitine on the central nervous system, highlighting protective effects against neurotoxicity-induced damage caused by various chemicals and encouraging a thorough evaluation of carnitine use as a therapy for patients suffering from neurotoxicant exposure.
Collapse
Affiliation(s)
- Leah E Latham
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas 72079, United States
| | - Cheng Wang
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas 72079, United States
| | - Tucker A Patterson
- Office of Director, National Center for Toxicological Research/FDA, Jefferson, Arkansas 72079, United States
| | - William Slikker
- Office of Director, National Center for Toxicological Research/FDA, Jefferson, Arkansas 72079, United States
| | - Fang Liu
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas 72079, United States
| |
Collapse
|
21
|
Zhang Y, Zhang P, Yu P, Shang X, Lu Y, Li Y. Transcriptome analysis reveals the mechanism of common carp brain injury after exposure to lead. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 743:140796. [PMID: 32673925 DOI: 10.1016/j.scitotenv.2020.140796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/29/2020] [Accepted: 07/05/2020] [Indexed: 06/11/2023]
Abstract
Lead, a widespread industrial pollutant, has been known as a powerful neurotoxin that could affect the central nervous system. Accumulating evidences demonstrated that lead exposure could result in the damage of brain tissues both in fish and human. However, the mechanism of lead induced brain injury has not been fully elucidated. The purpose of this study was to clarify the possible mechanism of common carp brain injury after exposure to lead through transcriptome analysis. Transcriptome analysis showed that 2141 differentially expressed genes were identified. Among these, 502 genes were up-regulated and 1639 genes were down-regulated. Meanwhile, GO enrichment analysis showed Transport, biological_process, DNA-templated (regulation of transcription) and signal transduction contained the most differential genes in the biological process. Furthermore, KEGG pathway enrichment analysis showed Ion channels, GnRH signaling pathway, cell adhesion molecules, Wnt signaling pathway, and calcium signaling pathway were significantly enriched. In addition, 10 differentially expressed genes were selected for qRT-PCR detection, and the results demonstrated that the selected genes exhibited the same trends with the RNA-Seq results, which indicates the transcriptome sequencing data is reliable. In conclusion, the above results provide a theoretical basis for clarifying the relationship between lead exposure and brain injury in common carp and for further studying of the genes related to lead poisoning.
Collapse
Affiliation(s)
- Yue Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Peijun Zhang
- Health Monitoring and Inspection Center of Jilin Province, Changchun 130062, China
| | - Peng Yu
- College of Electronic and Information Engineering, Changchun University of Science and Technology, Changchun 130022, China
| | - Xinchi Shang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Yuting Lu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Yuehong Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
22
|
Rangel-Barajas C, Coronel I, Zhang Y, Hernández M, Boehm Ii SL. Low-level developmental lead exposure does not predispose to adult alcohol self-administration, but does increase the risk of relapsing to alcohol seeking in mice: Contrasting role of GLT1 and xCT brain expression. Neuropharmacology 2020; 181:108339. [PMID: 33010299 DOI: 10.1016/j.neuropharm.2020.108339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/27/2022]
Abstract
Lead (Pb) is a neurotoxic heavy metal pollutant. Despite the efforts to reduce Pb environmental exposure and to prevent Pb poisoning, exposure in human populations persists. Studies of adults with history of childhood lead exposure have consistently demonstrated cognitive impairments that have been associated with sustained glutamate signaling. Additionally, some clinical studies have also found correlations between Pb exposure and increased proclivity to drug addiction. Thus, here we sought to investigate if developmental Pb exposure can increase propensity to alcohol consumption and relapse using an alcohol self-administration paradigm. Because Pb exposure is associated with increased glutamatergic tone, we also studied the effects on the expression of synaptic and non-synaptic glutamate transporters in brain regions associated with drug addiction such as the nucleus accumbens (NAc), dorsomedial striatum (DMS), dorsolateral striatum (DLS), and medial prefrontal cortex (mPFC). We found that while developmental Pb exposure did not increase risk for alcohol self-administration, it did play a role in relapsing to alcohol. The effects were associated with differential expression of the glutamate transporter 1 (GLT1) and the glutamate/cystine antiporter (xCT). In the NAc and DLS the expression of GLT1 was found to be significantly reduced, while no changes were found in DMS or mPFC. Contrastingly, xCT was found to be upregulated in NAc but downregulated in DLS, with no changes in DMS or mPFC. Our data suggest that lead exposure is involved in relapse to alcohol seeking, an effect that could be associated with downregulation of GLT1 and xCT in the DLS.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Israel Coronel
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Yanping Zhang
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Maribel Hernández
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Stephen L Boehm Ii
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA; Indiana Alcohol Research Center, Indiana University School of Medicine, 340 W 10th St, Indianapolis, IN, 462020, USA.
| |
Collapse
|
23
|
Kim M, Yun SM, Jeong J, Jo C, Koh YH. Association between blood lead level and risk of stroke in Korean adults: a cross-sectional study in the Korea National Health and Nutrition Examination Survey 2008-2013. BMJ Open 2020; 10:e035725. [PMID: 32907895 PMCID: PMC7482501 DOI: 10.1136/bmjopen-2019-035725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Although lead is a potential risk factor for cardiovascular diseases such as stroke, research on this association in the Korean population remains limited. Therefore, we aimed to investigate the association between lead level and stroke in Korean adults. DESIGN A population-based cross-sectional study. SETTING The Korea National Health and Nutrition Examination Survey 2008-2013, which enrolled a representative sample of the Korean population. PARTICIPANTS We excluded participants younger than 20 years, missing weight data, pregnant or lactating, and missing blood lead and stroke data. A total of 11 510 participants were included in this analysis. PRIMARY AND SECONDARY OUTCOME MEASUREMENT The participants were classified by blood lead concentration into the low-level (≤2.189 µg/dL, n=5756) and high-level (>2.189 µg/dL, n=5754) groups. The main outcome, stroke, was assessed by information from physician diagnosis, prevalence of stroke or treatment for stroke. The ORs and 95% CIs were calculated to evaluate the association between blood lead level and stroke using multivariate logistic regression analysis. RESULTS Although blood lead level was not significantly associated with stroke (OR: 1.30, 95% CI: 0.66-2.58) in the multivariate-adjusted model, in individuals with hypertension, the high-level group was 2.36-fold higher odds of stroke (OR: 2.36, 95% CI: 1.02-5.44) compared to that in the low-level group. No association was observed in individuals with normotension (OR: 0.42, 95% CI: 0.13-1.38, p for interaction=0.007). CONCLUSION The association between blood lead concentration and stroke may be influenced by hypertension status. Our findings suggest the need for closer attention to lead exposure in patients with hypertension.
Collapse
Affiliation(s)
- Minkyeong Kim
- Division of Brain Diseases, Korea National Institute of Health, Cheongju, South Korea
| | - Sang-Moon Yun
- Division of Brain Diseases, Korea National Institute of Health, Cheongju, South Korea
| | - Jihyun Jeong
- Division of Brain Diseases, Korea National Institute of Health, Cheongju, South Korea
| | - Chulman Jo
- Division of Brain Diseases, Korea National Institute of Health, Cheongju, South Korea
| | - Young Ho Koh
- Division of Brain Diseases, Korea National Institute of Health, Cheongju, South Korea
| |
Collapse
|
24
|
Zhou F, Du G, Xie J, Gu J, Jia Q, Fan Y, Yu H, Zha Z, Wang K, Ouyang L, Shao L, Feng C, Fan G. RyRs mediate lead-induced neurodegenerative disorders through calcium signaling pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 701:134901. [PMID: 31710906 DOI: 10.1016/j.scitotenv.2019.134901] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/21/2019] [Accepted: 10/07/2019] [Indexed: 06/10/2023]
Abstract
Heavy metal lead (Pb) is widely distributed in the environment and can induce neurodegeneration. Accumulating evidence has shown that ryanodine receptors (RyRs) play vital roles in neurodegenerative brain. However, whether aberrant RyRs levels contribute to Pb-induced neurodegeneration has largely remained unknown. In the present study, we report the important role of elevated levels of RyRs in Pb-induced neurodegeneration. Pb was found to upregulate the levels of RyRs in the rat hippocampal tissues and rat pheochromocytoma (PC12) cells. Furthermore, exposure to Pb induced neurodegenerative cognitive impairment in rats, depressed the long-term potentiation (LTP) in the rat brain slices, increased the neuronal intracellular free calcium concentration ([Ca2+]i), inhibited the phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and cyclic adenosine 3',5'-monophosphate (cAMP) response element binding protein (CREB) as well as the expression of anti-apoptotic protein B-cell lymphoma 2 (Bcl2), and activated the phosphorylation of extracellular regulated protein kinases (Erk) protein both in vitro and in vivo. In addition, the knockdown of RyR3 in PC12 cells significantly decreased the [Ca2+]i levels, increased the CaMKIIα and CREB phosphorylation, decrease the phosphorylation of Erk, and elongated the cognitive function-related neurite outgrowth after exposure to Pb. Moreover, treatment with a RyRs agonist showed the involvement of RyRs in Pb-induced depression in LTP in the rat brain slices. In summary, we determined that Pb-mediated upregulation of RyRs led to neurodegeneration via high levels of free calcium, depression of the calcium-dependent CaMKIIα/CREB mnemonic signaling pathway, and activation of the calcium-dependent Erk/Bcl2 apoptotic signaling pathway. These findings on the impact of Pb on the levels of RyRs could further improve our understanding of Pb-induced neurotoxicity and provide a promising molecular target to antagonize Pb-induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Fankun Zhou
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Guihua Du
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Jie Xie
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Junwang Gu
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Qiyue Jia
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Ying Fan
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Han Yu
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Zhipeng Zha
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Kai Wang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Lu Ouyang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Lijian Shao
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Chang Feng
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Guangqin Fan
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
25
|
Goudarzi N, Mohammad Valipour S, Nooritahneh A, Motaghinejad M, Motevalian M, Safari S, Gholami M, Vatandour S, Hekmati M. Pharmacological Evidences for Curcumin Neuroprotective Effects against Lead-Induced Neurodegeneration: Possible Role of Akt/GSK3 Signaling Pathway. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:494-508. [PMID: 33680047 PMCID: PMC7758019 DOI: 10.22037/ijpr.2020.1101210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
One of main herbal compounds with neuroprotective effects is curcumin. Lead poisoning cause neurodegeneration effect but its clear mechanism remains unknown. The current study evaluates the role of Akt/GSK3 signaling pathway in mediating the neuroprotective effects of curcumin against lead -induced neurodegeneration in rats. Sixty adult male rats were divided to: Group 1 and 2 receiving normal saline and drinking water containing 0.075% of lead acetate. Groups 3, 4, 5, and 6 were treated concurrently with lead acetate (0.075% in drinking water) and Curcumin (10, 20, 40, and 80 mg/kg I.P, respectively). Morris water maze (MWM) was used to evaluate cognitive activity, Hippocampal oxidative, anti-oxidant, as well as inflammatory and apoptotic factors and also Akt and GSK3 protein levels were studied. We found that lead poisoning disturbed the learning and memory and simultaneous treatment with Curcumin reduced the lead -induced cognition disturbances. In addition, lead acetate treatment increased lipid peroxidation and the levels of IL-1β, TNF-α , Bax, GSK3 (total and phosphorylated) while reducing reduced form of GSH, Bcl-2, and Akt3 (total and phosphorylated) levels in the hippocampus. Lead also reduced the activity of SOD, GPx, and GR in the hippocampus. In contrast, various doses of Curcumin attenuated lead -induced apoptosis, oxidative stress and inflammation; while elevating P-Akt and reduced of GSK3 levels. Thus, Curcumin via mediation of Akt/GSK3 signaling pathway confers neuroprotection against lead-induced neurodegeneration in hippocampus.
Collapse
Affiliation(s)
- Negin Goudarzi
- Department of Veterinary and Biomedical Science University of Minnesota, USA.
| | - Sanaz Mohammad Valipour
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Akram Nooritahneh
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Majid Motaghinejad
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Manijeh Motevalian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mina Gholami
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Safieh Vatandour
- Department of Animal science, Qaemshahr Branch, Islamic Azad University, Mazandaran, Iran.
| | - Malak Hekmati
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
26
|
Çelik H, Kucukler S, Çomaklı S, Özdemir S, Caglayan C, Yardım A, Kandemir FM. Morin attenuates ifosfamide-induced neurotoxicity in rats via suppression of oxidative stress, neuroinflammation and neuronal apoptosis. Neurotoxicology 2019; 76:126-137. [PMID: 31722249 DOI: 10.1016/j.neuro.2019.11.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/06/2019] [Accepted: 11/09/2019] [Indexed: 01/10/2023]
Abstract
Ifosfamide (IFA), a commonly used chemotherapeutic drug, has been frequently associated with encephalopathy and central nervous system toxicity. The present study aims to investigate whether morin could protect against acute IFA-induced neurotoxicity. Morin was administered to male rats once daily for 2 consecutive days at doses of 100 and 200 mg/kg body weight (BW) orally. IFA (500 mg/kg BW; i.p.) was administered on second day. The results showed that morin markedly inhibited the production of acetylcholinesterase (AChE), butrylcholinesterase (BChE), carbonic anhydrase (CA), glial fibrillary acidic protein (GFAP), brain-derived neurotrophic factor (BDNF) and nuclear factor erythroid 2-related factor 2 (Nrf-2) induced by IFA. Morin ameliorated IFA-induced lipid peroxidation, glutathione (GSH) depletion, and decrease antioxidant enzyme activities, catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Histopathological changes and immunohistochemical expressions of c-Jun N-terminal kinase (JNK) and c-Fos in the IFA-induced brain tissues were decreased after administration of morin. Furthermore, morin was able to down regulate the levels of inflammatory and apoptotic markers such as nuclear factor kappa B (NF-κB), neuronal nitric oxide synthase (nNOS), tumor necrosis factor-α (TNF-α), p53, cysteine aspartate specific protease-3 (caspase-3) and B-cell lymphoma-2 (Bcl-2). Taken together, our results demonstrated that morin elicited a typical chemoprotective effect on IFA-induced acute neurotoxicity.
Collapse
Affiliation(s)
- Hamit Çelik
- Department of Neurology, Private Buhara Hospital, Erzurum, Turkey
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Cuneyt Caglayan
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingol University, Bingol,Turkey.
| | - Ahmet Yardım
- Department of Neurosurgery, Private Buhara Hospital, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
27
|
Zhu G, Dai B, Chen Z, He L, Guo J, Dan Y, Liang S, Li G. Effects of chronic lead exposure on the sympathoexcitatory response associated with the P2X7 receptor in rat superior cervical ganglia. Auton Neurosci 2019; 219:33-41. [DOI: 10.1016/j.autneu.2019.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/01/2019] [Accepted: 03/20/2019] [Indexed: 12/23/2022]
|
28
|
Garza-Lombó C, Posadas Y, Quintanar L, Gonsebatt ME, Franco R. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid Redox Signal 2018; 28:1669-1703. [PMID: 29402131 PMCID: PMC5962337 DOI: 10.1089/ars.2017.7272] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Essential metals such as copper, iron, manganese, and zinc play a role as cofactors in the activity of a wide range of processes involved in cellular homeostasis and survival, as well as during organ and tissue development. Throughout our life span, humans are also exposed to xenobiotic metals from natural and anthropogenic sources, including aluminum, arsenic, cadmium, lead, and mercury. It is well recognized that alterations in the homeostasis of essential metals and an increased environmental/occupational exposure to xenobiotic metals are linked to several neurological disorders, including neurodegeneration and neurodevelopmental alterations. Recent Advances: The redox activity of essential metals is key for neuronal homeostasis and brain function. Alterations in redox homeostasis and signaling are central to the pathological consequences of dysfunctional metal ion homeostasis and increased exposure to xenobiotic metals. Both redox-active and redox-inactive metals trigger oxidative stress and damage in the central nervous system, and the exact mechanisms involved are starting to become delineated. CRITICAL ISSUES In this review, we aim to appraise the role of essential metals in determining the redox balance in the brain and the mechanisms by which alterations in the homeostasis of essential metals and exposure to xenobiotic metals disturb the cellular redox balance and signaling. We focus on recent literature regarding their transport, metabolism, and mechanisms of toxicity in neural systems. FUTURE DIRECTIONS Delineating the specific mechanisms by which metals alter redox homeostasis is key to understand the pathological processes that convey chronic neuronal dysfunction in neurodegenerative and neurodevelopmental disorders. Antioxid. Redox Signal. 28, 1669-1703.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska.,2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Yanahi Posadas
- 3 Departamentos de Farmacología y de, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México .,4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - Liliana Quintanar
- 4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - María E Gonsebatt
- 2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Rodrigo Franco
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska
| |
Collapse
|
29
|
Soussi A, Gargouri M, El Feki A. Effects of co-exposure to lead and zinc on redox status, kidney variables, and histopathology in adult albino rats. Toxicol Ind Health 2018; 34:469-480. [PMID: 29702030 DOI: 10.1177/0748233718770293] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Lead (Pb) is a toxic metal that induces a wide range of biochemical and physiological effects in humans. Oxidative damage has been proposed as a possible mechanism involved in Pb toxicity. The current study was carried out to evaluate the antioxidant activities of zinc (Zn) supplement against lead acetate-induced kidney injury in rats. In this study, adults male rats were treated for 15 days with Pb (0.344 g/kg body weight (bw)) associated or not with Zn (10 mg/kg bw). Our study showed that supplementation with Zn prevented renal dysfunction as indicated by plasma biomarkers (urea, uric acid, creatinine, lactate dehydrogenase, and alkaline phosphatase levels) and oxidative stress-related parameters (thiobarbituric acid reactive substances, protein carbonyl, advanced oxidation protein product, superoxide dismutase, catalase, glutathione peroxidase, and vitamins (A, E)) in kidney tissue. The corrective effect of Zn on Pb-induced kidney nephrotoxicity recovered normal kidney histology. Overall, this study indicates that Zn alleviated the toxic effects of this heavy metal on renal tissue, suggesting its role as a potential antioxidant and nephroprotective agent.
Collapse
Affiliation(s)
- Ahlem Soussi
- Laboratory of Animal Ecophysiology, Faculty of Sciences, University of Sfax, Sfax, Tunisia
| | - Manel Gargouri
- Laboratory of Animal Ecophysiology, Faculty of Sciences, University of Sfax, Sfax, Tunisia
| | - Abdelfattah El Feki
- Laboratory of Animal Ecophysiology, Faculty of Sciences, University of Sfax, Sfax, Tunisia
| |
Collapse
|
30
|
Yang M, Liu S, Hu L, Zhan J, Lei P, Wu M. Effects of the antidepressant, mianserin, on early development of fish embryos at low environmentally relevant concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 150:144-151. [PMID: 29272719 DOI: 10.1016/j.ecoenv.2017.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 12/03/2017] [Accepted: 12/09/2017] [Indexed: 06/07/2023]
Abstract
Pharmaceuticals have been considered as emerging organic contaminants in the environment that might pose huge risk to the non-target aquatic organisms. Mianserin, a tetracyclic antidepressant, is present at low detectable concentrations in the aquatic environment; however, limited attention has been devoted to its potential adverse effects on the aquatic animals. In the present study, we first performed an acute toxicity test for mianserin exposure using zebrafish (Danio rerio) embryos during 4-124h post fertilization (hpf). Time-dependent lethal concentrations of mianserin exposure on the zebrafish embryos were firstly determined at mg/L levels. Then, a series of sublethal concentrations of 0.01, 0.1, 1, 10, 100, and 1000μg/L of mianserin were prepared for the short-term exposure of zebrafish embryos for 120h. The results showed that mianserin exposure reduced the body length of zebrafish larvae, in addition to altering multiple physiological and biochemical parameters in the exposed embryos/larvae. A dose-dependent inhibition of the total antioxidant capacity and total cholinesterase activity was revealed in the exposed fish larvae upon increasing the concentrations of mianserin exposure. A U-shaped concentration-dependent response curve was observed for the adrenocorticotropic hormone; however, an inversed U-shaped response curve was obtained for the monoamine oxidase level in response to mianserin exposure. Activities of the total adenosine triphosphatase (T-ATPase), Na+/K+-ATPase, and Ca2+/Mg2+-ATPase were significantly increased in the fish larvae exposed to relatively high doses of mianserin; interestingly however, low dose of mianserin at 10ng/L inhibited their Na+/K+-ATPase and T-ATPase activities. Additionally, the coordinated regulation of cyclic adenosine monophosphate and protein kinase A was observed in the mianserin-exposed fish larvae, implying a reserved signaling pathway involved in the fish response to the antidepressant. Therefore, our study demonstrated that mianserin exposure significantly affected the early development of fish embryos at environmentally relevant concentrations, and suggested that the risk of pharmaceutical contamination of the aquatic environment, even at low doses, should receive more attention.
Collapse
Affiliation(s)
- Ming Yang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Shuai Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Lei Hu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China; School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Jing Zhan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Penghui Lei
- School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Minghong Wu
- Shanghai Applied Radiation Institute, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
31
|
Pellacani C, Costa LG. Role of autophagy in environmental neurotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 235:791-805. [PMID: 29353798 DOI: 10.1016/j.envpol.2017.12.102] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 12/08/2017] [Accepted: 12/25/2017] [Indexed: 06/07/2023]
Abstract
Human exposure to neurotoxic pollutants (e.g. metals, pesticides and other chemicals) is recognized as a key risk factor in the pathogenesis of neurodegenerative disorders. Emerging evidence indicates that an alteration in autophagic pathways may be correlated with the onset of the neurotoxicity resulting from chronic exposure to these pollutants. In fact, autophagy is a natural process that permits to preserving cell homeostasis, through the seizure and degradation of the cytosolic damaged elements. However, when an excessive level of intracellular damage is reached, the autophagic process may also induce cell death. A correct modulation of specific stages of autophagy is important to maintain the correct balance in the organism. In this review, we highlight the critical role that autophagy plays in neurotoxicity induced by the most common classes of environmental contaminants. The understanding of this mechanism may be helpful to discover a potential therapeutic strategy to reduce side effects induced by these compounds.
Collapse
Affiliation(s)
- C Pellacani
- Dept. of Medicine and Surgery, University of Parma, Parma, Italy.
| | - L G Costa
- Dept. of Medicine and Surgery, University of Parma, Parma, Italy; Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
32
|
Aaseth J, Ajsuvakova OP, Skalny AV, Skalnaya MG, Tinkov AA. Chelator combination as therapeutic strategy in mercury and lead poisonings. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2017.12.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
33
|
Burioli EAV, Squadrone S, Stella C, Foglini C, Abete MC, Prearo M. Trace element occurrence in the Pacific oyster Crassostrea gigas from coastal marine ecosystems in Italy. CHEMOSPHERE 2017; 187:248-260. [PMID: 28850909 DOI: 10.1016/j.chemosphere.2017.08.102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/17/2017] [Accepted: 08/18/2017] [Indexed: 06/07/2023]
Abstract
The Pacific oyster is one of the world's most widespread bivalves and a suitable species for biomonitoring trace elements in marine environments thanks to its bioaccumulation ability. As it is also an edible mollusc, concentrations of harmful elements in its tissues must be monitored. For these purposes, 464 wild individuals were collected from 12 sites along the Italian coasts. The concentration of fourteen trace elements (Al, As, Cd, Cr, Cu, Fe, Hg, Mn, Ni, Pb, Se, Sn, Tl, and Zn) in their tissues was quantified. Among the three heavy metals, cadmium, lead, and mercury, none exceeded the maximum limit for in food set by European Union regulations but Cd in one sample from the Varano Lagoon resulted extremely close to this value. Contamination by Hg of the northern Adriatic and Orbetello Lagoons was also observed. Moreover, there was a positive association between the lagoon's environmental conditions and the bioaccumulation of this element in oysters. Despite the ban instituted 15 years ago on the use of Sn in antifouling paints, this element is still present in several marine environments, as demonstrated in the oysters sampled from harbour areas. Samples collected from harbours also showed very high concentrations of Cu and Zn due to the ability of oysters to accumulate these elements, which have replaced Sn in antifouling paints. Analysis of the samples from most sites indicated a low risk of human exposure to harmful elements through oyster consumption; nonetheless, chemical sanitary controls should focus primarily on Cd, Cu, and Zn.
Collapse
Affiliation(s)
- E A V Burioli
- Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle D'Aosta, Turin, Italy.
| | - S Squadrone
- Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle D'Aosta, Turin, Italy
| | - C Stella
- Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle D'Aosta, Turin, Italy
| | - C Foglini
- Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle D'Aosta, Turin, Italy
| | - M C Abete
- Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle D'Aosta, Turin, Italy
| | - M Prearo
- Istituto Zooprofilattico Sperimentale Del Piemonte, Liguria e Valle D'Aosta, Turin, Italy
| |
Collapse
|
34
|
Increased GSNOR Expression during Aging Impairs Cognitive Function and Decreases S-Nitrosation of CaMKIIα. J Neurosci 2017; 37:9741-9758. [PMID: 28883020 DOI: 10.1523/jneurosci.0681-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 11/21/2022] Open
Abstract
As the population ages, an increasing number of people suffer from age-related cognitive impairment. However, the mechanisms underlying this process remain unclear. Here, we found that S-nitrosoglutathione reductase (GSNOR), the key enzyme that metabolizes intracellular nitric oxide (NO) and regulates S-nitrosation, was significantly increased in the hippocampus of both aging humans and mice. Transgenic mice overexpressing GSNOR exclusively in neurons showed cognitive impairment in behavioral tests, including the Morris water maze, fear conditioning, and the Y-maze test. We also found that GSNOR transgenic mice have LTP defects and lower dendrite spine density, whereas GSNOR knock-out mice rescued the age-related cognitive impairment. Analysis of S-nitrosation showed significantly decreased hippocampal CaMKIIα S-nitrosation in naturally aged mice and GSNOR transgenic mice. Consistent with the change in CaMKIIα S-nitrosation, the accumulation of CaMKIIα in the hippocampal synaptosomal fraction, as well as its downstream signaling targets p(S831)-GLUR1, was also significantly decreased. All these effects could be rescued in the GSNOR knock-out mice. We further verified that the S-nitrosation of CaMKIIα was responsible for the CaMKIIα synaptosomal accumulation by mutating CaMKIIα S-nitrosated sites (C280/C289). Upregulation of the NO signaling pathway rescued the cognitive impairment in GSNOR transgenic mice. In summary, our research demonstrates that GSNOR impairs cognitive function in aging and it could serve as a new potential target for the treatment of age-related cognitive impairment. In contrast to the free radical theory of aging, NO signaling deficiency may be the main mediator of age-related cognitive impairment.SIGNIFICANCE STATEMENT This study indicated that S-nitrosoglutathione reductase (GSNOR), a key protein S-nitrosation metabolic enzyme, is a new potential target in age-related cognitive impairment; and in contrast to the free radical theory of aging, NO signaling deficiency may be the main cause of this process. In addition, increased GSNOR expression during aging decreases S-nitrosation of CaMKIIα and reduces CaMKIIα synaptosomal accumulation. To our knowledge, it is for the first time to show the cellular function regulation of CaMKIIα by GSNOR-dependent S-nitrosation as a new post-translational modification after its phosphorylation was explored. These findings elucidate a novel mechanism of age-related cognitive impairment and may provide a new potential target and strategy for slowing down this process.
Collapse
|
35
|
El-Sherbini ES, El-Sayed G, El Shotory R, Gheith N, Abou-Alsoud M, Harakeh SM, Karrouf GI. Ameliorative effects of l-carnitine on rats raised on a diet supplemented with lead acetate. Saudi J Biol Sci 2017; 24:1410-1417. [PMID: 28855839 PMCID: PMC5562480 DOI: 10.1016/j.sjbs.2016.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 05/02/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022] Open
Abstract
Lead intoxication has been a major health hazard in humans. It affects people at all ages. Its toxicity is associated with various organs of the body and affects different metabolic pathways. Based on histological data, l-carnitine reduced the severity of tissue damage produced as a result of exposure of rats to lead acetate. The main objective of this study was to evaluate the underlying mechanism of protection offered by l-carnitine against lead acetate intoxication using male Sprague-Dawley rats. Forty male Sprague-Dawley rats were randomly divided into four groups with ten rats in each. The first group (G1) served as the control group and animals received standard diet only. The second group (G2) received lead acetate in their diet. The third group (G3) was the l-carnitine treated group and received the normal standard diet supplemented with l-carnitine. While the fourth group (G4) had a diet supplemented with both lead acetate and l-carnitine. At the end of each experiment, blood (serum and whole blood) were collected from each animal and analyzed for the following parameters: serum testosterone levels, serum nitric oxide and serum malondialdehyde. This is in addition to looking at the enzymatic activities of two important enzymes (superoxide dismutase and catalase) and on (glutathione reductase) which are indicative of the antioxidant activities in the whole blood. The results indicated that l-carnitine will counteract the undesirable effects of lead intoxication. It exerted its antioxidant potential by reducing the production of ROS and scavenging free radicals by maintaining and protecting the level of the of antioxidant enzymes SOD, CAT and glutathione peroxidase. Conclusion:l-Carnitine may play an important role in reversing the undesirable effects of lead intoxication. Future studies should be conducted to see whether such an effect is applicable in humans exposed to lead poising.
Collapse
Affiliation(s)
- El-Said El-Sherbini
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Gehad El-Sayed
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Rehab El Shotory
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Nervana Gheith
- Public Administration Departments, Faculty of Economic and Administration, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed Abou-Alsoud
- Faculty of Meteorology, Environment and Arid Land Agriculture, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Steve Mustapha Harakeh
- Special Infectious Agents Unit – King Fahd Medical Research Center, King Abdulaziz University, P.O. Box: 80216, Jeddah 21589; Saudi Arabia
| | - Gamal I. Karrouf
- Medical Physics Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Surgery, Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Dakahlia, Egypt
| |
Collapse
|
36
|
Wang Y, Ezemaduka AN, Li Z, Chen Z, Song C. Joint Toxicity of Arsenic, Copper and Glyphosate on Behavior, Reproduction and Heat Shock Protein Response in Caenorhabditis elegans. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2017; 98:465-471. [PMID: 28224177 DOI: 10.1007/s00128-017-2042-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 02/01/2017] [Indexed: 06/06/2023]
Abstract
The soil nematode Caenorhabditis elegans was used in 24-h acute exposures to arsenic (As), copper (Cu) and glyphosate (GPS) and to mixtures of As/Cu and As/GPS to investigate the effects of mixture exposures in the worms. A synergistic type of interaction was observed for acute toxicity with the As/Cu and As/GPS mixtures. Sublethal 24-h exposures of 1/1000, 1/100 and 1/10 of the LC50 concentrations for As, Cu and GPS individually and for As/Cu and As/GPS mixtures were conducted to observe responses in locomotory behavior (head thrashing), reproduction, and heat shock protein expression. Head thrash frequency and reproduction exhibited concentration dependent decreases in both individual and combined exposures to the tested chemical stressors, and showed synergistic interactions even at micromolar concentrations. Furthermore, the HSP70 protein level was significantly increased following exposure to individual and combined chemical stressors in wild-type C. elegans. Our findings establish for the first time the effects of exposure to As/GPS and As/Cu mixtures in C. elegans.
Collapse
Affiliation(s)
- Yunbiao Wang
- Key Laboratory of Wetland Ecology and Environment, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, 130102, China.
| | - Anastasia N Ezemaduka
- Key Laboratory of Wetland Ecology and Environment, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, 130102, China
| | - Zhuheng Li
- Jilin Provincial Institute of Education, Changchun, 130022, China
| | - Zhanyan Chen
- Key Laboratory of Wetland Ecology and Environment, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, 130102, China
| | - Chuantao Song
- School of Environment, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
37
|
Ferreira MCDF, Zucki F, Duarte JL, Iano FG, Ximenes VF, Buzalaf MAR, Oliveira RCD. Influence of iron on modulation of the antioxidant system in rat brains exposed to lead. ENVIRONMENTAL TOXICOLOGY 2017; 32:813-822. [PMID: 27170105 DOI: 10.1002/tox.22281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 06/05/2023]
Abstract
The objective of this study was to evaluate markers of oxidative stress in the brains of rats exposed to lead acetate (Pb(C2 H3 O2 )2 ), either associated or not associated with ferrous sulfate (FeSO4 ). A total of 36 weaning rats (Rattus norvegicus) were divided into 6 groups of six animals and exposed to lead acetate for six weeks. In the control group (control), the animals received deionized water. The Pb260 and Pb260 + Fe received 260 µM lead acetate, and the Pb1050 and Pb1050 + Fe received 1050 µM lead acetate. The Pb260 + Fe and Pb1050 + Fe were supplemented with 20 mg of ferrous sulfate/Kg body weight every 2 days. Group Fe received deionized water and ferrous sulfate. The rat brains were collected to analyze the enzymatic activity of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and the concentration of reduced glutathione (GSH), lipid peroxidation (TBARS), and total antioxidant substance (TAS) (DPPH• technique). The activity of SOD and GPx in the experimental groups decreased compared to the control, together with the concentration of GSH (p < 0.05). For CAT analysis, SOD tended to increase in concentration in the experimental groups without a concomitant exposure to FeSO4 , whereas GPx showed a slight tendency to increase in activity compared to the control. For TAS-DPPH• , there was a decrease in the experimental groups (p < 0.05). According to the results, SOD, GPx, and GSH were affected by lead acetate and exposure to ferrous sulfate changed this dynamic. However, further studies are needed to verify whether ferrous sulfate acts as a protectant against the toxic effects of lead. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 813-822, 2017.
Collapse
Affiliation(s)
- Maria Cecília de Freitas Ferreira
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Fernanda Zucki
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Josilene Luciene Duarte
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Flávia Godoy Iano
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Valdecir Farias Ximenes
- Department of Chemistry, Faculty of Sciences, São Paulo State University, Av. Engenheiro Luiz Edmundo Carrijo Coube, 1401 Bauru-SP 17033-360, Brazil
| | - Marília Afonso Rabelo Buzalaf
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Rodrigo Cardoso de Oliveira
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| |
Collapse
|
38
|
Liu X, Ye J, Wang L, Li Z, Zhang Y, Sun J, Du C, Wang C, Xu S. Protective Effects of PGC-1α Against Lead-Induced Oxidative Stress and Energy Metabolism Dysfunction in Testis Sertoli Cells. Biol Trace Elem Res 2017; 175:440-448. [PMID: 27392955 DOI: 10.1007/s12011-016-0799-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/04/2016] [Indexed: 12/06/2022]
Abstract
The reproductive system is sensitive to lead (Pb) toxicity, which has long been an area of research interest, but the underlying mechanisms remain to be illustrated. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) is pivotal in mitochondrial function. In this study, mouse testis Sertoli cells (TM4 cells), PGC-1α lower-expression (PGC-1α(-)) TM4 cells and PGC-1α overexpression (PGC-1α(+)) TM4 cells were used to explore the protective roles of PGC-1α against lead toxicity on the mouse reproductive system. Lead acetate (PbAc) exposure decreased the expression level of PGC-1α, increased the intracellular level of reactive oxygen species (ROS), and reduced the level of ATP in the three TM4 cell lines. The effects of PbAc on intracellular ATP level and on ROS content were significantly weakened in PGC-1α(+)TM4 cells versus TM4 cells and were significantly amplified in PGC-1α(-)TM4 cells versus TM4 cells. These results suggest that PGC-1α is a protective factor against PbAc-induced oxidative stress and energy metabolism dysfunction in the mouse reproductive system, thereby holding the potential of being developed as a preventive or therapeutic strategy against disorders induced by lead exposure.
Collapse
Affiliation(s)
- Xi Liu
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
| | - Jingping Ye
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
- Renmin hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Lu Wang
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
| | - Zhen Li
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
| | - Yucheng Zhang
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
| | - Jiantao Sun
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
| | - Chuang Du
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China
| | - Chunhong Wang
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan, 430071, People's Republic of China.
| | - Siyuan Xu
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Academy for Preventive Medicine, Wuhan, 430079, People's Republic of China.
| |
Collapse
|
39
|
Rameshrad M, Razavi BM, Hosseinzadeh H. Protective effects of green tea and its main constituents against natural and chemical toxins: A comprehensive review. Food Chem Toxicol 2016; 100:115-137. [PMID: 27915048 DOI: 10.1016/j.fct.2016.11.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 01/26/2023]
Abstract
Toxins are natural or chemical poisonous substances with severe side effects on health. Humans are generally exposed by widespread toxic contaminations via air, soil, water, food, fruits and vegetables. Determining a critical antidote agent with extensive effects on different toxins is an ultimate goal for all toxicologists. Traditional medicine is currently perceived as a safe and natural approach against toxins. In this regard, we focused on the protective effects of green tea (Camellia sinensis) and its main components such as catechin, epicatechin, epicatechin gallate, gallocatechin, epigallocatechin and epigallocatechin gallate as a principal source of antioxidants against both natural and chemical toxins. This literate review demonstrates that protective effects of green tea and its constituents were mainly attributed to their anti-oxidative, radical scavenging, chelating, anti-apoptotic properties and modulating inflammatory responses. Although, some studies reveal they have protective effects by increasing toxin metabolism and neutralizing PLA2, proteases, hyaluronidase and l-amino acid oxidase enzymes.
Collapse
Affiliation(s)
- Maryam Rameshrad
- Pharmaceutical Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Bibi Marjan Razavi
- Targeted Drug Delivery Research Center, Department of Pharmacodynamy and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
40
|
Meng H, Wang L, He J, Wang Z. The Protective Effect of Gangliosides on Lead (Pb)-Induced Neurotoxicity Is Mediated by Autophagic Pathways. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:365. [PMID: 27023584 PMCID: PMC4847027 DOI: 10.3390/ijerph13040365] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/08/2016] [Accepted: 03/19/2016] [Indexed: 12/27/2022]
Abstract
Lead (Pb) is a ubiquitous environmental and industrial pollutant and can affect intelligence development and the learning ability and memory of children. Therefore, necessary measures should be taken to protect the central nervous system (CNS) from Pb toxicity. Gangliosides are sialic acid-containing glycosphingolipids that are constituents of mammalian cell membranes and are more abundantly expressed in the CNS. Studies have shown that gangliosides constitute a useful tool in the attempt to promote functional recovery of CNS and can reverse Pb-induced impairments of synaptic plasticity in rats. However, the detailed mechanisms have yet to be fully understood. In our present study, we tried to investigate the role of gangliosides in Pb-induced injury in hippocampus neurons and to further confirm the detailed mechanism. Our results show that Pb-induced injuries in the spatial reference memory were associated with a reduction of cell viability and cell apoptosis, and treatment with gangliosides markedly ameliorated the Pb-induced injury by inhibition of apoptosis action. Gangliosides further attenuated Pb-induced the abnormal autophagic process by regulation of mTOR pathways. In summary, our study establishes the efficacy of gangliosides as neuroprotective agents and provides a strong rationale for further studies on the underlying mechanisms of their neuroprotective functions.
Collapse
Affiliation(s)
- Hongtao Meng
- Department of Neurology, Shanxi Hospital of the Armed Police Force, Xi'an 710054, China.
| | - Lan Wang
- Department of Neurology, Shanxi Hospital of the Armed Police Force, Xi'an 710054, China.
| | - Junhong He
- Department of Neurology, Shanxi Hospital of the Armed Police Force, Xi'an 710054, China.
| | - Zhufeng Wang
- Department of Neurology, Shanxi Hospital of the Armed Police Force, Xi'an 710054, China.
| |
Collapse
|
41
|
Abstract
Metals play important roles in the human body, maintaining cell structure and regulating gene expression, neurotransmission, and antioxidant response, to name a few. However, excessive metal accumulation in the nervous system may be toxic, inducing oxidative stress, disrupting mitochondrial function, and impairing the activity of numerous enzymes. Damage caused by metal accumulation may result in permanent injuries, including severe neurological disorders. Epidemiological and clinical studies have shown a strong correlation between aberrant metal exposure and a number of neurological diseases, including Alzheimer’s disease, amyotrophic lateral sclerosis, autism spectrum disorders, Guillain–Barré disease, Gulf War syndrome, Huntington’s disease, multiple sclerosis, Parkinson’s disease, and Wilson’s disease. Here, we briefly survey the literature relating to the role of metals in neurodegeneration.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Mahfuzur Rahman Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
42
|
Su P, Zhang J, Wang S, Aschner M, Cao Z, Zhao F, Wang D, Chen J, Luo W. Genistein alleviates lead-induced neurotoxicity in vitro and in vivo: Involvement of multiple signaling pathways. Neurotoxicology 2016; 53:153-164. [PMID: 26797587 DOI: 10.1016/j.neuro.2015.12.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/18/2015] [Accepted: 12/29/2015] [Indexed: 12/14/2022]
Abstract
Lead (Pb) is a ubiquitous environmental and industrial pollutant. It induces neurotoxicity and cell death by disrupting the pro- and anti-oxidative balance; however, the mechanisms of its toxicity have yet to be fully understood. The soy-derived isoflavonoid, genistein (GEN), was reported to possess neuroprotective and antioxidative properties. The present study investigated the molecular mechanisms of Pb-induced neurotoxicity in vivo and in vitro, addressing the efficacy of GEN in protecting against Pb-induced toxicity. Pb exposure was associated with reduction of cell viability and cell apoptosis, concomitant with reactive oxygen species (ROS) generation in vitro, and pre-treatment with GEN markedly ameliorated the Pb-induced oxidative injury by increasing the expression of key antioxidant enzymes and the antioxidant transcription factor, nuclear factor erythroid 2 p45-related factor 2 (Nrf2). Next, PKC-α activation was found after Pb exposure in vitro and pretreatment with GEN attenuated Pb-induced ROS generation by PKC-α inhibition. MAPK-NF-κB activation triggered by Pb was also inhibited by GEN. In summary, our study establishes that GEN alleviates Pb-induced impairment in spatial memory, and reduces cell apoptosis caused by Pb exposure and GEN protects neurons from Pb-induced neurotoxicity by downstream activation of antioxidant and anti-apoptotic pathways via regulation of Nrf2 and MAPK-NF-κB signaling.
Collapse
Affiliation(s)
- Peng Su
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianbin Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Siwang Wang
- Institute of Materia Medica, Fourth Military Medical University, Xi'an 710032, China
| | | | - Zipeng Cao
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Fang Zhao
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Diya Wang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiangyuan Chen
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Wenjing Luo
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
43
|
Zhang Z, Miah M, Culbreth M, Aschner M. Autophagy in Neurodegenerative Diseases and Metal Neurotoxicity. Neurochem Res 2016; 41:409-22. [PMID: 26869037 DOI: 10.1007/s11064-016-1844-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/20/2016] [Accepted: 01/22/2016] [Indexed: 02/07/2023]
Abstract
Autophagy generally refers to cell catabolic and recycling process in which cytoplasmic components are delivered to lysosomes for degradation. During the last two decades, autophagy research has experienced a recent boom because of a newfound connection between this process and many human diseases. Autophagy plays a significant role in maintaining cellular homeostasis and protects cells from varying insults, including misfolded and aggregated proteins and damaged organelles, which is particularly crucial in neuronal survival. Mounting evidence has implicated autophagic dysfunction in the pathogenesis of several major neurodegenerative disorders, such as Parkinson's disease, Alzheimer's disease and Huntington's disease, where deficient elimination of abnormal and toxic protein aggregates promotes cellular stress, failure and death. In addition, autophagy has also been found to affect neurotoxicity induced by exposure to essential metals, such as manganese, copper, and iron, and other heavy metals, such as cadmium, lead, and methylmercury. This review examines current literature on the role of autophagy in the mechanisms of disease pathogenesis amongst common neurodegenerative disorders and of metal-induced neurotoxicity.
Collapse
Affiliation(s)
- Ziyan Zhang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 209, Bronx, NY, 10461, USA
| | - Mahfuzur Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 209, Bronx, NY, 10461, USA
| | - Megan Culbreth
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 209, Bronx, NY, 10461, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 209, Bronx, NY, 10461, USA.
| |
Collapse
|
44
|
Yu H, Liao Y, Li T, Cui Y, Wang G, Zhao F, Jin Y. Alterations of Synaptic Proteins in the Hippocampus of Mouse Offspring Induced by Developmental Lead Exposure. Mol Neurobiol 2015; 53:6786-6798. [DOI: 10.1007/s12035-015-9597-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 12/01/2015] [Indexed: 12/22/2022]
|
45
|
Liu J, Liu X, Pak V, Wang Y, Yan C, Pinto-Martin J, Dinges D. Early Blood Lead Levels and Sleep Disturbance in Preadolescence. Sleep 2015; 38:1869-74. [PMID: 26194570 PMCID: PMC4667382 DOI: 10.5665/sleep.5230] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/11/2015] [Indexed: 12/30/2022] Open
Abstract
STUDY OBJECTIVES Little is known about the effect of lead exposure on children's sleep. This study examined the association between blood lead levels (BLL) and sleep problems in a longitudinal study of children. SETTING Four community-based elementary schools in Jintan City, China. PARTICIPANTS 1,419 Chinese children. MEASUREMENT AND RESULTS BLL were measured when children were aged 3-5 y, and sleep was assessed at ages 9-13 y. Sleep was assessed by both parents' report, using the Children's Sleep Habits Questionnaire (CSHQ), and children's report, using an adolescent sleep questionnaire. A total of 665 children with complete data on BLL and sleep at both ages were included in the current study. Mean age of the sample at BLL assessment was 4.74 y (standard deviation [SD] = 0.89) and at sleep assessment was 11.05 y (SD = 0.88). Mean BLL was 6.26 μg/dL (SD = 2.54). There were significant positive correlations between BLL and 3 CSHQ subscales: Sleep onset delay (r = 0.113, P < 0.01), sleep duration (r = 0.139, P < 0.001), and night waking (r = 0.089, P < 0.05). Excessive daytime sleepiness (EDS) (26.1% versus 9.0%, P < 0.001) and use of sleeping pills (6.5% versus 1.8%, P = 0.03) were more prevalent in children BLL ≥ 10.0 μg/dL than in those children BLL < 10.0 μg/dL. After adjusting for demographics, BLL ≥ 10.0 μg/dL was significantly associated with increased risk for insomnia symptoms (odds ratio [OR] = 2.01, 95% confidence interval [CI] = 1.03-3.95) and EDS (OR = 2.90, 95% CI = 1.27-6.61). CONCLUSION The findings indicate that elevated blood lead levels in early childhood are associated with increased risk for sleep problems and excessive daytime sleepiness in later childhood.
Collapse
Affiliation(s)
- Jianghong Liu
- School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Xianchen Liu
- Shandong University School of Public Health, Jinan, China
| | - Victoria Pak
- School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Yingjie Wang
- School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Chonghuai Yan
- Xinhua Hospital, MOE-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Jiaotong University School of Medicine, China
| | | | - David Dinges
- Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
46
|
Barcelos GRM, De Marco KC, de Rezende VB, Braga GÚL, Antunes LMG, Tanus-Santos JE, Barbosa F. Genetic Effects of eNOS Polymorphisms on Biomarkers Related to Cardiovascular Status in a Population Coexposed to Methylmercury and Lead. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2015; 69:173-180. [PMID: 25690149 DOI: 10.1007/s00244-015-0137-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 02/01/2015] [Indexed: 06/04/2023]
Abstract
The aim of the present study was to evaluate possible effects of endothelial nitric oxide synthase (eNOS) polymorphisms on systolic (SBP) and diastolic blood pressure (DBP) and on nitrite levels in plasma (NitP) in a population coexposed to methylhemoglobin (MeHg) and lead (Pb) in the Amazonian region, Brazil. Plasmatic levels of hemoglobin Hg (HgP) and Pb (PbP) were determined by inductively coupled plasma-mass spectrometry, whereas NitP were quantified by chemiluminescence. Genotyping was performed by conventional and restriction fragment length polymorphism-polymerase chain reaction assay. The population age ranged from 18 to 87 years (mean 40 ± 16), and the distribution between the sexes was homogenous (63 men and 50 women). Mean HgP and PbP were 7.1 ± 6.1 and 1.1 ± 1.1 µg L(-1), respectively. PbP was correlated to SBP and DBP, whereas no effects were observed for HgP on blood pressure. Subjects carrying the 4b allele in intron 4 presented greater SBP and DBP compared with those who had the 4a4a genotype. In addition, interactions between alcohol consumption and the -786 T/C polymorphism were observed on NitP, i.e., individuals carrying the polymorphic allele and drinkers had lower NitP. Taken together, our data give new insights concerning the genetic effects of eNOS polymorphisms on biomarkers related to cardiovascular status in populations coexposed to Hg and Pb.
Collapse
Affiliation(s)
- Gustavo Rafael Mazzaron Barcelos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, Ribeirão Preto, São Paulo, CEP 14040-903, Brazil,
| | | | | | | | | | | | | |
Collapse
|
47
|
Venkareddy LK, Muralidhara. Potential of casein as a nutrient intervention to alleviate lead (Pb) acetate-mediated oxidative stress and neurotoxicity: First evidence in Drosophila melanogaster. Neurotoxicology 2015; 48:142-51. [DOI: 10.1016/j.neuro.2015.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/27/2015] [Accepted: 03/06/2015] [Indexed: 12/25/2022]
|
48
|
Smirnova L, Hogberg HT, Leist M, Hartung T. Developmental neurotoxicity - challenges in the 21st century and in vitro opportunities. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2015; 31:129-56. [PMID: 24687333 DOI: 10.14573/altex.1403271] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 11/23/2022]
Abstract
In recent years neurodevelopmental problems in children have increased at a rate that suggests lifestyle factors and chemical exposures as likely contributors. When environmental chemicals contribute to neurodevelopmental disorders developmental neurotoxicity (DNT) becomes an enormous concern. But how can it be tackled? Current animal test- based guidelines are prohibitively expensive, at $ 1.4 million per substance, while their predictivity for human health effects may be limited, and mechanistic data that would help species extrapolation are not available. A broader screening for substances of concern requires a reliable testing strategy, applicable to larger numbers of substances, and sufficiently predictive to warrant further testing. This review discusses the evidence for possible contributions of environmental chemicals to DNT, limitations of the current test paradigm, emerging concepts and technologies pertinent to in vitro DNT testing and assay evaluation, as well as the prospect of a paradigm shift based on 21st century technologies.
Collapse
Affiliation(s)
- Lena Smirnova
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, USA
| | | | | | | |
Collapse
|
49
|
Wang J, Song L, Li K, Yan R, Hu X, Zhang W, Yin Y, Zhao S. Protective effects of lithium against lead-induced toxicities in multiple systems of adult mouse. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00071h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Occupational and environmental exposures to lead (Pb), one of the toxic metal pollutants, is of global concern.
Collapse
Affiliation(s)
- Jiutao Wang
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Lingzhen Song
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Kaikai Li
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Runchuan Yan
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Xinde Hu
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Wei Zhang
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Yupeng Yin
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| | - Shanting Zhao
- College of Veterinary Medicine
- Northwest A&F University
- Yangling
- People's Republic of China
| |
Collapse
|
50
|
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
|